1
|
Nowak AL, Saadat N, Sun J, Forsman AM, Liang X, Joyce C, Woo J, Engeland CG, Misra DP, Giurgescu C, Zhang W, Anderson CM. Preterm Birth in African American Women: A Multi-Omic Pilot Study in Early Pregnancy. Biol Res Nurs 2024:10998004241275049. [PMID: 39440846 DOI: 10.1177/10998004241275049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Preterm birth (PTB; <37 weeks completed gestation) is a devastating problem affecting over 13 million live births worldwide. In the U.S., African Americans experience significantly higher rates of PTB compared to non-Hispanic Whites. PTB disparities have been linked to social determinants of health (e.g., socioeconomic status, discrimination). However, the biological underpinnings related to these associations are unclear. DNA methylation (DNAm) is subject to environmental influences, and DNAm modifications are known to affect gene expression. Using a multi-omic approach, we examined differences in combined DNA methylation (DNAm) and messenger RNA (mRNA) transcriptomic data from 20 pregnant African American women (12 PTB; 8 term birth) early in pregnancy (8-18 weeks gestation). We found that the HLA-DQB2 gene was both differentially methylated (cg12296550; p = .02) and differentially expressed (p = .014; log2FC = 2.5) between women with PTB and term birth. Gene expression analysis showed HLA-DQB2 and HLA-DRB4 (p = .028; log2FC = -3.6) were the two most highly expressed genes. HLA-DQB2 expressed higher in PTB and HLA-DRB4 expressed higher in term birth. However, no genes remained significant (p < .05) after Bonferroni correction. HLA-DRB4 and AKR1C1 were identified as a potential biomarkers in dimensionality reduction models and are also important to immune function and allogenic breakdown. Altered gene expression may lead to inflammatory imbalances or allogenic intolerance resulting in PTB. This study provides proof-of-concept evidence for the feasibility and importance of future multi-omics studies with larger populations to further explore the genes and pathways identified here.
Collapse
Affiliation(s)
- Alexandra L Nowak
- Marcella Niehoff School of Nursing, Loyola University at Chicago, Maywood, IL, USA
| | - Nadia Saadat
- Department of Paediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Jiao Sun
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Anna M Forsman
- Department of Biology, Colby College, Waterville, ME, USA
| | - Xiaoyu Liang
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Cara Joyce
- Biostatistics Core, Department of Medicine, Center for Translational Research and Education, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Jennifer Woo
- University of Texas at Arlington, Arlington, TX, USA
| | - Christopher G Engeland
- Biobehavioral Health, College of Health and Human Development, Ross and Carol Ness College of Nursing, The Pennsylvania State University, University Park, PA, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, MSU College of Human Medicine, East Lansing, MI, USA
| | - Carmen Giurgescu
- Chatlos Foundation Endowed Chair in Nursing, University of Central Florida College of Nursing, Orlando, FL, USA
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Cindy M Anderson
- Maternal Infant Health, Martha S. Pitzer Center for Women, Children and Youth, The Ohio State University College of Nursing, Columbus, OH, USA
| |
Collapse
|
2
|
Dye TDV, Quiñones Tavárez Z, Rivera I, Cardona Cordero N. Social determinants of participation in genetic research among Puerto Ricans and in the Puerto Rican diaspora. Soc Sci Med 2024; 362:117437. [PMID: 39461167 DOI: 10.1016/j.socscimed.2024.117437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Puerto Ricans are underrepresented in genetic research. This underrepresentation denies Puerto Ricans the benefit from therapeutic developments that could mitigate health disparities arising from conditions for which genetically-derived treatments exist. The Puerto Rican diaspora, especially post-2017 due to economic and environmental crises, has expanded within the USA. Prior research suggests that Latin American diaspora communities are less likely to participate in genetic research. We hypothesized, specifically, that the Puerto Rican diaspora in the USA would be less likely to participate in genetic research than would Puerto Ricans in their homeland's archipelago, and that accounting for social and cultural determinants related to the diaspora experience would mitigate this disparity. We implemented an analytical cross-sectional study of archipelago-residing Puerto Ricans and of the USA-residing diaspora to evaluate this hypothesis. With 1582 Puerto Ricans (723 in Puerto Rico, 859 in the USA), we found that while most participants would participate in genetic research, participation rates varied significantly by diaspora status. Puerto Ricans born and living in the USA were initially more likely to decline participation compared to those in Puerto Rico (OR = 1.54, p < 0.01). However, once adjusted for social and cultural variables, this difference was eliminated (aOR = 1.08, p = n.s.). The factors influencing non-participation include oppression, discrimination, distrust, and social determinants, aligning with the theory of minoritization. An important community in the USA and in the world, Puerto Ricans have the right to participate in well-conducted research and to benefit from its findings, particularly around topics that could help address existing disparities in health outcomes.
Collapse
Affiliation(s)
- Timothy De Ver Dye
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | - Zahira Quiñones Tavárez
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | - Ivelisse Rivera
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | | |
Collapse
|
3
|
Maggs X. A synthetic review: natural history of amniote reproductive modes in light of comparative evolutionary genomics. Biol Rev Camb Philos Soc 2024. [PMID: 39300750 DOI: 10.1111/brv.13145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
There is a current lack of consensus on whether the ancestral parity mode was oviparity (egg-laying) or viviparity (live-birth) in amniotes and particularly in squamates (snakes, lizards, and amphisbaenids). How transitions between parity modes occur at the genomic level has primary importance for how science conceptualises the origin of amniotes, and highly variable parity modes in Squamata. Synthesising literature from medicine, poultry science, reproductive biology, and evolutionary biology, I review the genomics and physiology of five broad processes (here termed the 'Main Five') expected to change during transitions between parity modes: eggshell formation, embryonic retention, placentation, calcium transport, and maternal-fetal immune dynamics. Throughout, I offer alternative perspectives and testable hypotheses regarding proximate causes of parity mode evolution in amniotes and squamates. If viviparity did evolve early in the history of lepidosaurs, I offer the nucleation site hypothesis as a proximate explanation. The framework of this hypothesis can be extended to amniotes to infer their ancestral state. I also provide a mechanism and hypothesis on how squamates may transition from viviparity to oviparity and make predictions about the directionality of transitions in three species. After considering evidence for differing perspectives on amniote origins, I offer a framework that unifies (i) the extended embryonic retention model and (ii) the traditional model which describes the amniote egg as an adaptation to the terrestrial environment. Additionally, this review contextualises the origin of amniotes and parity mode evolution within Medawar's paradigm. Medawar posited that pregnancy could be supported by immunosuppression, inertness, evasion, or immunological barriers. I demonstrate that this does not support gestation or gravidity across most amniotes but may be an adequate paradigm to explain how the first amniote tolerated internal fertilization and delayed egg deposition. In this context, the eggshell can be thought of as an immunological barrier. If serving as a barrier underpins the origin of the amniote eggshell, there should be evidence that oviparous gravidity can be met with a lack of immunological responses in utero. Rare examples of two species that differentially express very few genes during gravidity, suggestive of an absent immunological reaction to oviparous gravidity, are two skinks Lampropholis guichenoti and Lerista bougainvillii. These species may serve as good models for the original amniote egg. Overall, this review grounds itself in the historical literature while offering a modern perspective on the origin of amniotes. I encourage the scientific community to utilise this review as a resource in evolutionary and comparative genomics studies, embrace the complexity of the system, and thoughtfully consider the frameworks proposed.
Collapse
Affiliation(s)
- X Maggs
- Richard Gilder Graduate School at The American Museum of Natural History, 200 Central Park West, New York, NY, 10024, USA
- Christopher S. Bond Life Science Center at the University of Missouri, 1201 Rollins St, Columbia, MO, 65201, USA
- School of Life and Environmental Sciences at the University of Sydney, Heydon-Laurence Building A08, Sydney, NSW, 2006, Australia
| |
Collapse
|
4
|
Eaves LA, Harrington CE, Fry RC. Epigenetic Responses to Nonchemical Stressors: Potential Molecular Links to Perinatal Health Outcomes. Curr Environ Health Rep 2024; 11:145-157. [PMID: 38580766 DOI: 10.1007/s40572-024-00435-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2024] [Indexed: 04/07/2024]
Abstract
PURPOSE OF REVIEW We summarize the recent literature investigating exposure to four nonchemical stressors (financial stress, racism, psychosocial stress, and trauma) and DNA methylation, miRNA expression, and mRNA expression. We also highlight the relationships between these epigenetic changes and six critical perinatal outcomes (preterm birth, low birth weight, preeclampsia, gestational diabetes, childhood allergic disease, and childhood neurocognition). RECENT FINDINGS Multiple studies have found financial stress, psychosocial stress, and trauma to be associated with DNA methylation and/or miRNA and mRNA expression. Fewer studies have investigated the effects of racism. The majority of studies assessed epigenetic or genomic changes in maternal blood, cord blood, or placenta. Several studies included multi-OMIC assessments in which DNA methylation and/or miRNA expression were associated with gene expression. There is strong evidence for the role of epigenetics in driving the health outcomes considered. A total of 22 biomarkers, including numerous HPA axis genes, were identified to be epigenetically altered by both stressors and outcomes. Epigenetic changes related to inflammation, the immune and endocrine systems, and cell growth and survival were highlighted across numerous studies. Maternal exposure to nonchemical stressors is associated with epigenetic and/or genomic changes in a tissue-specific manner among inflammatory, immune, endocrine, and cell growth-related pathways, which may act as mediating pathways to perinatal health outcomes. Future research can test the mediating role of the specific biomarkers identified as linked with both stressors and outcomes. Understanding underlying epigenetic mechanisms altered by nonchemical stressors can provide a better understanding of how chemical and nonchemical exposures interact.
Collapse
Affiliation(s)
- Lauren A Eaves
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Cailee E Harrington
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
5
|
Oskotsky TT, Yin O, Khan U, Arnaout L, Sirota M. Data-driven insights can transform women's reproductive health. NPJ WOMEN'S HEALTH 2024; 2:14. [PMID: 38770215 PMCID: PMC11104016 DOI: 10.1038/s44294-024-00019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/20/2024] [Indexed: 05/22/2024]
Abstract
This perspective explores the transformative potential of data-driven insights to understand and address women's reproductive health conditions. Historically, clinical studies often excluded women, hindering comprehensive research into conditions such as adverse pregnancy outcomes and endometriosis. Recent advances in technology (e.g., next-generation sequencing techniques, electronic medical records (EMRs), computational power) provide unprecedented opportunities for research in women's reproductive health. Studies of molecular data, including large-scale meta-analyses, provide valuable insights into conditions like preterm birth and preeclampsia. Moreover, EMRs and other clinical data sources enable researchers to study populations of individuals, uncovering trends and associations in women's reproductive health conditions. Despite these advancements, challenges such as data completeness, accuracy, and representation persist. We emphasize the importance of holistic approaches, greater inclusion, and refining and expanding on how we leverage data and computational integrative approaches for discoveries so that we can benefit not only women's reproductive health but overall human health.
Collapse
Affiliation(s)
- Tomiko T. Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA USA
| | - Ophelia Yin
- Maternal–Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA USA
| | - Umair Khan
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA USA
| | - Leen Arnaout
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA USA
| |
Collapse
|
6
|
Wei J, Zhang L, Xu H, Luo Q. Preterm birth, a consequence of immune deviation mediated hyperinflammation. Heliyon 2024; 10:e28483. [PMID: 38689990 PMCID: PMC11059518 DOI: 10.1016/j.heliyon.2024.e28483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Preterm birth represents a multifaceted syndrome with intricacies still present in our comprehension of its etiology. In the context of a semi-allograft, the prosperity from implantation to pregnancy to delivery hinges on the establishment of a favorable maternal-fetal immune microenvironment and a successful trilogy of immune activation, immune tolerance and then immune activation transitions. The occurrence of spontaneous preterm birth could be related to abnormalities within the immune trilogy, stemming from deviation in maternal and fetal immunity. These immune deviations, characterized by insufficient immune tolerance and early immune activation, ultimately culminated in an unsustainable pregnancy. In this review, we accentuated the role of both innate and adaptive immune reason in promoting spontaneous preterm birth, reviewed the risk of preterm birth from vaginal microbiome mediated by immune changes and the potential of vaginal microbiomes and metabolites as a new predictive marker, and discuss the changes in the role of progesterone and its interaction with immune cells in a preterm birth population. Our objective was to contribute to the growing body of knowledge in the field, shedding light on the immunologic reason of spontaneous preterm birth and effective biomarkers for early prediction, providing a roadmap for forthcoming investigations.
Collapse
Affiliation(s)
- Juan Wei
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - LiYuan Zhang
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - Heng Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| |
Collapse
|
7
|
Cifkova E, Karahoda R, Stranik J, Abad C, Kacerovsky M, Lisa M, Staud F. Metabolomic analysis of the human placenta reveals perturbations in amino acids, purine metabolites, and small organic acids in spontaneous preterm birth. EXCLI JOURNAL 2024; 23:264-282. [PMID: 38487084 PMCID: PMC10938235 DOI: 10.17179/excli2023-6785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 03/17/2024]
Abstract
Spontaneous preterm delivery presents one of the most complex challenges in obstetrics and is a leading cause of perinatal morbidity and mortality. Although it is a common endpoint for multiple pathological processes, the mechanisms governing the etiological complexity of spontaneous preterm birth and the placental responses are poorly understood. This study examined placental tissues collected between May 2019 and May 2022 from a well-defined cohort of women who experienced spontaneous preterm birth (n = 72) and healthy full-term deliveries (n = 30). Placental metabolomic profiling of polar metabolites was performed using Ultra-High Performance Liquid Chromatography/Mass Spectrometry (UHPLC/MS) analysis. The resulting data were analyzed using multi- and univariate statistical methods followed by unsupervised clustering. A comprehensive metabolomic evaluation of the placenta revealed that spontaneous preterm birth was associated with significant changes in the levels of 34 polar metabolites involved in intracellular energy metabolism and biochemical activity, including amino acids, purine metabolites, and small organic acids. We found that neither the preterm delivery phenotype nor the inflammatory response explain the reported differential placental metabolome. However, unsupervised clustering revealed two molecular subtypes of placentas from spontaneous preterm pregnancies exhibiting differential enrichment of clinical parameters. We also identified differences between early and late preterm samples, suggesting distinct placental functions in early spontaneous preterm delivery. Altogether, we present evidence that spontaneous preterm birth is associated with significant changes in the level of placental polar metabolites. Dysregulation of the placental metabolome may underpin important (patho)physiological mechanisms involved in preterm birth etiology and long-term neonatal outcomes.
Collapse
Affiliation(s)
- Eva Cifkova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Miroslav Lisa
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| |
Collapse
|
8
|
Reforma L, Greenberg S, Ledyard R, Burris H. Equitable Representation of Race, Ethnicity, and Ancestry Among Genomic Studies of Preterm Birth: A Systematic Review. Cureus 2024; 16:e53757. [PMID: 38465134 PMCID: PMC10921821 DOI: 10.7759/cureus.53757] [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] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
We conducted a systematic review of representation of race, ethnicity, and ancestry among genomic studies of preterm birth. Our data sources included CINHAL, EMBASE, MEDLINE (PubMed), and Scopus. Studies were included if they were human, genomic studies of preterm birth that analyzed greater than 1,000 genes and included race, ethnicity, and/or ancestry information. Two authors independently reviewed all abstracts and full-text manuscripts. Twelve studies were included. Ancestry was reported for 139,189 (93.6%) participants. Race was reported for 4,841 (3.3%) participants and ethnicity was reported for 7,154 (5.0%) participants. Of the 148,644 births represented in this systematic review, over 90% were reported to be of European ancestry, and race and ethnicity were not further described. When examining the smaller subset of individuals described by race alone, 2,444 individuals were identified as Black or African American and 1,853 were identified as White. Race, ethnicity, and ancestry were not reported in a uniform manner, which makes ascertainment of the genetic contribution to population differences in preterm birth inequities impossible. When reported as race, ethnicity and ancestry, Black or African American populations were under-represented among the studies in this review. Research of the genomics of preterm birth not only requires increased representation of populations that are disproportionately affected, but it also requires standardized reporting of race, ethnicity, and ancestry.
Collapse
Affiliation(s)
- Liberty Reforma
- Obstetrics and Gynecology, Boston Medical Center, Boston, USA
| | - Simone Greenberg
- School of Public Health, Columbia University College of Physicians and Surgeons, New York, USA
| | - Rachel Ledyard
- Biostatistics, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Heather Burris
- Neonatology, Children's Hospital of Philadelphia, Philadelphia, USA
| |
Collapse
|
9
|
Mead EC, Wang CA, Phung J, Fu JY, Williams SM, Merialdi M, Jacobsson B, Lye S, Menon R, Pennell CE. The Role of Genetics in Preterm Birth. Reprod Sci 2023; 30:3410-3427. [PMID: 37450251 PMCID: PMC10692032 DOI: 10.1007/s43032-023-01287-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
Preterm birth (PTB), defined as the birth of a child before 37 completed weeks gestation, affects approximately 11% of live births and is the leading cause of death in children under 5 years. PTB is a complex disease with multiple risk factors including genetic variation. Much research has aimed to establish the biological mechanisms underlying PTB often through identification of genetic markers for PTB risk. The objective of this review is to present a comprehensive and updated summary of the published data relating to the field of PTB genetics. A literature search in PubMed was conducted and English studies related to PTB genetics were included. Genetic studies have identified genes within inflammatory, immunological, tissue remodeling, endocrine, metabolic, and vascular pathways that may be involved in PTB. However, a substantial proportion of published data have been largely inconclusive and multiple studies had limited power to detect associations. On the contrary, a few large hypothesis-free approaches have identified and replicated multiple novel variants associated with PTB in different cohorts. Overall, attempts to predict PTB using single "-omics" datasets including genomic, transcriptomic, and epigenomic biomarkers have been mostly unsuccessful and have failed to translate to the clinical setting. Integration of data from multiple "-omics" datasets has yielded the most promising results.
Collapse
Affiliation(s)
- Elyse C Mead
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Carol A Wang
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
| | - Jason Phung
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia
- Department of Maternity and Gynaecology, John Hunter Hospital, Newcastle, NSW, 2305, Australia
| | - Joanna Yx Fu
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mario Merialdi
- Maternal Newborn Health Innovations, Geneva, PBC, Switzerland
| | - Bo Jacobsson
- Department of Obstetrics and Gynaecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Obstetrics and Gynaecology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Institute of Public Health, Oslo, Norway
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Craig E Pennell
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, 2308, Australia.
- Hunter Medical Research Institute, Newcastle, NSW, 2305, Australia.
- Department of Maternity and Gynaecology, John Hunter Hospital, Newcastle, NSW, 2305, Australia.
| |
Collapse
|
10
|
Shynlova O, Nadeem L, Lye S. Progesterone control of myometrial contractility. J Steroid Biochem Mol Biol 2023; 234:106397. [PMID: 37683774 DOI: 10.1016/j.jsbmb.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
During pregnancy, the primary function of the uterus is to be quiescent and not contract, which allows the growing fetus to develop and mature. A uterine muscle layer, myometrium, is composed of smooth muscle cells (SMCs). Before the onset of labor contractions, the uterine SMCs experience a complex biochemical and molecular transformation involving the expression of contraction-associated proteins. Labor is initiated when genes in SMCs are activated in response to a combination of hormonal, inflammatory and mechanical signals. In this review, we provide an overview of molecular mechanisms regulating the process of parturition in humans, focusing on the hormonal control of the myometrium, particularly the steroid hormone progesterone. The primary reason for discussing the regulation of myometrial contractility by progesterone is the importance of the clinical problem of preterm birth. It is thought that the hormonal mechanisms regulating premature uterine contractions represent an untimely triggering of the normal events occurring during term parturition. Yet, our knowledge of the complex and redundant hormonal pathways controlling uterine contractile activity leading to delivery of the neonate remains incomplete. Finally, we introduce recent animal studies using a novel class of drugs, Selective Progesterone Receptor Modulators, targeting progesterone signaling to prevent premature myometrial contractions.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada
| |
Collapse
|
11
|
Jin H, Zhang Y, Fan Z, Wang X, Rui C, Xing S, Dong H, Wang Q, Tao F, Zhu Y. Identification of novel cell-free RNAs in maternal plasma as preterm biomarkers in combination with placental RNA profiles. J Transl Med 2023; 21:256. [PMID: 37046301 PMCID: PMC10100253 DOI: 10.1186/s12967-023-04083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/25/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Preterm birth (PTB) is the main driver of newborn deaths. The identification of pregnancies at risk of PTB remains challenging, as the incomplete understanding of molecular mechanisms associated with PTB. Although several transcriptome studies have been done on the placenta and plasma from PTB women, a comprehensive description of the RNA profiles from plasma and placenta associated with PTB remains lacking. METHODS Candidate markers with consistent trends in the placenta and plasma were identified by implementing differential expression analysis using placental tissue and maternal plasma RNA-seq datasets, and then validated by RT-qPCR in an independent cohort. In combination with bioinformatics analysis tools, we set up two protein-protein interaction networks of the significant PTB-related modules. The support vector machine (SVM) model was used to verify the prediction potential of cell free RNAs (cfRNAs) in plasma for PTB and late PTB. RESULTS We identified 15 genes with consistent regulatory trends in placenta and plasma of PTB while the full term birth (FTB) acts as a control. Subsequently, we verified seven cfRNAs in an independent cohort by RT-qPCR in maternal plasma. The cfRNA ARHGEF28 showed consistence in the experimental validation and performed excellently in prediction of PTB in the model. The AUC achieved 0.990 for whole PTB and 0.986 for late PTB. CONCLUSIONS In a comparison of PTB versus FTB, the combined investigation of placental and plasma RNA profiles has shown a further understanding of the mechanism of PTB. Then, the cfRNA identified has the capacity of predicting whole PTB and late PTB.
Collapse
Affiliation(s)
- Heyue Jin
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
| | - Zhigang Fan
- Department of Neonatology, Ma'anshan Maternal and Child Health Hospital, Ma'anshan, Anhui, China
| | - Xianyan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Chen Rui
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Shaozhen Xing
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongmei Dong
- Department of Obstetrics, Ma'anshan Maternal and Child Health Hospital, Ma'anshan, Anhui, China
| | - Qunan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China.
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
| | - Yumin Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
12
|
Dauengauer-Kirlienė S, Domarkienė I, Pilypienė I, Žukauskaitė G, Kučinskas V, Matulevičienė A. Causes of preterm birth: Genetic factors in preterm birth and preterm infant phenotypes. J Obstet Gynaecol Res 2023; 49:781-793. [PMID: 36519629 DOI: 10.1111/jog.15516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
AIM The aim is to provide an overview of recent research on genetic factors that influence preterm birth in the context of neonatal phenotypic assessment. METHODS This is a nonsystematic review of the recent scientific literature. RESULTS Maternal and fetal genetic diversity and rare genome variants are linked with crucial immune response sites. In addition, more frequent in preterm neonates, de novo variants may lead to attention deficits, hyperactivity, autism spectrum disorders, and infertility of both sexes later in life. Environmental factors may also greatly burden fetal, and consequently, neonatal development and neurodevelopment through a failure in the fetal epigenome reprogramming process and even influence the initiation of spontaneous preterm pregnancy termination. Minimally invasive analysis of the transcription factors associated with preterm birth helps elucidate labor mechanisms and predict its timing. We also provide valuable summaries of genomic and transcriptomic factors that contribute to preterm birth. CONCLUSIONS Investigation of the human genome, epigenome, and transcriptome helps to identify molecular mechanisms linked with preterm delivery and premature newborn clinical appearance in early and late neonatal life and even predict developmental outcomes. Further studies are needed to fully understand the implications of genetic changes in preterm births. These data could be used to develop targeted interventions aimed at selecting the most effective individual treatment and rehabilitation plan.
Collapse
Affiliation(s)
- Svetlana Dauengauer-Kirlienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ingrida Domarkienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ingrida Pilypienė
- Clinic of Children's Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Gabrielė Žukauskaitė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vaidutis Kučinskas
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Aušra Matulevičienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
13
|
Li S, Yan B, Li TKT, Lu J, Gu Y, Tan Y, Gong F, Lam TW, Xie P, Wang Y, Lin G, Luo R. Ultra-low-coverage genome-wide association study-insights into gestational age using 17,844 embryo samples with preimplantation genetic testing. Genome Med 2023; 15:10. [PMID: 36788602 PMCID: PMC9926832 DOI: 10.1186/s13073-023-01158-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Very low-coverage (0.1 to 1×) whole genome sequencing (WGS) has become a promising and affordable approach to discover genomic variants of human populations for genome-wide association study (GWAS). To support genetic screening using preimplantation genetic testing (PGT) in a large population, the sequencing coverage goes below 0.1× to an ultra-low level. However, the feasibility and effectiveness of ultra-low-coverage WGS (ulcWGS) for GWAS remains undetermined. METHODS We built a pipeline to carry out analysis of ulcWGS data for GWAS. To examine its effectiveness, we benchmarked the accuracy of genotype imputation at the combination of different coverages below 0.1× and sample sizes from 2000 to 16,000, using 17,844 embryo PGT samples with approximately 0.04× average coverage and the standard Chinese sample HG005 with known genotypes. We then applied the imputed genotypes of 1744 transferred embryos who have gestational ages and complete follow-up records to GWAS. RESULTS The accuracy of genotype imputation under ultra-low coverage can be improved by increasing the sample size and applying a set of filters. From 1744 born embryos, we identified 11 genomic risk loci associated with gestational ages and 166 genes mapped to these loci according to positional, expression quantitative trait locus, and chromatin interaction strategies. Among these mapped genes, CRHBP, ICAM1, and OXTR were more frequently reported as preterm birth related. By joint analysis of gene expression data from previous studies, we constructed interrelationships of mainly CRHBP, ICAM1, PLAGL1, DNMT1, CNTLN, DKK1, and EGR2 with preterm birth, infant disease, and breast cancer. CONCLUSIONS This study not only demonstrates that ulcWGS could achieve relatively high accuracy of adequate genotype imputation and is capable of GWAS, but also provides insights into the associations between gestational age and genetic variations of the fetal embryos from Chinese population.
Collapse
Affiliation(s)
- Shumin Li
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Bin Yan
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Thomas K. T. Li
- grid.415550.00000 0004 1764 4144Department of Obstetrics & Gynecology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Jianliang Lu
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Yifan Gu
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Yueqiu Tan
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Fei Gong
- grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008 Hunan China ,grid.477823.d0000 0004 1756 593XClinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013 Hunan China
| | - Tak-Wah Lam
- grid.194645.b0000000121742757Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Pingyuan Xie
- Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China. .,National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China.
| | - Yuexuan Wang
- Department of Computer Science, The University of Hong Kong, Hong Kong, China. .,College of Computer Science and Technology, Zhejiang University, Hangzhou, China.
| | - Ge Lin
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, 410008, Hunan, China. .,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410013, Hunan, China. .,National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China.
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Paquette AG, MacDonald J, Bammler T, Day DB, Loftus CT, Buth E, Mason WA, Bush NR, Lewinn KZ, Marsit C, Litch JA, Gravett M, Enquobahrie DA, Sathyanarayana S. Placental transcriptomic signatures of spontaneous preterm birth. Am J Obstet Gynecol 2023; 228:73.e1-73.e18. [PMID: 35868418 PMCID: PMC9790028 DOI: 10.1016/j.ajog.2022.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/01/2022] [Accepted: 07/09/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Spontaneous preterm birth accounts for most preterm births and leads to significant morbidity in the newborn and childhood period. This subtype of preterm birth represents an increasing proportion of all preterm births when compared with medically indicated preterm birth, yet it is understudied in omics analyses. The placenta is a key regulator of fetal and newborn health, and the placental transcriptome can provide insight into pathologic changes that lead to spontaneous preterm birth. OBJECTIVE This analysis aimed to identify genes for which placental expression was associated with spontaneous preterm birth (including early preterm and late preterm birth). STUDY DESIGN The ECHO PATHWAYS consortium extracted RNA from placental samples collected from the Conditions Affecting Neurocognitive Development and Learning in Early Childhood and the Global Alliance to Prevent Prematurity and Stillbirth studies. Placental transcriptomic data were obtained by RNA sequencing. Linear models were fit to estimate differences in placental gene expression between term birth and spontaneous preterm birth (including gestational age subgroups defined by the American College of Obstetricians and Gynecologists). Models were adjusted for numerous confounding variables, including labor status, cohort, and RNA sequencing batch. This analysis excluded patients with induced labor, chorioamnionitis, multifetal gestations, or medical indications for preterm birth. Our combined cohort contained gene expression data for 14,023 genes in 48 preterm and 540 term samples. Genes and pathways were considered statistically significantly different at false discovery rate-adjusted P value of <.05. RESULTS In total, we identified 1728 genes for which placental expression was associated with spontaneous preterm birth with more differences in expression in early preterm samples than late preterm samples when compared with full-term samples. Of those, 9 genes were significantly decreased in both early and late spontaneous preterm birth, and the strongest associations involved placental expression of IL1B, ALPL, and CRLF1. In early and late preterm samples, we observed decreased expression of genes involved in immune signaling, signal transduction, and endocrine function. CONCLUSION This study provides a comprehensive assessment of the differences in the placental transcriptome associated with spontaneous preterm birth with robust adjustment for confounding. Results of this study are in alignment with the known etiology of spontaneous preterm birth, because we identified multiple genes and pathways for which the placental and chorioamniotic membrane expression was previously associated with prematurity, including IL1B. We identified decreased expression in key signaling pathways that are essential for placental growth and function, which may be related to the etiology of spontaneous preterm birth. We identified increased expression of genes within metabolic pathways associated exclusively with early preterm birth. These signaling and metabolic pathways may provide clinically targetable pathways and biomarkers. The findings presented here can be used to understand underlying pathologic changes in premature placentas, which can inform and improve clinical obstetrics practice.
Collapse
Affiliation(s)
- Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA; Department of Pediatrics, University of Washington, Seattle, WA.
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Drew B Day
- Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Erin Buth
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - W Alex Mason
- Department of Preventative Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, University of San Francisco, San Francisco, CA; Department of Pediatrics, University of San Francisco, San Francisco, CA
| | - Kaja Z Lewinn
- Department of Psychiatry and Behavioral Sciences, University of San Francisco, San Francisco, CA
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Emory University, Atlanta, GA
| | - James A Litch
- Global Alliance to Prevent Preterm Birth and Stillbirth (GAPPS), Lynnwood, WA
| | - Michael Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | | | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington, Seattle, WA; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA; Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA; Department of Epidemiology, University of Washington, Seattle, WA
| |
Collapse
|
15
|
Illarionov RA, Pachuliia OV, Vashukova ES, Tkachenko AA, Maltseva AR, Postnikova TB, Nasykhova YA, Bespalova ON, Glotov AS. Plasma miRNA Profile in High Risk of Preterm Birth during Early and Mid-Pregnancy. Genes (Basel) 2022; 13:genes13112018. [PMID: 36360255 PMCID: PMC9690526 DOI: 10.3390/genes13112018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
In recent years evidence has been accumulated showing that miRNAs can act as potential biomarkers or targets for therapy of preterm birth, one of the most important problems in modern obstetrics. We have performed a prospective study of the miRNA profile in the plasma during the first and second trimesters in pregnant women with high risk of preterm birth (n = 13 cases and n = 11 controls). For the study group plasma blood samples at 9–13 weeks before diagnosis and at 22–24 weeks after start of therapy were selected. Using high-throughput sequencing technology we detected differences in the levels of 15 miRNAs (3 upregulated—hsa-miR-122-5p, hsa-miR-34a-5p, hsa-miR-34c-5p; 12 downregulated—hsa-miR-487b-3p, hsa-miR-493-3p, hsa-miR-432-5p, hsa-miR-323b-3p, hsa-miR-369-3p, hsa-miR-134-5p, hsa-miR-431-5p, hsa-miR-485-5p, hsa-miR-382-5p, hsa-miR-369-5p, hsa-miR-485-3p, hsa-miR-127-3p) (log2(FC) ≥ 1.5; FDR ≤ 0.05) during the first trimester compared with the control (non-high-risk of preterm birth pregnant women). All downregulated miRNAs in the first trimester from the placenta-specific C14MC cluster. During the second trimester no differentially expressed miRNAs were found. Our results suggest that the miRNA profile in plasma during early pregnancy may predict a high risk of preterm birth, which is important in preventing gestational problems as early as possible.
Collapse
Affiliation(s)
- Roman A. Illarionov
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
- Resource Center “Biobank”, St. Petersburg State University, St. Petersburg 199034, Russia
| | - Olga V. Pachuliia
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Elena S. Vashukova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Alexander A. Tkachenko
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
- Institute of Applied Computer Sciences, ITMO University, St. Petersburg 197101, Russia
| | - Anastasia R. Maltseva
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Tatyana B. Postnikova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Yulia A. Nasykhova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Olesya N. Bespalova
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
| | - Andrey S. Glotov
- Department of Genomic Medicine, D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproduction, St. Petersburg 199034, Russia
- Correspondence:
| |
Collapse
|
16
|
Pearson C, Bartell T, Wang G, Hong X, Rusk SA, Fu L, Cerda S, Bustamante-Helfrich B, Kuohung W, Yarrington C, Adams WG, Wang X. Boston Birth Cohort Profile: Rationale and Study Design. PRECISION NUTRITION 2022; 1:e00011. [PMID: 36660305 PMCID: PMC9844822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In 1998, the Boston Birth Cohort (BBC) was initiated at Boston Medical Center (BMC) in response to persistently high rates of preterm birth (PTB, defined as birth before 37 weeks of gestation) in the US population and the longstanding profound PTB disparity among Black, Indigenous, and people of color (BIPOC). The BBC encompasses two linked study protocols: The Preterm Birth Study serves as the baseline recruitment in the BBC. It aims to address fundamental questions about the causes and consequences of PTB. The study oversamples preterm babies using a case/control study design, in which cases are defined as mothers who deliver a preterm and/or low birthweight baby (<2500 grams regardless of gestational age). Controls are enrolled at a 2:1 control/case ratio and matched by maternal age (±5 years), self-reported race and ethnicity, and date of delivery (± 7 days for case delivery). From inception, it was designed as a comprehensive gene-environmental study of PTB. As a natural extension, the Children's Health Study, under a separate but linked IRB protocol, is a longitudinal follow-up study of the participants who were recruited at birth in the Preterm Birth Study and who continue pediatric care at BMC. This linked model allows for investigation of early life origins of pediatric and chronic disease in a prospective cohort design. The BBC is one of the largest and longest NIH-funded prospective birth cohort studies in the US, consisting of 8733 mother-child dyads enrolled in the Preterm Birth Study at birth, and of those, 3,592 children have been enrolled in the Children's Health Study, with a median follow-up of 14.5 years. The BBC mirrors the urban, under-resourced and underrepresented BIPOC population served by BMC. A high proportion of BBC children were born prematurely and had chronic health conditions (e.g., asthma, obesity and elevated blood pressure) in childhood. The BBC's long-term goal has been to build a large, comprehensive database (epidemiological, clinical, multi-omics) and biospecimen repository to elucidate early life origins of pediatric and chronic diseases and identify modifiable upstream factors (e.g., psychosocial, environmental, nutritional) to improve health across the life course for BIPOC mothers and children.
Collapse
Affiliation(s)
- Colleen Pearson
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Tami Bartell
- Patrick M. Magoon Institute for Healthy Communities, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Serena A. Rusk
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - LingLing Fu
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Sandra Cerda
- Department of Pathology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | | | - Wendy Kuohung
- Department of Obstetrics and Gynecology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Christina Yarrington
- Department of Obstetrics and Gynecology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - William G. Adams
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Jain VG, Monangi N, Zhang G, Muglia LJ. Genetics, epigenetics, and transcriptomics of preterm birth. Am J Reprod Immunol 2022; 88:e13600. [PMID: 35818963 PMCID: PMC9509423 DOI: 10.1111/aji.13600] [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: 03/25/2022] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Preterm birth contributes significantly to neonatal mortality and morbidity. Despite its global significance, there has only been limited progress in preventing preterm birth. Spontaneous preterm birth (sPTB) results from a wide variety of pathological processes. Although many non-genetic risk factors influence the timing of gestation and labor, compelling evidence supports the role of substantial genetic and epigenetic influences and their interactions with the environment contributing to sPTB. To investigate a common and complex disease such as sPTB, various approaches such as genome-wide association studies, whole-exome sequencing, transcriptomics, and integrative approaches combining these with other 'omics studies have been used. However, many of these studies were typically small or focused on a single ethnicity or geographic region with limited data, particularly in populations at high risk for sPTB, or lacked a robust replication. These studies found many genes involved in the inflammation and immunity-related pathways that may affect sPTB. Recent studies also suggest the role of epigenetic modifications of gene expression by the environmental signals as a potential contributor to the risk of sPTB. Future genetic studies of sPTB should continue to consider the contributions of both maternal and fetal genomes as well as their interaction with the environment.
Collapse
Affiliation(s)
- Viral G. Jain
- Division of Neonatology, Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nagendra Monangi
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ge Zhang
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Louis J. Muglia
- Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children’s Hospital Medical Center and March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Burroughs Wellcome Fund, Research Triangle Park, North Carolina, USA
| |
Collapse
|
18
|
Loveday C, Garrett A, Law P, Hanks S, Poyastro-Pearson E, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Chapman C, Cook J, Davidson R, Donaldson A, Douglas F, Greenhalgh L, Henderson A, Izatt L, Kumar A, Lalloo F, Miedzybrodzka Z, Morrison PJ, Paterson J, Porteous M, Rogers MT, Walker L, Eccles D, Evans DG, Snape K, Hanson H, Houlston RS, Turnbull C. Analysis of rare disruptive germline mutations in 2,135 enriched BRCA-negative breast cancers excludes additional high-impact susceptibility genes. Ann Oncol 2022; 33:1318-1327. [PMID: 36122798 DOI: 10.1016/j.annonc.2022.09.152] [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: 03/03/2022] [Revised: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Breast cancer has a significant heritable basis, of which approximately 60% remains unexplained. Testing for BRCA1/BRCA2 offers useful discrimination of breast cancer risk within families, and identification of additional breast cancer susceptibility genes could offer clinical utility. PATIENTS AND METHODS We included 2,135 invasive breast cancer cases recruited via the BOCS study, a retrospective UK study of familial breast cancer. ELIGIBILITY CRITERIA female, BRCA-negative, white European ethnicity, and one of: i) breast cancer family history, ii) bilateral disease, iii) young age of onset (<30 years), iv) concomitant ovarian cancer. We undertook exome sequencing of cases and performed gene-level burden testing of rare damaging variants against those from 51,377 ethnicity-matched population controls from gnomAD. RESULTS 159/2135 (7.4%) cases had a qualifying variant in an established breast cancer susceptibility gene, with minimal evidence of signal in other cancer susceptibility genes. Known breast cancer susceptibility genes PALB2, CHEK2 and ATM were the only genes to retain statistical significance after correcting for multiple testing. Due to the enrichment of hereditary cases in the series, we had good power (>80%) to detect a gene of BRCA1-like risk (odds ratio = 10.6) down to a population minor allele frequency of 4.6 x 10-5 (1 in 10,799, less than one tenth that of BRCA1)and of PALB2-like risk (odds ratio = 5.0) down to a population minor allele frequency of 2.8 x 10-4 (1 in 1,779, less than half that of PALB2). Power was lower for identification of novel moderate penetrance genes (odds ratio = 2-3) like CHEK2 and ATM. CONCLUSIONS This is the largest case-control whole-exome analysis of enriched breast cancer published to date. Whilst additional breast cancer susceptibility genes likely exist, those of high penetrance are likely to be of very low mutational frequency. Contention exists regarding the clinical utility of such genes.
Collapse
Affiliation(s)
- C Loveday
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - A Garrett
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - P Law
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - S Hanks
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - E Poyastro-Pearson
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - J W Adlard
- Yorkshire Regional Genetics Service, St James's University Hospital, Leeds, UK
| | - J Barwell
- Leicestershire Genetics Centre, University Hospitals of Leicester National Health Service (NHS) Trust, Leicester, UK
| | - J Berg
- Division of Medical Sciences, Human Genetics, University of Dundee, Dundee, UK
| | - A F Brady
- North West Thames Regional Genetics Service, Kennedy Galton Centre, London, UK
| | - C Brewer
- Peninsula Regional Genetics Service, Royal Devon & Exeter Hospital, Exeter, UK
| | - C Chapman
- West Midlands Regional Genetics Service, Birmingham Women's Hospital, Birmingham, UK
| | - J Cook
- Sheffield Regional Genetics Service, Sheffield Children's NHS Foundation Trust, Sheffield, UK
| | - R Davidson
- West of Scotland Regional Genetics Service, Ferguson Smith Centre for Clinical Genetics, Glasgow, UK
| | - A Donaldson
- South Western Regional Genetics Service, University Hospitals of Bristol NHS Foundation Trust, Bristol, UK
| | - F Douglas
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - L Greenhalgh
- Cheshire and Merseyside Clinical Genetics Service, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - A Henderson
- Northern Genetics Service (Cumbria), Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - L Izatt
- South East Thames Regional Genetics Service, Guy's and St. Thomas NHS Foundation Trust, London, UK
| | - A Kumar
- North East Thames Regional Genetics Service, Great Ormond St. Hospital, London, UK
| | - F Lalloo
- University Department of Medical Genetics & Regional Genetics Service, St. Mary's Hospital, Manchester, UK
| | - Z Miedzybrodzka
- University of Aberdeen and North of Scotland Clinical Genetics Service, Aberdeen Royal Infirmary, Aberdeen, UK
| | - P J Morrison
- Belfast Health and Social Care (HSC) Trust & Department of Medical Genetics, Northern Ireland Regional Genetics Service, Queen's University Belfast, Belfast, UK
| | - J Paterson
- East Anglian Regional Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - M Porteous
- South East of Scotland Clinical Genetics Service, Western General Hospital, Edinburgh, UK
| | - M T Rogers
- All Wales Medical Genetics Service, University Hospital of Wales, Cardiff, UK
| | - L Walker
- Oxford Regional Genetics Service, Oxford Radcliffe Hospitals NHS Trust, Oxford, UK
| | -
- Individual collaborators and their affiliations are listed in the Appendix
| | - D Eccles
- Faculty of Medicine, University of Southampton, Southampton University Hospitals NHS Trust, Southampton, UK
| | - D G Evans
- University Department of Medical Genetics & Regional Genetics Service, St. Mary's Hospital, Manchester, UK
| | - K Snape
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.; South West Thames Regional Genetics Service, St. George's Hospital, London, UK
| | - H Hanson
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.; South West Thames Regional Genetics Service, St. George's Hospital, London, UK
| | - R S Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - C Turnbull
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.; Royal Marsden NHS Foundation Hospital, London, UK.
| |
Collapse
|
19
|
Pearson C, Bartell T, Wang G, Hong X, Rusk SA, Fu L, Cerda S, Bustamante-Helfrich B, Kuohung W, Yarrington C, Adams WG, Wang X. Boston Birth Cohort profile: rationale and study design. PRECISION NUTRITION 2022; 1:e00011. [PMID: 37745944 PMCID: PMC9844822 DOI: 10.1097/pn9.0000000000000011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 09/26/2023]
Abstract
In1998, the Boston Birth Cohort (BBC) was initiated at Boston Medical Center (BMC) in response to persistently high rates of preterm birth (PTB, defined as birth before 37 weeks of gestation) in the US population and the longstanding profound PTB disparity among Black, Indigenous, and people of color (BIPOC). The BBC encompasses two linked study protocols: The PTB Study serves as the baseline recruitment in the BBC. It aims to address fundamental questions about the causes and consequences of PTB. The study oversamples preterm babies using a case/control study design, in which cases are defined as mothers who deliver a preterm and/or low birthweight baby (<2500 grams regardless of gestational age). Controls are enrolled at a 2:1 control/case ratio and matched by maternal age (±5 years), self-reported race and ethnicity, and date of delivery (± 7 days for case delivery). From inception, it was designed as a comprehensive gene-environmental study of PTB. As a natural extension, the Children's Health Study, under a separate but linked Institutional Review Board protocol, is a longitudinal follow-up study of the participants who were recruited at birth in the PTB Study and who continue pediatric care at BMC. This linked model allows for investigation of early life origins of pediatric and chronic disease in a prospective cohort design. The BBC is one of the largest and longest National Institutes of Health-funded prospective birth cohort studies in the United States, consisting of 8733 mother-child dyads enrolled in the PTB Study at birth, and of those, 3592 children have been enrolled in the Children's Health Study, with a median follow-up of 14.5 years. The BBC mirrors the urban, underresourced, and underrepresented BIPOC population served by BMC. A high proportion of BBC children were born prematurely and had chronic health conditions (e.g., asthma, obesity, and elevated blood pressure) in childhood. The BBC's long-term goal has been to build a large, comprehensive database (epidemiological, clinical, and multiomics) and biospecimen repository to elucidate early life origins of pediatric and chronic diseases and identify modifiable upstream factors (e.g., psychosocial, environmental, and nutritional) to improve health across the life course for BIPOC mothers and children.
Collapse
Affiliation(s)
- Colleen Pearson
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Tami Bartell
- Patrick M. Magoon Institute for Healthy Communities, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Serena A. Rusk
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - LingLing Fu
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Sandra Cerda
- Department of Pathology, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | | | - Wendy Kuohung
- Department of Obstetrics and Gynecology, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Christina Yarrington
- Department of Obstetrics and Gynecology, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - William G. Adams
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Asif H, Barnett SD, Buxton ILO. Title: β3 Adrenergic Receptor Signaling in the Human Myometrium. Reprod Sci 2022; 30:124-134. [PMID: 35380411 PMCID: PMC8980516 DOI: 10.1007/s43032-022-00917-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/11/2022] [Indexed: 01/06/2023]
Abstract
Preterm labor leading to preterm birth is the leading cause of infant morbidity and mortality. Although β2 adrenergic agonists fail to provide adequate tocolysis, the expression of the β3 adrenergic receptor in myometrium and its unique signaling suggest a role for β3 agonist in the management of preterm labor. Western blot analysis showed that the β3 adrenergic receptor expression increased in human pregnancy myometrium compared to nonpregnant tissues (p < 0.0001). There was no difference in β3 adrenergic receptor expression throughout pregnancy (p > 0.05). The addition of the β3 agonist mirabegron in the tissue bath relaxed oxytocin contracted myometrium with an EC50 of 41.5 µM. Relaxation was partially blocked by the addition of the eNOS blocker Nω-nitro-L-arginine, or the large conductance potassium channel blocker paxilline. Combination of Nω-nitro-L-arginine and paxilline prevented mirabegron-mediated relaxation. Imaging revealed that the β3 adrenergic receptors are expressed by both myocyte and microvascular endothelial cells isolated from human myometrium. Nitric oxide production measured by 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate revealed that mirabegron stimulated nitric oxide production in myometrial endothelial cells. These data suggest that both endothelial and smooth muscle cells contribute to relaxation through disparate signaling pathways. Repurposing of approved medications tested in human myometrium as uterine tocolytics can advance prevention of preterm birth. These data argue that further examination of β3 adrenergic receptor signaling in myometrium may reveal mirabegron as a useful tocolytic in combination tocolysis regimens.
Collapse
Affiliation(s)
- Hazik Asif
- School of Medicine, Department of Pharmacology, Myometrial Function Laboratory, University of Nevada, Reno, NV 89557-0318 USA
| | - Scott D. Barnett
- School of Medicine, Department of Pharmacology, Myometrial Function Laboratory, University of Nevada, Reno, NV 89557-0318 USA
| | - Iain L. O. Buxton
- School of Medicine, Department of Pharmacology, Myometrial Function Laboratory, University of Nevada, Reno, NV 89557-0318 USA
| |
Collapse
|
21
|
Winchester P, Nilsson E, Beck D, Skinner MK. Preterm birth buccal cell epigenetic biomarkers to facilitate preventative medicine. Sci Rep 2022; 12:3361. [PMID: 35232984 PMCID: PMC8888575 DOI: 10.1038/s41598-022-07262-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 02/11/2022] [Indexed: 11/09/2022] Open
Abstract
Preterm birth is the major cause of newborn and infant mortality affecting nearly one in every ten live births. The current study was designed to develop an epigenetic biomarker for susceptibility of preterm birth using buccal cells from the mother, father, and child (triads). An epigenome-wide association study (EWAS) was used to identify differential DNA methylation regions (DMRs) using a comparison of control term birth versus preterm birth triads. Epigenetic DMR associations with preterm birth were identified for both the mother and father that were distinct and suggest potential epigenetic contributions from both parents. The mother (165 DMRs) and female child (136 DMRs) at p < 1e-04 had the highest number of DMRs and were highly similar suggesting potential epigenetic inheritance of the epimutations. The male child had negligible DMR associations. The DMR associated genes for each group involve previously identified preterm birth associated genes. Observations identify a potential paternal germline contribution for preterm birth and identify the potential epigenetic inheritance of preterm birth susceptibility for the female child later in life. Although expanded clinical trials and preconception trials are required to optimize the potential epigenetic biomarkers, such epigenetic biomarkers may allow preventative medicine strategies to reduce the incidence of preterm birth.
Collapse
Affiliation(s)
- Paul Winchester
- Department of Pediatrics, St. Franciscan Hospital, School of Medicine, Indiana University, Indianapolis, IN, 46202-5201, USA
| | - Eric Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, 99164-4236, USA
| | - Daniel Beck
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, 99164-4236, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, 99164-4236, USA.
| |
Collapse
|
22
|
Ran Y, He J, Peng W, Liu Z, Mei Y, Zhou Y, Yin N, Qi H. Development and validation of a transcriptomic signature-based model as the predictive, preventive, and personalized medical strategy for preterm birth within 7 days in threatened preterm labor women. EPMA J 2022; 13:87-106. [PMID: 35273661 PMCID: PMC8897543 DOI: 10.1007/s13167-021-00268-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 12/08/2022]
Abstract
Preterm birth (PTB) is the leading cause of neonatal death. The essential strategy to prevent PTB is the accurate identification of threatened preterm labor (TPTL) women who will have PTB in a short time (< 7 days). Here, we aim to propose a clinical model to contribute to the effective prediction, precise prevention, and personalized medical treatment for PTB < 7 days in TPTL women through bioinformatics analysis and prospective cohort studies. In this study, the 1090 key genes involved in PTB < 7 days in the peripheral blood of TPTL women were ascertained using WGCNA. Based on this, the biological basis of immune-inflammatory activation (e.g., IFNγ and TNFα signaling) as well as immune cell disorders (e.g., monocytes and Th17 cells) in PTB < 7 days were revealed. Then, four core genes (JOSD1, IDNK, ZMYM3, and IL1B) that best represent their transcriptomic characteristics were screened by SVM and LASSO algorithm. Therefore, a prediction model with an AUC of 0.907 was constructed, which was validated in a larger population (AUC = 0.783). Moreover, the predictive value (AUC = 0.957) and clinical feasibility of this model were verified through the clinical prospective cohort we established. In conclusion, in the context of Predictive, Preventive, and Personalized Medicine (3PM), we have developed and validated a model to predict PTB < 7 days in TPTL women. This is promising to greatly improve the accuracy of clinical prediction, which would facilitate the personalized management of TPTL women to precisely prevent PTB < 7 days and improve maternal-fetal outcomes.
Collapse
Affiliation(s)
- Yuxin Ran
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Health Center for Women and Children, No. 120 Longshan Road, Yubei District, Chongqing, 401120 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Jie He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Wei Peng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Zheng Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Youwen Mei
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Yunqian Zhou
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| | - Nanlin Yin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Center for Reproductive Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Rd, Yuzhong District, Chongqing, 400016 China
| | - Hongbo Qi
- Chongqing Health Center for Women and Children, No. 120 Longshan Road, Yubei District, Chongqing, 401120 China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
- Joint International Research Laboratory of Reproduction and Development of Chinese Ministry of Education, Chongqing Medical University, No. 1 Yixueyuan Rd, Yuzhong District, Chongqing, 400016 China
| |
Collapse
|
23
|
Protein interaction networks define the genetic architecture of preterm birth. Sci Rep 2022; 12:438. [PMID: 35013336 PMCID: PMC8748950 DOI: 10.1038/s41598-021-03427-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 02/10/2021] [Indexed: 11/20/2022] Open
Abstract
The likely genetic architecture of complex diseases is that subgroups of patients share variants in genes in specific networks sufficient to express a shared phenotype. We combined high throughput sequencing with advanced bioinformatic approaches to identify such subgroups of patients with variants in shared networks. We performed targeted sequencing of patients with 2 or 3 generations of preterm birth on genes, gene sets and haplotype blocks that were highly associated with preterm birth. We analyzed the data using a multi-sample, protein–protein interaction (PPI) tool to identify significant clusters of patients associated with preterm birth. We identified shared protein interaction networks among preterm cases in two statistically significant clusters, p < 0.001. We also found two small control-dominated clusters. We replicated these data on an independent, large birth cohort. Separation testing showed significant similarity scores between the clusters from the two independent cohorts of patients. Canonical pathway analysis of the unique genes defining these clusters demonstrated enrichment in inflammatory signaling pathways, the glucocorticoid receptor, the insulin receptor, EGF and B-cell signaling, These results support a genetic architecture defined by subgroups of patients that share variants in genes in specific networks and pathways which are sufficient to give rise to the disease phenotype.
Collapse
|
24
|
Preterm Labor, a Syndrome Attributed to the Combination of External and Internal Factors. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
25
|
Li X, Liu L, Whitehead C, Li J, Thierry B, Le TD, Winter M. OUP accepted manuscript. Brief Funct Genomics 2022; 21:296-309. [PMID: 35484822 PMCID: PMC9328024 DOI: 10.1093/bfgp/elac006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia is a pregnancy-specific disease that can have serious effects on the health of both mothers and their offspring. Predicting which women will develop preeclampsia in early pregnancy with high accuracy will allow for improved management. The clinical symptoms of preeclampsia are well recognized, however, the precise molecular mechanisms leading to the disorder are poorly understood. This is compounded by the heterogeneous nature of preeclampsia onset, timing and severity. Indeed a multitude of poorly defined causes including genetic components implicates etiologic factors, such as immune maladaptation, placental ischemia and increased oxidative stress. Large datasets generated by microarray and next-generation sequencing have enabled the comprehensive study of preeclampsia at the molecular level. However, computational approaches to simultaneously analyze the preeclampsia transcriptomic and network data and identify clinically relevant information are currently limited. In this paper, we proposed a control theory method to identify potential preeclampsia-associated genes based on both transcriptomic and network data. First, we built a preeclampsia gene regulatory network and analyzed its controllability. We then defined two types of critical preeclampsia-associated genes that play important roles in the constructed preeclampsia-specific network. Benchmarking against differential expression, betweenness centrality and hub analysis we demonstrated that the proposed method may offer novel insights compared with other standard approaches. Next, we investigated subtype specific genes for early and late onset preeclampsia. This control theory approach could contribute to a further understanding of the molecular mechanisms contributing to preeclampsia.
Collapse
Affiliation(s)
- Xiaomei Li
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Lin Liu
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Clare Whitehead
- Pregnancy Research Centre, Dept of Obstetrics & Gynaecology, University of Melbourne, Royal Women’s Hospital, Melbourne, 3052, VIC, Australia
| | - Jiuyong Li
- UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia
| | - Thuc D Le
- Corresponding authors: Thuc D. Le, UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail: ; M. Winter, Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail:
| | - Marnie Winter
- Corresponding authors: Thuc D. Le, UniSA STEM, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail: ; M. Winter, Future Industries Institute, University of South Australia, Mawson Lakes, 5095, SA, Australia. E-mail:
| |
Collapse
|
26
|
Ferreira G, Blasina F, Rodríguez Rey M, Anesetti G, Sapiro R, Chavarría L, Cardozo R, Rey G, Sobrevia L, Nicolson GL. Pathophysiological and molecular considerations of viral and bacterial infections during maternal-fetal and -neonatal interactions of SARS-CoV-2, Zika, and Mycoplasma infectious diseases. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166285. [PMID: 34624499 PMCID: PMC8492386 DOI: 10.1016/j.bbadis.2021.166285] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 01/04/2023]
Abstract
During pregnancy, a series of physiological changes are determined at the molecular, cellular and macroscopic level that make the mother and fetus more susceptible to certain viral and bacterial infections, especially the infections in this and the companion review. Particular situations increase susceptibility to infection in neonates. The enhanced susceptibility to certain infections increases the risk of developing particular diseases that can progress to become morbidly severe. For example, during the current pandemic caused by the SARS-CoV-2 virus, epidemiological studies have established that pregnant women with COVID-19 disease are more likely to be hospitalized. However, the risk for intensive care unit admission and mechanical ventilation is not increased compared with nonpregnant women. Although much remains unknown with this particular infection, the elevated risk of progression during pregnancy towards more severe manifestations of COVID-19 disease is not associated with an increased risk of death. In addition, the epidemiological data available in neonates suggest that their risk of acquiring COVID-19 is low compared with infants (<12 months of age). However, they might be at higher risk for progression to severe COVID-19 disease compared with older children. The data on clinical presentation and disease severity among neonates are limited and based on case reports and small case series. It is well documented the importance of the Zika virus infection as the main cause of several congenital anomalies and birth defects such as microcephaly, and also adverse pregnancy outcomes. Mycoplasma infections also increase adverse pregnancy outcomes. This review will focus on the molecular, pathophysiological and biophysical characteristics of the mother/placental-fetal/neonatal interactions and the possible mechanisms of these pathogens (SARS-CoV-2, ZIKV, and Mycoplasmas) for promoting disease at this level.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay.
| | - Fernanda Blasina
- Dept. of Neonatology, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Marianela Rodríguez Rey
- Dept. of Neonatology, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Gabriel Anesetti
- Dept. of Histology and Development, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Rosana Sapiro
- Dept. of Histology and Development, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Luisina Chavarría
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Romina Cardozo
- Laboratory of Ion Channels, Biological Membranes and Cell Signaling, Dept. of Biophysics, Facultad de Medicina, Universidad de la Republica, Montevideo, Uruguay
| | - Grazzia Rey
- Dept. of Clinical Ginecology and Obstetrics B, Facultad de Medicina, Universidad de la Republica, Uruguay
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; Medical School, Faculty of Medicine, Sao Paulo State University (UNESP), Brazil; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands
| | - Garth L Nicolson
- Department of Molecular Pathology, The Institute for Molecular Medicine, Huntington Beach, CA, USA
| |
Collapse
|
27
|
Wood TR, Parikh P, Comstock BA, Law JB, Bammler TK, Kuban KC, Mayock DE, Heagerty PJ, Juul S. Early Biomarkers of Hypoxia and Inflammation and Two-Year Neurodevelopmental Outcomes in the Preterm Erythropoietin Neuroprotection (PENUT) Trial. EBioMedicine 2021; 72:103605. [PMID: 34619638 PMCID: PMC8498235 DOI: 10.1016/j.ebiom.2021.103605] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/24/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND In the Preterm Erythropoietin (Epo) NeUroproTection (PENUT) Trial, potential biomarkers of neurological injury were measured to determine their association with outcomes at two years of age and whether Epo treatment decreased markers of inflammation in extremely preterm (<28 weeks' gestation) infants. METHODS Plasma Epo was measured (n=391 Epo, n=384 placebo) within 24h after birth (baseline), 30min after study drug administration (day 7), 30min before study drug (day 9), and on day 14. A subset of infants (n=113 Epo, n=107 placebo) had interferon-gamma (IFN-γ), Interleukin (IL)-6, IL-8, IL-10, Tau, and tumour necrosis factor-α (TNF-α) levels evaluated at baseline, day 7 and 14. Infants were then evaluated at 2 years using the Bayley Scales of Infant and Toddler Development, 3rd Edition (BSID-III). FINDINGS Elevated baseline Epo was associated with increased risk of death or severe disability (BSID-III Motor and Cognitive subscales <70 or severe cerebral palsy). No difference in other biomarkers were seen between treatment groups at any time, though Epo appeared to mitigate the association between elevated baseline IL-6 and lower BSID-III scores in survivors. Elevated baseline, day 7 and 14 Tau concentrations were associated with worse BSID-III Cognitive, Motor, and Language skills at two years. INTERPRETATION Elevated Epo at baseline and elevated Tau in the first two weeks after birth predict poor outcomes in infants born extremely preterm. However, no clear prognostic cut-off values are apparent, and further work is required before these biomarkers can be widely implemented in clinical practice. FUNDING PENUT was funded by the National Institute of Neurological Disorders and Stroke (U01NS077955 and U01NS077953).
Collapse
Affiliation(s)
- Thomas R. Wood
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - Pratik Parikh
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | | | - Janessa B. Law
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Karl C. Kuban
- Department of Pediatrics, Boston University School of Medicine, Boston, MA
| | - Dennis E. Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | | | - Sandra Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
| | - for the PENUT Trial consortium
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
- Department of Pediatrics, Boston University School of Medicine, Boston, MA
| |
Collapse
|
28
|
The Preventive Effects of Quercetin on Preterm Birth Based on Network Pharmacology and Bioinformatics. Reprod Sci 2021; 29:193-202. [PMID: 34231170 DOI: 10.1007/s43032-021-00674-4] [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: 03/01/2021] [Accepted: 06/17/2021] [Indexed: 10/20/2022]
Abstract
Our previous study has shown that quercetin prevented lipopolysaccharide-induced preterm birth. This study aims to clarify the potential targets and biological mechanisms of quercetin in preventing preterm birth. We used bioinformatics databases to collect the candidate targets for quercetin and preterm birth. The biological functions and enriched pathways of the intersecting targets were analyzed by gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. Then, the hub targets were identified by cytoscape plugin cytoHubba from the protein-protein interaction network. We obtained 105 targets for quercetin in preventing preterm birth. The biological processes of the intersecting targets are mainly involved in steroid metabolic process, drug metabolic process, oxidation-reduction process, omega-hydroxylase P450 pathway, positive regulation of cell migration, negative regulation of apoptotic process, and positive regulation of cell proliferation. The highly enriched pathways were steroid hormone biosynthesis, metabolism of xenobiotics by cytochrome P450, proteoglycans in cancer, focal adhesion, and arachidonic acid metabolism. The ten hub targets for quercetin in preventing preterm birth were AKT serine/threonine kinase 1, mitogen-activated protein kinase 3, epidermal growth factor receptor, prostaglandin-endoperoxide synthase 2, mitogen-activated protein kinase 1, estrogen receptor 1, heat shock protein 90 alpha family class A member 1, mitogen-activated protein kinase 8, androgen receptor, and matrix metallopeptidase 9. Molecular docking analysis showed good bindings between these proteins and quercetin. In conclusion, these findings highlight the key targets and molecular mechanisms of quercetin in preventing preterm birth.
Collapse
|
29
|
Tarca AL, Pataki BÁ, Romero R, Sirota M, Guan Y, Kutum R, Gomez-Lopez N, Done B, Bhatti G, Yu T, Andreoletti G, Chaiworapongsa T, Hassan SS, Hsu CD, Aghaeepour N, Stolovitzky G, Csabai I, Costello JC. Crowdsourcing assessment of maternal blood multi-omics for predicting gestational age and preterm birth. Cell Rep Med 2021; 2:100323. [PMID: 34195686 PMCID: PMC8233692 DOI: 10.1016/j.xcrm.2021.100323] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/18/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Identification of pregnancies at risk of preterm birth (PTB), the leading cause of newborn deaths, remains challenging given the syndromic nature of the disease. We report a longitudinal multi-omics study coupled with a DREAM challenge to develop predictive models of PTB. The findings indicate that whole-blood gene expression predicts ultrasound-based gestational ages in normal and complicated pregnancies (r = 0.83) and, using data collected before 37 weeks of gestation, also predicts the delivery date in both normal pregnancies (r = 0.86) and those with spontaneous preterm birth (r = 0.75). Based on samples collected before 33 weeks in asymptomatic women, our analysis suggests that expression changes preceding preterm prelabor rupture of the membranes are consistent across time points and cohorts and involve leukocyte-mediated immunity. Models built from plasma proteomic data predict spontaneous preterm delivery with intact membranes with higher accuracy and earlier in pregnancy than transcriptomic models (AUROC = 0.76 versus AUROC = 0.6 at 27-33 weeks of gestation).
Collapse
Affiliation(s)
- Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA
| | - Bálint Ármin Pataki
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL 33199, USA
| | - Marina Sirota
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rintu Kutum
- Informatics and Big Data Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | | | - Gaia Andreoletti
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tinnakorn Chaiworapongsa
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - The DREAM Preterm Birth Prediction Challenge Consortium
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL 33199, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Informatics and Big Data Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Sage Bionetworks, Seattle, WA, USA
- Office of Women’s Health, Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Pediatrics, and Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sonia S. Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Office of Women’s Health, Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Department of Pediatrics, and Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gustavo Stolovitzky
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Istvan Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
30
|
Spontaneous preterm birth: the underpinnings in the maternal and fetal genomes. NPJ Genom Med 2021; 6:43. [PMID: 34103530 PMCID: PMC8187433 DOI: 10.1038/s41525-021-00209-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022] Open
Abstract
Preterm birth (PTB) is a major cause of neonatal mortality and health complications in infants. Elucidation of its genetic underpinnings can lead to improved understanding of the biological mechanisms and boost the development of methods to predict PTB. Although recent genome-based studies of both mother and fetus have identified several genetic loci which might be implicated in PTB, these results suffer from a lack of consistency across multiple studies and populations. Moreover, results of functional validation of most of these findings are unavailable. Since medically indicated preterm deliveries have well-known heterogeneous causes, we have reviewed only those studies which investigated spontaneous preterm birth (sPTB) and have attempted to suggest probable biological mechanisms by which the implicated genetic factors might result in sPTB. We expect our review to provide a panoramic view of the genetics of sPTB.
Collapse
|
31
|
Wei CF, Lin CC, Tsai MS, Guo YL, Lin SJ, Liao HF, Hsieh WS, Chen MH, Chen PC. Associations between household incense burning and delayed motor development among preterm infants modified by gestational age and maternal educational status. INDOOR AIR 2021; 31:660-672. [PMID: 33090582 DOI: 10.1111/ina.12762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/20/2020] [Accepted: 09/30/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Household incense burning is a common ritual behavior in the Asia-Pacific region but has been associated with inferior developmental outcomes in term infants. We aimed to examine these associations among preterm infants. METHODS Information from 1190 mother-infant pairs during 6- and 18-month follow-up to the Taiwan Birth Cohort Study was examined for associations between household incense burning exposure and infant neurodevelopmental milestone achievement using multivariable Cox proportional hazard model with propensity score weighting, along with stratified, sensitivity, and decomposition analysis. RESULTS Household incense burning exposure was associated with delayed gross motor milestone achievement among all preterm infants according to the Cox model and after propensity score weighting. Meanwhile, associations for delayed development were found in gross motor domain milestones among late preterm infants, while fine motor domain delay was found among other preterm infants. Furthermore, the associations between household incense burning status and gross motor milestone delays were attenuated by the interaction between higher education level and household incense burning exposure status. CONCLUSIONS Household incense burning exposure was associated with delays, and the motor domains affected differed according to degree of prematurity. These associations were modified by the attenuation upon higher maternal educational status and exposure status interaction.
Collapse
Affiliation(s)
- Chih-Fu Wei
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
- Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Ching-Chun Lin
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Meng-Shan Tsai
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Yueliang L Guo
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
- Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Shio-Jean Lin
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Hua-Fang Liao
- School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wu-Shiun Hsieh
- Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan
- Department of Pediatrics, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - Mei-Huei Chen
- Department of Pediatrics, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Pau-Chung Chen
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health, Taipei, Taiwan
- Department of Environmental and Occupational Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
- Department of Public Health, National Taiwan University College of Public Health, Taipei, Taiwan
- Innovation and Policy Center for Population Health and Sustainable Environment, National Taiwan University College of Public Health, Taipei, Taiwan
| |
Collapse
|
32
|
Identification of candidate genes and pathways in retinopathy of prematurity by whole exome sequencing of preterm infants enriched in phenotypic extremes. Sci Rep 2021; 11:4966. [PMID: 33654115 PMCID: PMC7925531 DOI: 10.1038/s41598-021-83552-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/01/2021] [Indexed: 01/05/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative retinal disease affecting premature infants. In addition to prematurity itself and oxygen treatment, genetic factors have been suggested to predispose to ROP. We aimed to identify potentially pathogenic genes and biological pathways associated with ROP by analyzing variants from whole exome sequencing (WES) data of premature infants. As part of a multicenter ROP cohort study, 100 non-Hispanic Caucasian preterm infants enriched in phenotypic extremes were subjected to WES. Gene-based testing was done on coding nonsynonymous variants. Genes showing enrichment of qualifying variants in severe ROP compared to mild or no ROP from gene-based tests with adjustment for gestational age and birth weight were selected for gene set enrichment analysis (GSEA). Mean BW of included infants with pre-plus, type-1 or type 2 ROP including aggressive posterior ROP (n = 58) and mild or no ROP (n = 42) were 744 g and 995 g, respectively. No single genes reached genome-wide significance that could account for a severe phenotype. GSEA identified two significantly associated pathways (smooth endoplasmic reticulum and vitamin C metabolism) after correction for multiple tests. WES of premature infants revealed potential pathways that may be important in the pathogenesis of ROP and in further genetic studies.
Collapse
|
33
|
Barnett SD, Asif H, Anderson M, Buxton ILO. Novel Tocolytic Strategy: Modulating Cx43 Activity by S-Nitrosation. J Pharmacol Exp Ther 2021; 376:444-453. [PMID: 33384302 PMCID: PMC7919864 DOI: 10.1124/jpet.120.000427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Currently available tocolytics are ineffective at significantly delaying preterm birth. This is due in part to our failure to better understand the mechanisms that drive spontaneous preterm labor (sPTL). Cyclic nucleotides are not the primary contributors to myometrial quiescence, but instead nitric oxide (NO)-mediated protein S-nitrosation (SNO) is integral to the relaxation of the tissue. Connexin-43 (Cx43), a myometrial "contractile-associated protein" that functions as either a gap junction channel or an hemichannel (HC), was the focus of this study. Protein analysis determined that Cx43 is downregulated in sPTL myometrium. Furthermore, Cx43 is S-nitrosated by NO, which correlates with an increase of phosphorylated Cx43 at serine 368 (Cx43-pS368 -gap junction inhibition) as well as an increase in the HC open-state probability (quiescence). Pharmacologic inhibition of Cx43 with 18β-glycyrrhetinic acid (18β-GA) exhibits a negative inotropic effect on the myometrium in a dose-dependent manner, as does administration of nebivolol, an NO synthase activator that increases total protein SNOs. When 18β-GA and nebivolol were coadministered at their IC50 values, the effect on contractile dynamics was additive and all but eliminated contractions. The development of new tocolytics demands a better understanding of the underlying mechanisms of sPTL. Here it has been shown that 18β-GA and nebivolol leverage dysregulated pathways in the myometrium, resulting in a novel approach for the treatment of sPTL. SIGNIFICANCE STATEMENT: Although there are many known causes of preterm labor (PTL), the mechanisms of "spontaneous" PTL (sPTL) remain obfuscated, which is why treating this condition is so challenging. Here we have identified that connexin-43 (Cx43), an important contractile-associated protein, is dysregulated in sPTL myometrium and that the pharmacologic inhibition of Cx43 and its S-nitrosation with 18β-glycyrrhetinic acid and nebivolol, respectively, significantly blunts contraction in human myometrial tissue, presenting a novel approach to tocolysis that leverages maladjusted pathways in women who experience sPTL.
Collapse
Affiliation(s)
- Scott D Barnett
- Department of Pharmacology, Myometrial Function Group, University of Nevada, Reno School of Medicine, Reno, NV
| | - Hazik Asif
- Department of Pharmacology, Myometrial Function Group, University of Nevada, Reno School of Medicine, Reno, NV
| | - Mitchell Anderson
- Department of Pharmacology, Myometrial Function Group, University of Nevada, Reno School of Medicine, Reno, NV
| | - Iain L O Buxton
- Department of Pharmacology, Myometrial Function Group, University of Nevada, Reno School of Medicine, Reno, NV
| |
Collapse
|
34
|
Gudicha DW, Romero R, Kabiri D, Hernandez-Andrade E, Pacora P, Erez O, Kusanovic JP, Jung E, Paredes C, Berry SM, Yeo L, Hassan SS, Hsu CD, Tarca AL. Personalized assessment of cervical length improves prediction of spontaneous preterm birth: a standard and a percentile calculator. Am J Obstet Gynecol 2021; 224:288.e1-288.e17. [PMID: 32918893 PMCID: PMC7914140 DOI: 10.1016/j.ajog.2020.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND A sonographic short cervix (length <25 mm during midgestation) is the most powerful predictor of preterm birth. Current clinical practice assumes that the same cervical length cutoff value should apply to all women when screening for spontaneous preterm birth, yet this approach may be suboptimal. OBJECTIVE This study aimed to (1) create a customized cervical length standard that considers relevant maternal characteristics and gestational age at sonographic examination and (2) assess whether the customization of cervical length evaluation improves the prediction of spontaneous preterm birth. STUDY DESIGN This retrospective analysis comprises a cohort of 7826 pregnant women enrolled in a longitudinal protocol between January 2006 and April 2017 at the Detroit Medical Center. Study participants met the following inclusion criteria: singleton pregnancy, ≥1 transvaginal sonographic measurements of the cervix, delivery after 20 weeks of gestation, and available relevant demographics and obstetrical history information. Data from women without a history of preterm birth or cervical surgery who delivered at term without progesterone treatment (N=5188) were used to create a customized standard of cervical length. The prediction of the primary outcome, spontaneous preterm birth at <37 weeks of gestation, was assessed in a subset of pregnancies (N=7336) that excluded cases with induced labor before 37 weeks of gestation. Area under the receiver operating characteristic curve and sensitivity at a fixed false-positive rate were calculated for screening at 20 to 23 6/7, 24 to 27 6/7, 28 to 31 6/7, and 32 to 35 6/7 weeks of gestation in asymptomatic patients. Survival analysis was used to determine which method is better at predicting imminent delivery among symptomatic women. RESULTS The median cervical length remained fundamentally unchanged until 20 weeks of gestation and subsequently decreased nonlinearly with advancing gestational age among women who delivered at term. The effects of parity and maternal weight and height on the cervical length were dependent on the gestational age at ultrasound examination (interaction, P<.05 for all). Parous women had a longer cervix than nulliparous women, and the difference increased with advancing gestation after adjusting for maternal weight and height. Similarly, maternal weight was nonlinearly associated with a longer cervix, and the effect was greater later in gestation. The sensitivity at a 10% false-positive rate for prediction of spontaneous preterm birth at <37 weeks of gestation by a short cervix ranged from 29% to 40% throughout pregnancy, yet it increased to 50%, 50%, 53%, and 54% at 20 to 23 6/7, 24 to 27 6/7, 28 to 31 6/7, and 32 to 35 6/7 weeks of gestation, respectively, for a low, customized percentile (McNemar test, P<.001 for all). When a cervical length <25 mm was compared to the customized screening at 20 to 23 6/7 weeks of gestation by using a customized percentile cutoff value that ensured the same negative likelihood ratio for both screening methods, the customized approach had a significantly higher (about double) positive likelihood ratio in predicting spontaneous preterm birth at <33, <34, <35, <36, and <37 weeks of gestation. Among symptomatic women, the difference in survival between women with a customized cervical length percentile of ≥10th and those with a customized cervical length percentile of <10th was greater than the difference in survival between women with a cervical length ≥25 mm and those with a cervical length <25 mm. CONCLUSION Compared to the use of a cervical length <25 mm, a customized cervical length assessment (1) identifies more women at risk of spontaneous preterm birth and (2) improves the distinction between patients at risk for impending preterm birth in those who have an episode of preterm labor.
Collapse
Affiliation(s)
- Dereje W Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI; Detroit Medical Center, Detroit, MI; Department of Obstetrics and Gynecology, Florida International University, Miami, FL
| | - Doron Kabiri
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Maternity Department "D," Division of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Beer-Sheva, Israel; Faculty of Health Sciences, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Center for Research and Innovation in Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sótero del Río Hospital, Santiago, Chile; Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Carmen Paredes
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Office of Women's Health, Integrative Biosciences Center, Wayne State University, Detroit, MI; Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, and Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Computer Science, Wayne State University College of Engineering, Detroit, MI.
| |
Collapse
|
35
|
Paquette AG, Hood L, Price ND, Sadovsky Y. Deep phenotyping during pregnancy for predictive and preventive medicine. Sci Transl Med 2021; 12:12/527/eaay1059. [PMID: 31969484 DOI: 10.1126/scitranslmed.aay1059] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023]
Abstract
Deep phenotyping during pregnancy offers an opportunity to define the antecedents of lifelong health and wellness, and to improve pregnancy outcomes.
Collapse
Affiliation(s)
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | - Yoel Sadovsky
- Magee-Womens Research Institute and Departments of OBGYN and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
36
|
Integrative analysis of transcriptomic data for identification of T-cell activation-related mRNA signatures indicative of preterm birth. Sci Rep 2021; 11:2392. [PMID: 33504832 PMCID: PMC7841165 DOI: 10.1038/s41598-021-81834-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Preterm birth (PTB), defined as birth at less than 37 weeks of gestation, is a major determinant of neonatal mortality and morbidity. Early diagnosis of PTB risk followed by protective interventions are essential to reduce adverse neonatal outcomes. However, due to the redundant nature of the clinical conditions with other diseases, PTB-associated clinical parameters are poor predictors of PTB. To identify molecular signatures predictive of PTB with high accuracy, we performed mRNA sequencing analysis of PTB patients and full-term birth (FTB) controls in Korean population and identified differentially expressed genes (DEGs) as well as cellular pathways represented by the DEGs between PTB and FTB. By integrating the gene expression profiles of different ethnic groups from previous studies, we identified the core T-cell activation pathway associated with PTB, which was shared among all previous datasets, and selected three representative DEGs (CYLD, TFRC, and RIPK2) from the core pathway as mRNA signatures predictive of PTB. We confirmed the dysregulation of the candidate predictors and the core T-cell activation pathway in an independent cohort. Our results suggest that CYLD, TFRC, and RIPK2 are potentially reliable predictors for PTB.
Collapse
|
37
|
Predominance of Atopobium vaginae at Midtrimester: a Potential Indicator of Preterm Birth Risk in a Nigerian Cohort. mSphere 2021; 6:6/1/e01261-20. [PMID: 33504666 PMCID: PMC7885325 DOI: 10.1128/msphere.01261-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Giving birth too soon accounts for half of all newborn deaths worldwide. Clinical symptoms alone are not sufficient to identify women at risk of giving birth too early, as such a pragmatic approach to reducing the incidence of preterm birth entails developing early strategies for intervention before it materializes. Preterm birth (PTB) is the largest contributor to infant death in sub-Saharan Africa and globally. With a global estimate of 773,600, Nigeria has the third highest rate of PTB worldwide. There have been a number of microbiome profiling studies to identify vaginal microbiomes suggestive of preterm and healthy birth outcome. However, studies on the pregnancy vaginal microbiome in Africa are sparse with none performed in Nigeria. Moreover, few studies have considered the concurrent impact of steroid hormones and the vaginal microbiome on pregnancy outcome. We assessed two key determinants of pregnancy progression to gain a deeper understanding of the interactions between vaginal microbiome composition, steroid hormone concentrations, and pregnancy outcome. Vaginal swabs and blood samples were prospectively collected from healthy midtrimester pregnant women. Vaginal microbiome compositions were assessed by analysis of the V3-V5 region of 16S rRNA genes, and potential functional metabolic traits of identified vaginal microbiomes were imputed by PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states) analysis, while plasma estradiol (E2) and progesterone (P1) levels were quantified by the competitive enzyme-linked immunosorbent assay (ELISA). PTB vaginal samples were characterized by increased microbial richness, high diversity, and depletion of lactobacilli compared to term delivery samples. Women who delivered preterm were characterized by an Atopobium vaginae-dominated vagitype. High relative abundance of Atopobium vaginae at the midtrimester was highly predictive of PTB (area under the receiving operator characteristics [AUROC] of 0.983). There was a marked overlap in the range of plasma E2 and P1 values between term and PTB groups. IMPORTANCE Giving birth too soon accounts for half of all newborn deaths worldwide. Clinical symptoms alone are not sufficient to identify women at risk of giving birth too early, as such a pragmatic approach to reducing the incidence of preterm birth entails developing early strategies for intervention before it materializes. In view of the role played by the vaginal microbiome and maternal steroid hormones in determining obstetric outcome, we assessed the vaginal microbiome composition and steroid hormone during pregnancy and examined their relationship in predicting preterm birth risk in Nigerian women. This study highlights a potential early-driver microbial marker for prediction of preterm birth risk and supports the notion that vaginal microbiome composition varies across populations. A knowledge of relevant preterm birth microbial markers specific to populations would enhance the development of personalized therapeutic interventions toward restoring a microbiome that optimizes reproductive health fitness, therefore reducing the incidence of preterm birth.
Collapse
|
38
|
Maternal DNA Methylation During Pregnancy: a Review. Reprod Sci 2021; 28:2758-2769. [PMID: 33469876 DOI: 10.1007/s43032-020-00456-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022]
Abstract
Multiple environmental, behavioral, and hereditary factors affect pregnancy. Recent studies suggest that epigenetic modifications, such as DNA methylation (DNAm), affect both maternal and fetal health during the period of gestation. Some of the pregnancy-related risk factors can influence maternal DNAm, thus predisposing both the mother and the neonate to clinical adversities with long-lasting consequences. DNAm alterations in the promoter and enhancer regions modulate gene expression changes which play vital physiological role. In this review, we have discussed the recent advances in our understanding of maternal DNA methylation changes during pregnancy and its associated complications such as gestational diabetes and anemia, adverse pregnancy outcomes like preterm birth, and preeclampsia. We have also highlighted some major gaps and limitations in the area which if addressed might improve our understanding of pregnancy and its associated adverse clinical conditions, ultimately leading to healthy pregnancies and reduction of public health burden.
Collapse
|
39
|
Nair J, Maheshwari A. Epigenetics in Necrotizing Enterocolitis. Curr Pediatr Rev 2021; 17:172-184. [PMID: 33882811 DOI: 10.2174/1573396317666210421110608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic alterations in our genetic material can lead to heritable changes in the risk, clinical manifestations, course, and outcomes of many diseases. Understanding these epigenetic mechanisms can help in identifying potential therapeutic targets. This is especially important in necrotizing enterocolitis (NEC), where prenatal as well as postnatal factors impact susceptibility to this devastating condition, but our therapeutic options are limited. Developmental factors affecting intestinal structure and function, our immune system, gut microbiome, and postnatal enteral nutrition are all thought to play a prominent role in this disease. In this manuscript, we have reviewed the epigenetic mechanisms involved in NEC. These include key developmental changes in DNA methylation in the immature intestine, the role of long non-coding RNA (lncRNA) in maintaining intestinal barrier function, epigenetic influences of prenatal inflammation on immunological pathways in NEC pathogenesis such as Toll-Like Receptor 4 (TLR4) and epigenetic changes associated with enteral feeding causing upregulation of pro-inflammatory genes. We have assimilated research findings from our own laboratory with an extensive review of the literature utilizing key terms in multiple databases, including PubMed, EMBASE, and Science Direct.
Collapse
Affiliation(s)
- Jayasree Nair
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University, Baltimore, MA, United States
| |
Collapse
|
40
|
Jehan F, Sazawal S, Baqui AH, Nisar MI, Dhingra U, Khanam R, Ilyas M, Dutta A, Mitra DK, Mehmood U, Deb S, Mahmud A, Hotwani A, Ali SM, Rahman S, Nizar A, Ame SM, Moin MI, Muhammad S, Chauhan A, Begum N, Khan W, Das S, Ahmed S, Hasan T, Khalid J, Rizvi SJR, Juma MH, Chowdhury NH, Kabir F, Aftab F, Quaiyum A, Manu A, Yoshida S, Bahl R, Rahman A, Pervin J, Winston J, Musonda P, Stringer JSA, Litch JA, Ghaemi MS, Moufarrej MN, Contrepois K, Chen S, Stelzer IA, Stanley N, Chang AL, Hammad GB, Wong RJ, Liu C, Quaintance CC, Culos A, Espinosa C, Xenochristou M, Becker M, Fallahzadeh R, Ganio E, Tsai AS, Gaudilliere D, Tsai ES, Han X, Ando K, Tingle M, Marić I, Wise PH, Winn VD, Druzin ML, Gibbs RS, Darmstadt GL, Murray JC, Shaw GM, Stevenson DK, Snyder MP, Quake SR, Angst MS, Gaudilliere B, Aghaeepour N. Multiomics Characterization of Preterm Birth in Low- and Middle-Income Countries. JAMA Netw Open 2020; 3:e2029655. [PMID: 33337494 PMCID: PMC7749442 DOI: 10.1001/jamanetworkopen.2020.29655] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE Worldwide, preterm birth (PTB) is the single largest cause of deaths in the perinatal and neonatal period and is associated with increased morbidity in young children. The cause of PTB is multifactorial, and the development of generalizable biological models may enable early detection and guide therapeutic studies. OBJECTIVE To investigate the ability of transcriptomics and proteomics profiling of plasma and metabolomics analysis of urine to identify early biological measurements associated with PTB. DESIGN, SETTING, AND PARTICIPANTS This diagnostic/prognostic study analyzed plasma and urine samples collected from May 2014 to June 2017 from pregnant women in 5 biorepository cohorts in low- and middle-income countries (LMICs; ie, Matlab, Bangladesh; Lusaka, Zambia; Sylhet, Bangladesh; Karachi, Pakistan; and Pemba, Tanzania). These cohorts were established to study maternal and fetal outcomes and were supported by the Alliance for Maternal and Newborn Health Improvement and the Global Alliance to Prevent Prematurity and Stillbirth biorepositories. Data were analyzed from December 2018 to July 2019. EXPOSURES Blood and urine specimens that were collected early during pregnancy (median sampling time of 13.6 weeks of gestation, according to ultrasonography) were processed, stored, and shipped to the laboratories under uniform protocols. Plasma samples were assayed for targeted measurement of proteins and untargeted cell-free ribonucleic acid profiling; urine samples were assayed for metabolites. MAIN OUTCOMES AND MEASURES The PTB phenotype was defined as the delivery of a live infant before completing 37 weeks of gestation. RESULTS Of the 81 pregnant women included in this study, 39 had PTBs (48.1%) and 42 had term pregnancies (51.9%) (mean [SD] age of 24.8 [5.3] years). Univariate analysis demonstrated functional biological differences across the 5 cohorts. A cohort-adjusted machine learning algorithm was applied to each biological data set, and then a higher-level machine learning modeling combined the results into a final integrative model. The integrated model was more accurate, with an area under the receiver operating characteristic curve (AUROC) of 0.83 (95% CI, 0.72-0.91) compared with the models derived for each independent biological modality (transcriptomics AUROC, 0.73 [95% CI, 0.61-0.83]; metabolomics AUROC, 0.59 [95% CI, 0.47-0.72]; and proteomics AUROC, 0.75 [95% CI, 0.64-0.85]). Primary features associated with PTB included an inflammatory module as well as a metabolomic module measured in urine associated with the glutamine and glutamate metabolism and valine, leucine, and isoleucine biosynthesis pathways. CONCLUSIONS AND RELEVANCE This study found that, in LMICs and high PTB settings, major biological adaptations during term pregnancy follow a generalizable model and the predictive accuracy for PTB was augmented by combining various omics data sets, suggesting that PTB is a condition that manifests within multiple biological systems. These data sets, with machine learning partnerships, may be a key step in developing valuable predictive tests and intervention candidates for preventing PTB.
Collapse
Affiliation(s)
- Fyezah Jehan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sunil Sazawal
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Abdullah H. Baqui
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Usha Dhingra
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Rasheda Khanam
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Muhammad Ilyas
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Arup Dutta
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Dipak K. Mitra
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Saikat Deb
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Public Health Laboratory-Ivo de Carneri, Pemba Island, Zanzibar
| | - Arif Mahmud
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Sayedur Rahman
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Ambreen Nizar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Mamun Ibne Moin
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sajid Muhammad
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Nazma Begum
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sayan Das
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Salahuddin Ahmed
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tarik Hasan
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Javairia Khalid
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Jafar Raza Rizvi
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Nabidul Haque Chowdhury
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Fahad Aftab
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Abdul Quaiyum
- International Center for Maternal and Newborn Health, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Alexander Manu
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Sachiyo Yoshida
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Rajiv Bahl
- Maternal, Newborn, Child and Adolescent Health Research, World Health Organization, Geneva, Switzerland
| | - Anisur Rahman
- Matlab Health Research Centre, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Jennifer Winston
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill
| | - Patrick Musonda
- School of Public Health, University of Zambia, Lusaka, Zambia
| | - Jeffrey S. A. Stringer
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill
| | - James A. Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Seattle, Washington
| | - Mohammad Sajjad Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Ontario, Canada
| | - Mira N. Moufarrej
- Department of Bioengineering, Stanford University, Stanford, California
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ghaith Bany Hammad
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ronald J. Wong
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Candace Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Edward Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Amy S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Dyani Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Eileen S. Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Xiaoyuan Han
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Kazuo Ando
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Martha Tingle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Ivana Marić
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Paul H. Wise
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Maurice L. Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Ronald S. Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Gary L. Darmstadt
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | | | - Gary M. Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - David K. Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, California
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California
- Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
41
|
Schoorlemmer J, Macías-Redondo S, Strunk M, Ramos-Ruíz R, Calvo P, Benito R, Paules C, Oros D. Altered DNA methylation in human placenta after (suspected) preterm labor. Epigenomics 2020; 12:1769-1782. [PMID: 33107765 DOI: 10.2217/epi-2019-0346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The aim of this study was to determine if alterations in DNA methylation in the human placenta would support suspected preterm labor as a pathologic insult associated with diminished placental health. Methods: We evaluated placental DNA methylation at seven loci differentially methylated in placental pathologies using targeted bisulfite sequencing, in placentas associated with preterm labor (term birth after suspected preterm labor [n = 15] and preterm birth [n = 15]), and controls (n = 15). Results: DNA methylation levels at the NCAM1 and PLAGL1 loci in placentas associated with preterm labor did differ significantly (p < 0.05) from controls. Discussion: Specific alterations in methylation patterns indicative of an unfavourable placental environment are associated with preterm labor per se and not restricted to preterm birth.
Collapse
Affiliation(s)
- Jon Schoorlemmer
- Instituto Aragonés de Ciencias de la Salud (IACS) & Aragon Institute for Health Research (IIS Aragón), Zaragoza, Spain.,Placental pathophysiology & fetal programming research group, B05 DGA & GIIS-028 del IISA.,ARAID Foundation, Zaragoza, Spain
| | - Sofía Macías-Redondo
- Instituto Aragonés de Ciencias de la Salud (IACS) & Aragon Institute for Health Research (IIS Aragón), Zaragoza, Spain
| | - Mark Strunk
- Instituto Aragonés de Ciencias de la Salud (IACS), Sequencing & Functional Genomics, Aragon Biomedical Research Center (CIBA), Zaragoza, Spain
| | - Ricardo Ramos-Ruíz
- Unidad de Genómica, Fundación Parque Científico de Madrid, Madrid, Spain
| | - Pilar Calvo
- Placental pathophysiology & fetal programming research group, B05 DGA & GIIS-028 del IISA.,Aragon Institute for Health Research (IIS Aragón), Obstetrics Department, Hospital Clínico Universitario Zaragoza, Spain
| | - Rafael Benito
- Aragon Institute for Health Research (IIS Aragón), Microbiology Department, Hospital Clínico Universitario Zaragoza, Spain
| | - Cristina Paules
- Placental pathophysiology & fetal programming research group, B05 DGA & GIIS-028 del IISA.,Aragon Institute for Health Research (IIS Aragón), Obstetrics Department, Hospital Clínico Universitario Zaragoza, Spain
| | - Daniel Oros
- Placental pathophysiology & fetal programming research group, B05 DGA & GIIS-028 del IISA.,Aragon Institute for Health Research (IIS Aragón), Obstetrics Department, Hospital Clínico Universitario Zaragoza, Spain.,Red de Salud Materno Infantil y del Desarrollo (SAMID), RETICS, Instituto de Salud Carlos III (ISCIII), Subdirección General de Evaluación y Fomento de la Investigación, Fondo Europeo de Desarrollo Regional (FEDER), Spain
| |
Collapse
|
42
|
Rapid Phenotype-Driven Gene Sequencing with the NeoSeq Panel: A Diagnostic Tool for Critically Ill Newborns with Suspected Genetic Disease. J Clin Med 2020; 9:jcm9082362. [PMID: 32718099 PMCID: PMC7464859 DOI: 10.3390/jcm9082362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Abstract
New genomic sequencing techniques have shown considerable promise in the field of neonatology, increasing the diagnostic rate and reducing time to diagnosis. However, several obstacles have hindered the incorporation of this technology into routine clinical practice. We prospectively evaluated the diagnostic rate and diagnostic turnaround time achieved in newborns with suspected genetic diseases using a rapid phenotype-driven gene panel (NeoSeq) containing 1870 genes implicated in congenital malformations and neurological and metabolic disorders of early onset (<2 months of age). Of the 33 newborns recruited, a genomic diagnosis was established for 13 (39.4%) patients (median diagnostic turnaround time, 7.5 days), resulting in clinical management changes in 10 (76.9%) patients. An analysis of 12 previous prospective massive sequencing studies (whole genome (WGS), whole exome (WES), and clinical exome (CES) sequencing) in newborns admitted to neonatal intensive care units (NICUs) with suspected genetic disorders revealed a comparable median diagnostic rate (37.2%), but a higher median diagnostic turnaround time (22.3 days) than that obtained with NeoSeq. Our phenotype-driven gene panel, which is specific for genetic diseases in critically ill newborns is an affordable alternative to WGS and WES that offers comparable diagnostic efficacy, supporting its implementation as a first-tier genetic test in NICUs.
Collapse
|
43
|
Spada E, Calzari L, Corsaro L, Fazia T, Mencarelli M, Di Blasio AM, Bernardinelli L, Zangheri G, Vignali M, Gentilini D. Epigenome Wide Association and Stochastic Epigenetic Mutation Analysis on Cord Blood of Preterm Birth. Int J Mol Sci 2020; 21:ijms21145044. [PMID: 32708910 PMCID: PMC7403978 DOI: 10.3390/ijms21145044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
Preterm birth (PTB) can be defined as the endpoint of a complex process that could be influenced by maternal and environmental factors. Epigenetics recently emerged as an interesting field of investigation since it represents an important mechanism of regulation. This study evaluates epigenetic impact of preterm birth on DNA methylation. Genome-wide DNAm was measured using the Illumina 450K array in cord blood samples obtained from 72 full term and 18 preterm newborns. Lymphocyte composition was calculated based on specific epigenetic markers that are present on the 450k array. Differential methylation analysis was performed both at site and region level; moreover, stochastic epigenetic mutations (SEMs) were also evaluated. The study showed significant differences in blood cell composition between the two groups. Moreover, after multiple testing correction, statistically significant differences in DNA methylation levels emerged between the two groups both at site and region levels. Results obtained were compared to those reported by previous EWAS, leading to a list of more consistent genes associated with PTB. Finally, the SEMs analysis revealed that the burden of SEMs resulted significantly higher in the preterm group. In conclusion, PTB resulted associated to specific epigenetic signatures that involve immune system. Moreover, SEMs analysis revealed an increased epigenetic drift at birth in the preterm group.
Collapse
Affiliation(s)
- Elena Spada
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (E.S.); (L.C.); (T.F.); (L.B.)
| | - Luciano Calzari
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milano, Italy;
| | - Luigi Corsaro
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (E.S.); (L.C.); (T.F.); (L.B.)
| | - Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (E.S.); (L.C.); (T.F.); (L.B.)
| | - Monica Mencarelli
- Molecular Biology Laboratory, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milano, Italy; (M.M.); (A.M.D.B.)
| | - Anna Maria Di Blasio
- Molecular Biology Laboratory, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milano, Italy; (M.M.); (A.M.D.B.)
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (E.S.); (L.C.); (T.F.); (L.B.)
| | - Giulia Zangheri
- Department of Biomedical Science for the Health, University of Milan, Macedonio Melloni Hospital, 20129 Milan, Italy; (G.Z.); (M.V.)
| | - Michele Vignali
- Department of Biomedical Science for the Health, University of Milan, Macedonio Melloni Hospital, 20129 Milan, Italy; (G.Z.); (M.V.)
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (E.S.); (L.C.); (T.F.); (L.B.)
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Cusano Milanino, 20095 Milano, Italy;
- Correspondence: ; Tel.: +39-0382987541
| |
Collapse
|
44
|
Conde-Agudelo A, Romero R, Nicolaides KH. Cervical pessary to prevent preterm birth in asymptomatic high-risk women: a systematic review and meta-analysis. Am J Obstet Gynecol 2020; 223:42-65.e2. [PMID: 32027880 DOI: 10.1016/j.ajog.2019.12.266] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Randomized controlled trials that have assessed the efficacy of cervical pessary to prevent preterm birth in asymptomatic high-risk women have reported conflicting results. OBJECTIVE To evaluate the efficacy and safety of cervical pessary to prevent preterm birth and adverse perinatal outcomes in asymptomatic high-risk women. DATA SOURCES MEDLINE, EMBASE, POPLINE, CINAHL, and LILACS (from their inception to October 31, 2019), Cochrane databases, Google Scholar, bibliographies, and conference proceedings. STUDY ELIGIBILITY CRITERIA Randomized controlled trials that compared cervical pessary with standard care (no pessary) or alternative interventions in asymptomatic women at high risk for preterm birth. STUDY APPRAISAL AND SYNTHESIS METHODS The systematic review was conducted according to the Cochrane Handbook guidelines. The primary outcome was spontaneous preterm birth <34 weeks of gestation. Secondary outcomes included adverse pregnancy, maternal, and perinatal outcomes. Pooled relative risks with 95% confidence intervals were calculated. Quality of evidence was assessed using the GRADE methodology. RESULTS Twelve studies (4687 women and 7167 fetuses/infants) met the inclusion criteria: 8 evaluated pessary vs no pessary in women with a short cervix, 2 assessed pessary vs no pessary in unselected multiple gestations, and 2 compared pessary vs vaginal progesterone in women with a short cervix. There were no significant differences between the pessary and no pessary groups in the risk of spontaneous preterm birth <34 weeks of gestation among singleton gestations with a cervical length ≤25 mm (relative risk, 0.80; 95% confidence interval, 0.43-1.49; 6 trials, 1982 women; low-quality evidence), unselected twin gestations (relative risk, 1.05; 95% confidence interval, 0.79-1.41; 1 trial, 1177 women; moderate-quality evidence), twin gestations with a cervical length <38 mm (relative risk, 0.75; 95% confidence interval, 0.41-1.36; 3 trials, 1128 women; low-quality evidence), and twin gestations with a cervical length ≤25 mm (relative risk; 0.72, 95% confidence interval, 0.25-2.06; 2 trials, 348 women; low-quality evidence). Overall, no significant differences were observed between the pessary and no pessary groups in preterm birth <37, <32, and <28 weeks of gestation, and most adverse pregnancy, maternal, and perinatal outcomes (low- to moderate-quality evidence for most outcomes). There were no significant differences in the risk of spontaneous preterm birth <34 weeks of gestation between pessary and vaginal progesterone in singleton gestations with a cervical length ≤25 mm (relative risk, 0.99; 95% confidence interval, 0.54-1.83; 1 trial, 246 women; low-quality evidence) and twin gestations with a cervical length <38 mm (relative risk, 0.73; 95% confidence interval, 0.46-1.18; 1 trial, 297 women; very low-quality evidence). Vaginal discharge was significantly more frequent in the pessary group than in the no pessary and vaginal progesterone groups (relative risks, ∼2.20; high-quality evidence). CONCLUSION Current evidence does not support the use of cervical pessary to prevent preterm birth or to improve perinatal outcomes in singleton or twin gestations with a short cervix and in unselected twin gestations.
Collapse
Affiliation(s)
- Agustin Conde-Agudelo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services, Bethesda, MD and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI; Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI; Detroit Medical Center, Detroit, MI; Department of Obstetrics and Gynecology, Florida International University, Miami, FL.
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| |
Collapse
|
45
|
York TP, Latendresse SJ, Jackson-Cook C, Lapato DM, Moyer S, Wolen AR, Roberson-Nay R, Do EK, Murphy SK, Hoyo C, Fuemmeler BF, Strauss JF. Replicated umbilical cord blood DNA methylation loci associated with gestational age at birth. Epigenetics 2020; 15:1243-1258. [PMID: 32448018 DOI: 10.1080/15592294.2020.1767277] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
DNA methylation is highly sensitive to in utero perturbations and has an established role in both embryonic development and regulation of gene expression. The foetal genetic component has been previously shown to contribute significantly to the timing of birth, yet little is known about the identity and behaviour of individual genes. The aim of this study was to test the extent genome-wide DNA methylation levels in umbilical cord blood were associated with gestational age at birth (GA). Findings were validated in an independent sample and evidence for the regulation of gene expression was evaluated for cis gene relationships in specimens with multi-omic data. Genome-wide DNA methylation, measured by the Illumina Infinium Human Methylation 450 K BeadChip, was associated with GA for 2,372 CpG probes (5% FDR) in both the Pregnancy, Race, Environment, Genes (PREG) and Newborn Epigenetic Study (NEST) cohorts. Significant probes mapped to 1,640 characterized genes and an association with nearby gene expression measures obtained by the Affymetrix HG-133A microarray was found for 11 genes. Differentially methylated positions were enriched for actively transcribed and enhancer chromatin states, were predominately located outside of CpG islands, and mapped to genes enriched for inflammation and innate immunity ontologies. In both PREG and NEST, the first principal component derived from these probes explained approximately one-half (58.1% and 47.8%, respectively) of the variation in GA. Gene pathways identified are consistent with the hypothesis of pathogen detection and response by the immune system to elicit premature labour as a consequence of unscheduled inflammation.
Collapse
Affiliation(s)
- Timothy P York
- Department of Human and Molecular Genetics, Virginia Commonwealth University , Richmond, VA, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University , Richmond, VA, USA
| | - Shawn J Latendresse
- Department of Psychology and Neuroscience, Baylor University , Waco, TX, USA
| | - Colleen Jackson-Cook
- Department of Human and Molecular Genetics, Virginia Commonwealth University , Richmond, VA, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University , Richmond, VA, USA.,Department of Pathology, Virginia Commonwealth University , Richmond, VA, USA
| | - Dana M Lapato
- Department of Human and Molecular Genetics, Virginia Commonwealth University , Richmond, VA, USA
| | - Sara Moyer
- Department of Human and Molecular Genetics, Virginia Commonwealth University , Richmond, VA, USA
| | - Aaron R Wolen
- Transplant Research Institute, Department of Surgery, University of Tennessee Health Science Center , Memphis, TN, USA
| | - Roxann Roberson-Nay
- Department of Psychiatry, Virginia Commonwealth University , Richmond, VA, USA
| | - Elizabeth K Do
- Department of Health Behavior and Policy, Virginia Commonwealth University , Richmond, VA, USA
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University , Durham, North Carolina, USA
| | - Catherine Hoyo
- Epidemiology and Environmental Epigenomics Laboratory, Center for Human Health and the Environment, North Carolina State University , Raleigh, NC, USA
| | - Bernard F Fuemmeler
- Department of Health Behavior and Policy, Virginia Commonwealth University , Richmond, VA, USA
| | - Jerome F Strauss
- Department of Human and Molecular Genetics, Virginia Commonwealth University , Richmond, VA, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University , Richmond, VA, USA
| |
Collapse
|
46
|
Wong HS, Wadon M, Evans A, Kirov G, Modi N, O'Donovan MC, Thapar A. Contribution of de novo and inherited rare CNVs to very preterm birth. J Med Genet 2020; 57:552-557. [PMID: 32051258 DOI: 10.1136/jmedgenet-2019-106619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/23/2019] [Accepted: 01/12/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND The genomic contribution to adverse health sequelae in babies born very preterm (<32 weeks' gestation) is unknown. We conducted an investigation of rare CNVs in infants born very preterm as part of a study to determine the feasibility and acceptability of a larger, well-powered genome-wide investigation in the UK, with follow-up using linked National Health Service records and DNA storage for additional research. METHODS We studied 488 parent-offspring trios. We performed genotyping using Illumina Infinium OmniExpress Arrays. CNV calling and quality control (QC) were undertaken using published protocols. We examined de novo CNVs in infants and the rate of known pathogenic variants in infants, mothers and fathers and compared these with published comparator data. We defined rare pathogenic CNVs as those consistently reported to be associated with clinical phenotypes. RESULTS We identified 14 de novo CNVs, representing a mutation rate of 2.9%, compared with 2.1% reported in control populations. The median size of these CNV was much higher than in comparator data (717 kb vs 255 kb). The rate of pathogenic CNVs was 4.3% in infants, 2.7% in mothers and 2% in fathers, compared with 2.3% in UK Biobank participants. CONCLUSION Our findings suggest that the rate of de novo CNVs, especially rare pathogenic CNVs, could be elevated in those born very preterm. However, we will need to conduct a much larger study to corroborate this conclusion.
Collapse
Affiliation(s)
- Hilary S Wong
- Department of Paediatrics, Cambridge University, Cambridge, UK
| | - Megan Wadon
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Alexandra Evans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - George Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Neena Modi
- Section of Neonatal Medicine, Imperial College London, London, UK
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Anita Thapar
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| |
Collapse
|
47
|
Park B, Khanam R, Vinayachandran V, Baqui AH, London SJ, Biswal S. Epigenetic biomarkers and preterm birth. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa005. [PMID: 32551139 PMCID: PMC7293830 DOI: 10.1093/eep/dvaa005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 05/06/2023]
Abstract
Preterm birth (PTB) is a major public health challenge, and novel, sensitive approaches to predict PTB are still evolving. Epigenomic markers are being explored as biomarkers of PTB because of their molecular stability compared to gene expression. This approach is also relatively new compared to gene-based diagnostics, which relies on mutations or single nucleotide polymorphisms. The fundamental principle of epigenome diagnostics is that epigenetic reprogramming in the target tissue (e.g. placental tissue) might be captured by more accessible surrogate tissue (e.g. blood) using biochemical epigenome assays on circulating DNA that incorporate methylation, histone modifications, nucleosome positioning, and/or chromatin accessibility. Epigenomic-based biomarkers may hold great potential for early identification of the majority of PTBs that are not associated with genetic variants or mutations. In this review, we discuss recent advances made in the development of epigenome assays focusing on its potential exploration for association and prediction of PTB. We also summarize population-level cohort studies conducted in the USA and globally that provide opportunities for genetic and epigenetic marker development for PTB. In addition, we summarize publicly available epigenome resources and published PTB studies. We particularly focus on ongoing genome-wide DNA methylation and epigenome-wide association studies. Finally, we review the limitations of current research, the importance of establishing a comprehensive biobank, and possible directions for future studies in identifying effective epigenome biomarkers to enhance health outcomes for pregnant women at risk of PTB and their infants.
Collapse
Affiliation(s)
- Bongsoo Park
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Vinesh Vinayachandran
- School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Abdullah H Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Stephanie J London
- Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
48
|
The promise and pitfalls of precision medicine to resolve black-white racial disparities in preterm birth. Pediatr Res 2020; 87:221-226. [PMID: 31382269 DOI: 10.1038/s41390-019-0528-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/28/2022]
Abstract
Differences in preterm birth rates between black and white women are the largest contributor to racial disparities in infant mortality. In today's age of precision medicine, analysis of the genome, epigenome, metabolome, and microbiome has generated interest in determining whether these biomarkers can help explain racial disparities. We propose that there are pitfalls as well as opportunities when using precision medicine analyses to interrogate disparities in health. To conclude that racial disparities in complex conditions are genetic in origin ignores robust evidence that social and environmental factors that track with race are major contributors to disparities. Biomarkers measured in omic assays that may be more environmentally responsive than genomics, such as the epigenome or metabolome, may be on the causal pathway of race and preterm birth, but omic observational studies suffer from the same limitations as traditional cohort studies. Confounding can lead to false conclusions about the causal relationship between omics and preterm birth. Methodological strategies (including stratification and causal mediation analyses) may help to ensure that associations between biomarkers and exposures, as well as between biomarkers and outcomes, are valid signals. These epidemiologic strategies present opportunities to assess whether precision medicine biomarkers can uncover biology underlying perinatal health disparities.
Collapse
|
49
|
Stress During Pregnancy and Epigenetic Modifications to Offspring DNA: A Systematic Review of Associations and Implications for Preterm Birth. J Perinat Neonatal Nurs 2020; 34:134-145. [PMID: 32332443 PMCID: PMC7185032 DOI: 10.1097/jpn.0000000000000471] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Offspring born preterm (ie, before 37 weeks of gestation) are more likely to die or experience long-standing illness than full-term offspring. Maternal genetic variants (ie, heritable, stable variations in the genetic code) and epigenetic modifications (ie, chemical modifications to the genetic code that can affect which genes are turned on or off) in response to stress have been implicated in preterm birth. Fetal genetic variants have been linked to preterm birth though the role of offspring epigenetics in preterm birth remains understudied. This systematic review synthesizes the literature examining associations among stress during pregnancy and epigenetic modifications to offspring DNA, with 25 reports identified. Ten reports examined DNA methylation (ie, addition/removal of methyl groups to/from DNA) across the epigenome. The remainder examined DNA methylation near genes of interest, primarily genes linked to hypothalamic-pituitary-adrenal axis function (NR3C1, FKBP51), growth/immune function (IGF2), and socioemotional regulation (SLC6A4, OXTR). The majority of reports noted associations among stress and offspring DNA methylation, primarily when perceived stress, anxiety, or depression served as the predictor. Findings suggest that differences in offspring epigenetic patterns may play a role in stress-associated preterm birth and serve as targets for novel interventions.
Collapse
|
50
|
Pique-Regi R, Romero R, Tarca AL, Sendler ED, Xu Y, Garcia-Flores V, Leng Y, Luca F, Hassan SS, Gomez-Lopez N. Single cell transcriptional signatures of the human placenta in term and preterm parturition. eLife 2019; 8:52004. [PMID: 31829938 PMCID: PMC6949028 DOI: 10.7554/elife.52004] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/12/2019] [Indexed: 01/02/2023] Open
Abstract
More than 135 million births occur each year; yet, the molecular underpinnings of human parturition in gestational tissues, and in particular the placenta, are still poorly understood. The placenta is a complex heterogeneous organ including cells of both maternal and fetal origin, and insults that disrupt the maternal-fetal dialogue could result in adverse pregnancy outcomes such as preterm birth. There is limited knowledge of the cell type composition and transcriptional activity of the placenta and its compartments during physiologic and pathologic parturition. To fill this knowledge gap, we used scRNA-seq to profile the placental villous tree, basal plate, and chorioamniotic membranes of women with or without labor at term and those with preterm labor. Significant differences in cell type composition and transcriptional profiles were found among placental compartments and across study groups. For the first time, two cell types were identified: 1) lymphatic endothelial decidual cells in the chorioamniotic membranes, and 2) non-proliferative interstitial cytotrophoblasts in the placental villi. Maternal macrophages from the chorioamniotic membranes displayed the largest differences in gene expression (e.g. NFKB1) in both processes of labor; yet, specific gene expression changes were also detected in preterm labor. Importantly, several placental scRNA-seq transcriptional signatures were modulated with advancing gestation in the maternal circulation, and specific immune cell type signatures were increased with labor at term (NK-cell and activated T-cell signatures) and with preterm labor (macrophage, monocyte, and activated T-cell signatures). Herein, we provide a catalogue of cell types and transcriptional profiles in the human placenta, shedding light on the molecular underpinnings and non-invasive prediction of the physiologic and pathologic parturition.
Collapse
Affiliation(s)
- Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Roberto Romero
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, United States.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, United States.,Detroit Medical Center, Detroit, United States
| | - Adi L Tarca
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Computer Science, College of Engineering, Wayne State University, Detroit, United States
| | - Edward D Sendler
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States
| | - Yi Xu
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Valeria Garcia-Flores
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Yaozhu Leng
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, United States.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Department of Physiology, Wayne State University School of Medicine, Detroit, United States
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, United States.,Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, United States.,Department of Immunology, Microbiology, and Biochemistry, Wayne State University School of Medicine, Detroit, United States
| |
Collapse
|