1
|
Mladenić T, Barišić A, Pereza N, Ostojić S, Peterlin B, Dević Pavlić S. Maternal genetic risk factors for spontaneous preterm birth: A systematic review and meta-analysis. Int J Gynaecol Obstet 2025; 169:458-473. [PMID: 39620886 DOI: 10.1002/ijgo.16056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 04/22/2025]
Abstract
BACKGROUND Despite various genomic approaches used in prior studies investigating the association of maternal genetic variability with spontaneous preterm birth (sPTB), results show inconsistency and contradictions. OBJECTIVES To conduct a systematic review of studies analyzing the association between maternal genetic variants and sPTB, evaluate retrieved studies based on selection criteria, classify studies into hypothesis-based and hypothesis-free, and perform a meta-analysis to identify the strongest associations. SEARCH STRATEGY PubMed, Scopus, and reference lists were searched until October 2024. SELECTION CRITERIA English-language, case-control, cross-sectional, and prospective cohort studies examining the association between maternal genetic variations and sPTB were included. DATA COLLECTION AND ANALYSIS Data on authors, publication year, ethnicity, genes/variants, P values, study type, sample size, inclusion criteria, and methods were collected. The association strength was estimated using odds ratios with 95% confidence intervals. RESULTS Eighty-one studies met eligibility criteria: 73 utilized a hypothesis-based and 14 a hypothesis-free approach. Thirty-five studies qualified for a meta-analysis, revealing a significant association in tumor necrosis factor α (rs1800629) gene for alleles and additive and recessive genetic models (P ≤ 0.05). From the hypothesis-free approach, 13 genes reached global significance in association with sPTB (P < 5 × 10-8). CONCLUSIONS No single gene or variant was consistently associated with sPTB risk among studies. Hypothesis-based analyses highlighted tumor necrosis factor α (rs1800629) as a modest signal, while hypothesis-free approaches identified 13 genes with genome-wide significance, pointing to new research directions in understanding sPTB genetics.
Collapse
Affiliation(s)
- Tea Mladenić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Anita Barišić
- Department of Gynecology and Obstetrics, University Hospital Rijeka, Rijeka, Croatia
| | - Nina Pereza
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Saša Ostojić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Sanja Dević Pavlić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
2
|
Huri M, Strambi N, Finazzi M, Manciucca G, Catalano G, Seravalli V, Di Tommaso M. The role of family history of preterm delivery in the individual risk of spontaneous preterm delivery: a case-control study. Arch Gynecol Obstet 2024; 309:2515-2519. [PMID: 37466687 PMCID: PMC11147892 DOI: 10.1007/s00404-023-07144-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE To investigate the role of family history of preterm delivery (PTD) in the individual risk of spontaneous preterm delivery. METHODS A retrospective case-control study was conducted on 354 patients who delivered between 2018 and 2020. 177 women who delivered preterm were matched with 177 controls who had full-term delivery. A questionnaire was administered to investigate the family history of PTD of both the patient and her partner. Cases and controls were matched for the anamnestic risk factors for PTD. RESULTS Seventeen of 173 women (9.8%) in the PTD group reported being born preterm, compared to five of 169 women (2.9%) in the control group (p = 0.01), with an odds ratio (OR) of 3.57 (95% confidence interval, CI 1.29-9.92). Women who delivered preterm also reported more frequently having a sibling who was born preterm (12.4% vs. 4.2%, p = 0.01), with an OR of 3.18 (95% CI 1.31-7.7). No association was found between the partner's family history of premature delivery and the patient's risk of preterm delivery in the present pregnancy. CONCLUSIONS Pregnant patients who were born prematurely or who have siblings born preterm have an increased risk of preterm delivery in their own pregnancies. Assessment of female personal and family history of PTD should be used to identify women at risk of having a PTD in the present pregnancy.
Collapse
Affiliation(s)
- Mor Huri
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Noemi Strambi
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Marta Finazzi
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Giulia Manciucca
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Giovanna Catalano
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Viola Seravalli
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy.
| | - Mariarosaria Di Tommaso
- Obstetrics and Gynecology Unit, Division of Obstetrics and Gynecology, Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
3
|
Kadivnik M, Plečko D, Kralik K, Arvaj N, Wagner J. Role of IL-6, IL-10 and TNFα Gene Variants in Preterm Birth. J Clin Med 2024; 13:2429. [PMID: 38673702 PMCID: PMC11051338 DOI: 10.3390/jcm13082429] [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: 03/19/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Background: The association of gene variants for interleukin 6 (IL-6) (rs1800796), interleukin 10 (IL-10) (rs1800896) and tumor necrosis factorα (TNFα (rs1800629) with the occurrence of spontaneous preterm birth (PTB) was investigated to determine whether these genetic variants are a risk factor. Methods: A total of 199 blood samples from pregnant women who had given birth prematurely and 200 control blood samples were analyzed to determine single nucleotide polymorphisms (SNPs) of genes for IL-6 (rs1800796), IL-10 (rs1800896) and TNFα (rs1800629). The control samples were samples from pregnant women with term delivery. The isolation of DNA was performed on mini-spin columns according to the manufacturer's protocol. The quality and purity of the isolated DNA were tested using a Qubit 3 fluorometer. Genotyping was performed with an ABI PRISM 7500 SDS using TaqMan SNP genotyping assays. The genotypes obtained were analyzed using the 7500 Software v2.3 package. Results: Carriers of the A/A genotype for the rs1800629 SNP of the TNFα gene have a 4.81 times greater chance of late-onset PTB compared to carriers of the G/G and A/G genotypes in the recessive inheritance model. The presence of the G/G genotype in the recessive inheritance model compared with the G/A and A/A genotypes for the rs1800896 SNP of the IL-10 gene represents a potentially protective factor, with mothers in the term-birth group having an almost 2-fold lower odds of PTB in general and an almost 10-fold lower odds of early PTB. On the other hand, carriers of the A/G genotype of rs1800896 have a 1.54-fold higher chance of preterm birth in general and a 1.6-fold higher chance of late preterm birth in the superdominant inheritance model compared to the A/A and G/G genotypes in the group of mothers with PTB. In this study, no association was found between PTB and the rs1800796 SNP of the IL-6 gene. Conclusions: rs1800629 in mothers was associated with PTB. rs1800896 shows a potentially protective effect for the occurrence of PTB in this study. No association was found between PTB and rs1800796.
Collapse
Affiliation(s)
- Mirta Kadivnik
- Clinic of Obstetrics and Gynecology, University Hospital Center Osijek, J. Huttlera 4, 31000 Osijek, Croatia;
- Department of Obstetrics and Gynecology, Faculty of Medicine, J.J. Strossmayer University, J. Huttlera 4, 31000 Osijek, Croatia
| | - Deni Plečko
- Department of Medical Biology and Genetics, Faculty of Medicine, J.J. Strossmayer University, J. Huttlera 4, 31000 Osijek, Croatia; (D.P.)
| | - Kristina Kralik
- Department of Medical Statistics and Informatics, Faculty of Medicine, J.J. Strossmayer University, J. Huttlera 4, 31000 Osijek, Croatia
| | - Nena Arvaj
- Department of Medical Biology and Genetics, Faculty of Medicine, J.J. Strossmayer University, J. Huttlera 4, 31000 Osijek, Croatia; (D.P.)
| | - Jasenka Wagner
- Department of Medical Biology and Genetics, Faculty of Medicine, J.J. Strossmayer University, J. Huttlera 4, 31000 Osijek, Croatia; (D.P.)
| |
Collapse
|
4
|
Hughes ZH, Hughes LM, Khan SS. Genetic contributions to risk of adverse pregnancy outcomes. CURRENT CARDIOVASCULAR RISK REPORTS 2023; 17:185-193. [PMID: 38186860 PMCID: PMC10768680 DOI: 10.1007/s12170-023-00729-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 01/09/2024]
Abstract
Purpose of Review Adverse pregnancy outcomes (APOs), including hypertensive disorders of pregnancy (HDP), low birthweight (LBW), and preterm birth (PTB), along with peripartum cardiomyopathy (PPCM) are associated with short- and long-term maternal and fetal cardiovascular risks. This review focuses on the genetic contributions to the risk of APOs and PPCM. Recent Findings The expansion of genome-wide association studies (GWAS) has led to better understanding of the biologic mechanisms underpinning APO, PPCM, and the predisposition to cardiovascular disease across the life course. Genetic loci known to be involved with the risk of hypertension (FTO, ZNF831) have been associated with the development of overall HDP and preeclampsia. Additionally, four loci significantly associated with type 2 diabetes have been associated with GDM (CDKAL1, MTNR1B, TCF7L2, CDK2NA-CDKN2B). Variants in loci known to affect genes coding for proteins involved in immune cell function and placental health (EBF1, EEFSEC, AGTR2, 2q13) have been implicated in the development of PTB and future cardiovascular risks for both the mother and the offspring. Genetic similarities in rare variants between PPCM and dilated cardiomyopathy have been described suggesting shared pathophysiologic origins as well as predisposition for future risk of heart failure, highlighting the need for the development PPCM genetic counseling guidelines. Summary Genetics may inform mechanisms, risk, and counseling for individuals after an APO or PPCM. Through recent advances in genetic techniques and analytic approaches, new insights into the underlying biologic mechanisms and genetic variants leading to these risks have been discovered.
Collapse
Affiliation(s)
- Zachary H. Hughes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, UA
| | - Lydia M. Hughes
- Department of Obstetrics & Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, UA
| | - Sadiya S. Khan
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Huang G, Yao D, Yan X, Zheng M, Yan P, Chen X, Wang D. Emerging role of toll-like receptors signaling and its regulators in preterm birth: a narrative review. Arch Gynecol Obstet 2023; 308:319-339. [PMID: 35916961 DOI: 10.1007/s00404-022-06701-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/03/2022] [Indexed: 11/02/2022]
Abstract
INTRODUCTION Despite intensive research, preterm birth (PTB) rates have not decreased significantly in recent years due to a lack of understanding of the underlying causes and insufficient treatment options for PTB. We are committed to finding promising biomarkers for the treatment of PTB. METHODS An extensive search of the literature was conducted with MEDLINE/PubMed, and in total, 151 studies were included and summarized in the present review. RESULTS Substantial evidence supports that the infection and/or inflammatory cascade associated with infection is an early event in PTB. Toll-like receptor (TLR) is a prominent pattern recognition receptor (PRR) found on both immune and non-immune cells, including fetal membrane cells. The activation of TLR downstream molecules, followed by TLR binding to its ligand, is critical for infection and inflammation, leading to the involvement of the TLR signaling pathway in PTB. TLR ligands are derived from microbial components and molecules released by damaged and dead cells. Particularly, TLR4 is an essential TLR because of its ability to recognize lipopolysaccharide (LPS). In this comprehensive overview, we discuss the role of TLR signaling in PTB, focus on numerous host-derived genetic and epigenetic regulators of the TLR signaling pathway, and cover ongoing research and prospective therapeutic options for treating PTB by inhibiting TLR signaling. CONCLUSION This is a critical topic because TLR-related molecules and mechanisms may enable obstetricians to better understand the physiological changes in PTB and develop new treatment and prevention strategies.
Collapse
Affiliation(s)
- Ge Huang
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Yao
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoli Yan
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mingyu Zheng
- Department of Pharmacy, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ping Yan
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoxia Chen
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Wang
- Department of Gynecology and Obstetrics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
6
|
Chang R, Dai J, Wang L, Liu H, Jiang H, Liu X, Jiang L, He F, Hu L. PlGF/FLT-1 deficiency leads to reduced STAT3-C/EBPβ signaling and aberrant polarization in decidual macrophages during early spontaneous abortion. Front Immunol 2023; 14:1061949. [PMID: 37033974 PMCID: PMC10074254 DOI: 10.3389/fimmu.2023.1061949] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
IntroductionDysregulated macrophage polarization (excessive M1-like or limited M2-like macrophages) in the early decidua contributes to allogeneic fetal rejection and thus early spontaneous abortion. However, the modulators of M1/M2 balance at the early maternal-fetal interface remain mostly unknown.MethodsFirst-trimester decidual tissues were collected from normal pregnant women undergoing elective pregnancy terminations and patients with spontaneous abortion. We measured the expression of placental growth factor (PlGF) and Fms-like-tyrosine-kinase receptor 1 (FLT-1), and characterized the profiles of macrophages in decidua. Notably, we investigated the effect of recombinant human PlGF (rhPlGF) on decidual macrophages (dMös) from normal pregnancy and revealed the underlying mechanisms both in vitro and in vivo.ResultsThe downregulated expression of PlGF/ FLT-1 may result in spontaneous abortion by inducing the M1-like deviation of macrophages in human early decidua. Moreover, the CBA/J×DBA/2 abortion-prone mice displayed a lower FLT-1 expression in uterine macrophages than did CBA/J×BALB/c control pregnant mice. In in vitro models, rhPlGF treatment was found to drive the M2-like polarization of dMös via the STAT3/CEBPB signaling pathway. These findings were further supported by a higher embryo resorption rate and uterine macrophage dysfunction in Pgf knockout mice, in addition to the reduced STAT3 transcription and C/EBPâ expression in uterine macrophages.DiscussionPlGF plays a key role in early pregnancy maintenance by skewing dMös toward an M2-like phenotype via the FLT-1-STAT3-C/EBP â signaling pathway. Excitingly, our results highlight a rationale that PlGF is a promising target to prevent early spontaneous abortion.
Collapse
Affiliation(s)
- Ruiqi Chang
- The Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Lab for Reproduction and Development, Ministry of Education, Chongqing, China
- Reproduction and Stem Cell Therapy Research Center of Chongqing, Chongqing Medical University, Chongqing, China
| | - Jingcong Dai
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Liu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huanhuan Jiang
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Liu
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Linying Jiang
- Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan He
- The Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Lab for Reproduction and Development, Ministry of Education, Chongqing, China
- Reproduction and Stem Cell Therapy Research Center of Chongqing, Chongqing Medical University, Chongqing, China
- *Correspondence: Fan He, ; Lina Hu,
| | - Lina Hu
- The Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Joint International Research Lab for Reproduction and Development, Ministry of Education, Chongqing, China
- Reproduction and Stem Cell Therapy Research Center of Chongqing, Chongqing Medical University, Chongqing, China
- *Correspondence: Fan He, ; Lina Hu,
| |
Collapse
|
7
|
Richardson LS, Emezienna N, Burd I, Taylor BD, Peltier MR, Han A, Menon R. Adapting an organ-on-chip device to study the effect of fetal sex and maternal race/ethnicity on preterm birth related intraamniotic inflammation leading to fetal neuroinflammation. Am J Reprod Immunol 2022; 88:e13638. [PMID: 36308737 PMCID: PMC9712252 DOI: 10.1111/aji.13638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/23/2022] [Accepted: 10/11/2022] [Indexed: 01/05/2023] Open
Abstract
PROBLEM Fetal neuroinflammation has been linked to preterm birth-related intraamniotic infection and inflammation; However, the contribution of fetal sex and maternal race/ethnicity is unknown. To determine if fetal sex and maternal race/ethnicity influence neuroinflammation, an organ-on-chip (OOC) model were established under normal or pathologic conditions utilizing amniotic fluid. METHOD OF STUDY OOC is composed of two-cell culture chambers connected by Type IV collagen-coated microchannels. Human fetal astroglia (SVGp12) and microglia (HMC3) were co-cultured at an 80:20 ratio in the inner chamber. The outer chamber contained amniotic fluid (AF) from male and female fetuses of White Hispanic (WH) and African-American (AA) pregnant women with or without lipopolysaccharide (LPS-100 ng/ml) and incubated for 48 h. Glial migration (brightfield microscopy), activation (Immunocytochemistry), and cytokine production (Luminex assays) were quantified and compared (N = 4 for each category of sex and race/ethnicity). RESULTS In a pooled analysis, AF+LPS did not induce glial activation or inflammatory changes compared to AF alone. When stratified by sex, male AF+LPS promoted significant glial activation (high CD11b:p < 0.05; low Iba1:p < 0.01) compared to male AF without LPS; however, this was not associated with changes in pro-inflammatory cytokines. When stratified by race/ethnicity, AF+LPS induced glial activation in both groups, but a differential increase in pro-inflammatory cytokines was seen between WH and AA AF (WH-interleukin-1β: p < 0.05; AA-interleukin-8: p < 0.01). CONCLUSION This OOC model of fetal neuroinflammation has determined that race/ethnicity differences do exist for perinatal brain injury. The fetal sex of neonates was not a determining factor of susceptibility to intraamniotic inflammation leading to neuroinflammation.
Collapse
Affiliation(s)
- Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Nkechinyere Emezienna
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Johns Hopkin University, Baltimore, Maryland, USA
| | - Brandie D. Taylor
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Morgan R. Peltier
- Department of Psychiatry and Behavioral Health, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
- Department of Psychiatry and Behavioral Health, Jersey Shore University Medical Center, Neptune, New Jersey, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
8
|
Lee SM, Park KH, Joo E, Jeong DE, Lee JE, Lee KN, Shin S. High-throughput analysis of amniotic fluid proteins associated with histological chorioamnionitis in preterm premature rupture of membranes using an antibody-based microarray. Am J Reprod Immunol 2022; 88:e13595. [PMID: 35792516 DOI: 10.1111/aji.13595] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/30/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022] Open
Abstract
PROBLEM To identify potential proteins in the amniotic fluid (AF) that may be associated with histologic chorioamnionitis (HCA) in patients with preterm premature rupture of membranes (PPROM) using antibody-based microarray analysis. METHOD OF STUDY This was a retrospective cohort study involving 100 singleton pregnant women with PPROM at 24-34 weeks who underwent amniocentesis and delivered within 120 h of amniocentesis. First, the AF proteomes of 15 patients with PPROM and HCA were compared with those of 15 gestational age-matched patients without HCA using a protein microarray. Next, 12 candidate proteins associated with HCA were further validated in 100 consecutive patients with PPROM by ELISA. RESULTS Of 507 proteins assessed in the microarray analysis, 46 showed significant intergroup differences. Further quantification confirmed that the levels of EN-RAGE, IL-6, MMP-9, TNFR2, SPARC, TSP2, and uPA were higher in the AF of PPROM patients with HCA than in those without. Multivariate analyses also showed that elevated AF EN-RAGE, IL-6, MMP-9, and TNFR2 levels were independently associated with HCA when adjusted for baseline variables. The frequency of the highest quartile of the aforementioned proteins significantly increased as the total grade of HCA increased; the risk of HCA significantly increased with increasing AF levels of each protein (P for trend < .001). CONCLUSIONS Using protein-antibody microarray technology, we discovered several potential AF proteins (EN-RAGE, IL-6, MMP-9, and TNFR2) independently associated with HCA in patients with PPROM. Furthermore, we demonstrated a direct correlation between the gradation of the intra-amniotic inflammatory response and HCA severity.
Collapse
Affiliation(s)
- Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Eunwook Joo
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Da Eun Jeong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ji Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sue Shin
- Department of Laboratory Medicine, Seoul National University Boramae Hospital, Seoul, South Korea
| |
Collapse
|
9
|
Gupta JK, Alfirevic A. Systematic review of preterm birth multi-omic biomarker studies. Expert Rev Mol Med 2022; 24:1-24. [PMID: 35379367 PMCID: PMC9884789 DOI: 10.1017/erm.2022.13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 02/16/2022] [Accepted: 03/30/2022] [Indexed: 11/07/2022]
Abstract
Preterm birth (PTB) is one of the leading causes of deaths in infants under the age of five. Known risk factors of PTB include genetic factors, lifestyle choices or infection. Identification of omic biomarkers associated with PTB could aid clinical management of women at high risk of early labour and thereby reduce neonatal morbidity. This systematic literature review aimed to identify and summarise maternal omic and multi-omic (genomics, transcriptomics, proteomics and metabolites) biomarker studies of PTB. Original research articles were retrieved from three databases: PubMed, Web of Science and Science Direct, using specified search terms for each omic discipline. PTB studies investigating genomics, transcriptomics, proteomics or metabolomics biomarkers prior to onset of labour were included. Data were collected and reviewed independently. Pathway analyses were completed on the biomarkers from non-targeted omic studies using Reactome pathway analysis tool. A total of 149 omic studies were identified; most of the literature investigated proteomic biomarkers. Pathway analysis identified several cellular processes associated with the omic biomarkers reported in the literature. Study heterogeneity was observed across the research articles, including the use of different gestation cut-offs to define PTB. Infection/inflammatory biomarkers were identified across majority of papers using a range of targeted and non-targeted approaches.
Collapse
Affiliation(s)
- Juhi K. Gupta
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Harris-Wellbeing Research Centre, University Department, Liverpool Women's Hospital, Liverpool L8 7SS, UK
| | - Ana Alfirevic
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Harris-Wellbeing Research Centre, University Department, Liverpool Women's Hospital, Liverpool L8 7SS, UK
| |
Collapse
|
10
|
Menon R, Dixon CL, Cayne S, Radnaa E, Salomon C, Sheller-Miller S. Differences in cord blood extracellular vesicle cargo in preterm and term births. Am J Reprod Immunol 2022; 87:e13521. [PMID: 35007379 DOI: 10.1111/aji.13521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE This study determined the cord plasma-derived extracellular vesicle (exosomes; 30-160 nm particles) proteomic profile in patients who had spontaneous preterm birth (PTB) or preterm premature rupture of membranes (pPROM), compared to those who delivered at term regardless of labor status. METHODS This is a cross-sectional analysis of a retrospective cohort that quantified and determined the proteomic cargo content of exosomes present in cord blood plasma samples in PTB or pPROM, and normal term in labor (TL) or term not in labor (TNIL) pregnancies. Exosomes were isolated by differential centrifugation followed by size exclusion chromatography. Exosomes were characterized by nanoparticle tracking analysis (quantity and size) and markers (dot blots for exosome markers). The exosomal proteomic profile was identified by liquid chromatography-mass spectrometry (LC-MS/MS). Ingenuity pathway analysis determined canonical pathways and biofunctions associated with dysregulated proteins. RESULTS Cord plasma exosomes have similar quantity and exhibit both tetraspanin and ESCRT protein markers specific of exosomes regardless of the conditions. Proteomics analysis exhibited several similar markers as well as very unique markers in exosomes from each condition; however, bioinformatics analysis revealed a generalized and non-specific inflammatory condition represented in exosomes from different condition that is not indicative of any specific underlying biological functions indicative of an underlying pathology. CONCLUSIONS Compared to maternal plasma and amniotic fluid exosomes, the value of cord plasma derived exosomes is limited. Quantity, character, and proteomic cargo contents in exosomes or the pathways and functions represented by differentially expressed proteins do not distinguish specific conditions regarding normal and abnormal parturition. The value of cord plasma exosome proteomic cargo has limited value as an indicator of an underlying physiology or as a biomarker of fetal well-being.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Christopher Luke Dixon
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Samir Cayne
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Enkhtuya Radnaa
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane, Australia.,Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Samantha Sheller-Miller
- Division of Basic and Translation Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
11
|
Genome and transcriptome profiling of spontaneous preterm birth phenotypes. Sci Rep 2022; 12:1003. [PMID: 35046466 PMCID: PMC8770724 DOI: 10.1038/s41598-022-04881-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/23/2021] [Indexed: 12/27/2022] Open
Abstract
Preterm birth (PTB) occurs before 37 weeks of gestation. Risk factors include genetics and infection/inflammation. Different mechanisms have been reported for spontaneous preterm birth (SPTB) and preterm birth following preterm premature rupture of membranes (PPROM). This study aimed to identify early pregnancy biomarkers of SPTB and PPROM from the maternal genome and transcriptome. Pregnant women were recruited at the Liverpool Women’s Hospital. Pregnancy outcomes were categorised as SPTB, PPROM (≤ 34 weeks gestation, n = 53), high-risk term (HTERM, ≥ 37 weeks, n = 126) or low-risk (no history of SPTB/PPROM) term (LTERM, ≥ 39 weeks, n = 188). Blood samples were collected at 16 and 20 weeks gestation from which, genome (UK Biobank Axiom array) and transcriptome (Clariom D Human assay) data were acquired. PLINK and R were used to perform genetic association and differential expression analyses and expression quantitative trait loci (eQTL) mapping. Several significant molecular signatures were identified across the analyses in preterm cases. Genome-wide significant SNP rs14675645 (ASTN1) was associated with SPTB whereas microRNA-142 transcript and PPARG1-FOXP3 gene set were associated with PPROM at week 20 of gestation and is related to inflammation and immune response. This study has determined genomic and transcriptomic candidate biomarkers of SPTB and PPROM that require validation in diverse populations.
Collapse
|
12
|
Zhang J, Chen G, Liang S, Liu J, Zhang J, Shen H, Chen Y, Duan J, Sun Z. PM 2.5 exposure exaggerates the risk of adverse birth outcomes in pregnant women with pre-existing hyperlipidemia: Modulation role of adipokines and lipidome. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 787:147604. [PMID: 33992945 DOI: 10.1016/j.scitotenv.2021.147604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 06/12/2023]
Abstract
The in-utero environmental exposure to fine particulate matter (PM2.5) might lead to adverse birth outcomes, such as low birth weight (LBW) and preterm birth (PTB), thereby increasing susceptibility to diseases in later life. However, no studies have examined the underlying mechanism through cross-omics of lipidome and adipokines profiling, as well as the possible effect modification by maternal hyperlipidemia. In total, 203 mother-newborn pairs were recruited in the birth cohort study ongoing since February 2017 in Beijing, China. Individual-level of PM2.5 exposure was estimated using a satellite data based random forest model. Cord blood lipidome and adipokines were assessed through the lipidomic approaches and antibody-based array. Multivariable logistic/linear regression models and moderation analysis were employed in this study. We observed a significantly increased risk of PTB associated with PM2.5 exposure during the second trimester, especially in pregnant women with pre-existing hyperlipidemia. 9 lipid classes and 21 adipokines were associated with PM2.5 exposure independently or significantly influenced by the interaction of maternal PM2.5 exposure and hyperlipidemia. In addition, 4 adipokines (ANGPTL4, IGFBP-2, IL-12p40, and TNF-RII) and 3 lipid classes [phosphatidylcholines (PCs), phosphatidylinositols (PIs), and triglycerides (TGs)] were related to the increased risk of PTB, indicating that inflammation, IGF/IGFBP axis, and lipolysis induced lipid homeostasis disorder of PCs, TGs, and PIs might be the possible mediators for the PM2.5-induced adverse birth outcomes. Our results substantiated the need for reducing exposure in susceptible populations.
Collapse
Affiliation(s)
- Jingyi Zhang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, People's Republic of China
| | - Gongbo Chen
- Guangdong Provincial Engineering Technology Research Center of Environmental and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Shuang Liang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, People's Republic of China
| | - Jiangyan Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, People's Republic of China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, People's Republic of China
| | - Heqing Shen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, People's Republic of China
| | - Yi Chen
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, People's Republic of China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, People's Republic of China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, People's Republic of China.
| |
Collapse
|
13
|
Couceiro J, Matos I, Mendes JJ, Baptista PV, Fernandes AR, Quintas A. Inflammatory factors, genetic variants, and predisposition for preterm birth. Clin Genet 2021; 100:357-367. [PMID: 34013526 DOI: 10.1111/cge.14001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022]
Abstract
Preterm birth is a major clinical and public health challenge, with a prevalence of 11% worldwide. It is the leading cause of death in children younger than 5 years old and represents 70% of neonatal deaths and 75% of neonatal morbidity. Despite the clinical and public health significance, this condition's etiology is still unclear, and most of the cases are spontaneous. There are several known preterm birth risk factors, including inflammatory diseases and the genetic background, although the underlying molecular mechanisms are far from understood. The present review highlights the research advances on the association between inflammatory-related genes and the increased risk for preterm delivery. The most associated genetic variants are the TNFα rs1800629, the IL1α rs17561, and the IL1RN rs2234663. Moreover, many of the genes discussed in this review are also implicated in pathologies involving inflammatory or autoimmune systems, such as periodontal disease, bowel inflammatory disease, and autoimmune rheumatic diseases. This review presents evidence suggesting a common genetic background to preterm birth, autoimmune and inflammatory diseases susceptibility.
Collapse
Affiliation(s)
- Joana Couceiro
- Centro de Investigação Interdisciplinar Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal.,UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica, Portugal.,Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal
| | - Irina Matos
- Centro de Investigação Interdisciplinar Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal
| | - José João Mendes
- Centro de Investigação Interdisciplinar Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Alexandre Quintas
- Centro de Investigação Interdisciplinar Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal.,Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário Quinta da Granja, Caparica, Portugal
| |
Collapse
|
14
|
Munro SK, Balakrishnan B, Lissaman AC, Gujral P, Ponnampalam AP. Cytokines and pregnancy: Potential regulation by histone deacetylases. Mol Reprod Dev 2021; 88:321-337. [PMID: 33904218 DOI: 10.1002/mrd.23430] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 12/26/2022]
Abstract
Cytokines are important regulators of pregnancy and parturition. Aberrant expression of proinflammatory cytokines during pregnancy contributes towards preterm labor, pre-eclampsia, and gestational diabetes mellitus. The regulation of cytokine expression in human cells is highly complex, involving interactions between environment, transcription factors, and feedback mechanisms. Recent developments in epigenetic research have made tremendous advancements in exploring histone modifications as a key epigenetic regulator of cytokine expression and the effect of their signaling molecules on various organ systems in the human body. Histone acetylation and subsequent deacetylation by histone deacetylases (HDACs) are major epigenetic regulators of protein expression in the human body. The expression of various proinflammatory cytokines, their role in normal and abnormal pregnancy, and their epigenetic regulation via HDACs will be discussed in this review.
Collapse
Affiliation(s)
- Sheryl K Munro
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Biju Balakrishnan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Abbey C Lissaman
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Palak Gujral
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Anna P Ponnampalam
- Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Physiology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Prairie E, Côté F, Tsakpinoglou M, Mina M, Quiniou C, Leimert K, Olson D, Chemtob S. The determinant role of IL-6 in the establishment of inflammation leading to spontaneous preterm birth. Cytokine Growth Factor Rev 2021; 59:118-130. [PMID: 33551331 DOI: 10.1016/j.cytogfr.2020.12.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022]
Abstract
Preterm birth (PTB) and its consequences are a major public health concern as preterm delivery is the main cause of mortality and morbidity at birth. There are many causes of PTB, but inflammation is undeniably associated with the process of premature childbirth and fetal injury. At present, treatments clinically available mostly involve attempt to arrest contractions (tocolytics) but do not directly address upstream maternal inflammation on development of the fetus. One of the possible solutions may lie in the modulation of inflammatory mediators. Of the many pro-inflammatory cytokines involved in the induction of PTB, IL-6 stands out for its pleiotropic effects and its involvement in both acute and chronic inflammation. Here, we provide a detailed review of the effects of IL-6 on the timing of childbirth, its occurrence during PTB and its indissociable roles with associated fetal tissue damage.
Collapse
Affiliation(s)
- Elizabeth Prairie
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - France Côté
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Marika Tsakpinoglou
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Michael Mina
- Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada.
| | - Kelycia Leimert
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - David Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, H3T 1C5, Canada; Department of Pharmacology, Université de Montréal, Montreal, H3T 1J4, Canada.
| |
Collapse
|
16
|
Menon R, Dixon CL, Sheller-Miller S, Fortunato SJ, Saade GR, Palma C, Lai A, Guanzon D, Salomon C. Quantitative Proteomics by SWATH-MS of Maternal Plasma Exosomes Determine Pathways Associated With Term and Preterm Birth. Endocrinology 2019; 160:639-650. [PMID: 30668697 PMCID: PMC6388657 DOI: 10.1210/en.2018-00820] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/13/2019] [Indexed: 12/11/2022]
Abstract
Exosomes are membrane-bound nanovesicles that transport molecular signals between cells. This study determined changes in maternal plasma exosome proteomics contents in term and preterm births. Maternal plasma (MP) samples were collected from group 1: term not in labor (TNIL, n = 13); group 2: term in labor (TL, n = 11); group 3: preterm premature rupture of membranes (pPROM, n = 8); and group 4: preterm birth (PTB, n = 13). Exosomes isolated from plasma by differential density centrifugation followed by size exclusion chromatography were characterized by morphology (electron microscopy), quantity and size (nanoparticle tracking analysis), and markers (western blot). A quantitative, information-independent acquisition [sequential windowed acquisition of all theoretical mass spectra (SWATH-MS)] approach was used to determine the protein profile in exosomes. Ingenuity Pathway Analysis determined pathways associated with the protein profile identified in exosomes. MP exosomes were spherical, had a mean diameter of 120 nm, and were positive for exosomal proteins CD63 and TSG101 irrespective of pregnancy status. No distinct changes in exosome quantities were seen in maternal circulation across the groups. SWATH-MS identified 72 statistically significant proteins across the groups studied. Bioinformatics analysis showed the proteins within the exosomes in TNIL, TL, pPROM, and PTB target pathways mainly associated with inflammatory and metabolic signals. Exosomal data suggest that homeostatic imbalances, specifically inflammatory and endocrine signaling, might disrupt pregnancy maintenance resulting in labor-related changes both at term and preterm. Reflection of physiologic changes in exosomes is suggestive of its usefulness as biomarkers and cellular function indicators.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
- Correspondence: Ramkumar Menon, MS, PhD, Department of Obstetrics and Gynecology, 11.138 Medical Research Building, The University of Texas Medical Branch, Galveston, Texas 77555. E-mail: ; or Carlos Salomon, PhD, Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Faculty of Health Sciences, University of Queensland, Building 71/918, Royal Brisbane Hospital, Herston, Queensland 4029, Brisbane, Australia. E-mail:
| | - Christopher Luke Dixon
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Stephen J Fortunato
- Department of Obstetrics and Gynecology, Ochsner Baptist Hospital, New Orleans, Louisiana
| | - George R Saade
- Division of Maternal-Fetal Medicine & Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Carlos Palma
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Herston, Queensland, Brisbane, Australia
| | - Andrew Lai
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Herston, Queensland, Brisbane, Australia
| | - Dominic Guanzon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Herston, Queensland, Brisbane, Australia
| | - Carlos Salomon
- Department of Obstetrics and Gynecology, Ochsner Baptist Hospital, New Orleans, Louisiana
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Herston, Queensland, Brisbane, Australia
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University
of Concepción, Concepción, Chile
- Correspondence: Ramkumar Menon, MS, PhD, Department of Obstetrics and Gynecology, 11.138 Medical Research Building, The University of Texas Medical Branch, Galveston, Texas 77555. E-mail: ; or Carlos Salomon, PhD, Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Faculty of Health Sciences, University of Queensland, Building 71/918, Royal Brisbane Hospital, Herston, Queensland 4029, Brisbane, Australia. E-mail:
| |
Collapse
|
17
|
Bhavnani SK, Dang B, Kilaru V, Caro M, Visweswaran S, Saade G, Smith AK, Menon R. Methylation differences reveal heterogeneity in preterm pathophysiology: results from bipartite network analyses. J Perinat Med 2018; 46:509-521. [PMID: 28665803 PMCID: PMC5971156 DOI: 10.1515/jpm-2017-0126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/26/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recent studies have shown that epigenetic differences can increase the risk of spontaneous preterm birth (PTB). However, little is known about heterogeneity underlying such epigenetic differences, which could lead to hypotheses for biological pathways in specific patient subgroups, and corresponding targeted interventions critical for precision medicine. Using bipartite network analysis of fetal DNA methylation data we demonstrate a novel method for classification of PTB. METHODS The data consisted of DNA methylation across the genome (HumanMethylation450 BeadChip) in cord blood from 50 African-American subjects consisting of 22 cases of early spontaneous PTB (24-34 weeks of gestation) and 28 controls (>39 weeks of gestation). These data were analyzed using a combination of (1) a supervised method to select the top 10 significant methylation sites, (2) unsupervised "subject-variable" bipartite networks to visualize and quantitatively analyze how those 10 methylation sites co-occurred across all the subjects, and across only the cases with the goal of analyzing subgroups and their underlying pathways, and (3) a simple linear regression to test whether there was an association between the total methylation in the cases, and gestational age. RESULTS The bipartite network analysis of all subjects and significant methylation sites revealed statistically significant clustering consisting of an inverse symmetrical relationship in the methylation profiles between a case-enriched subgroup and a control-enriched subgroup: the former was predominantly hypermethylated across seven methylation sites, and hypomethylated across three methylation sites, whereas the latter was predominantly hypomethylated across the above seven methylation sites and hypermethylated across the three methylation sites. Furthermore, the analysis of only cases revealed one subgroup that was predominantly hypomethylated across seven methylation sites, and another subgroup that was hypomethylated across all methylation sites suggesting the presence of heterogeneity in PTB pathophysiology. Finally, the analysis found a strong inverse linear relationship between total methylation and gestational age suggesting that methylation differences could be used as predictive markers for gestational length. CONCLUSIONS The results demonstrate that unsupervised bipartite networks helped to identify a complex but comprehensible data-driven hypotheses related to patient subgroups and inferences about their underlying pathways, and therefore were an effective complement to supervised approaches currently used.
Collapse
Affiliation(s)
- Suresh K. Bhavnani
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Bryant Dang
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Varun Kilaru
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Maria Caro
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Shyam Visweswaran
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Saade
- Department of Obstetrics and Gynecology, Division of Maternal Fetal-Medicine Perinatal Research, University of Texas Medical Branch, Galveston, Texas
| | - Alicia K. Smith
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Maternal Fetal-Medicine Perinatal Research, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
18
|
Strauss JF, Romero R, Gomez-Lopez N, Haymond-Thornburg H, Modi BP, Teves ME, Pearson LN, York TP, Schenkein HA. Spontaneous preterm birth: advances toward the discovery of genetic predisposition. Am J Obstet Gynecol 2018; 218:294-314.e2. [PMID: 29248470 PMCID: PMC5834399 DOI: 10.1016/j.ajog.2017.12.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 02/08/2023]
Abstract
Evidence from family and twin-based studies provide strong support for a significant contribution of maternal and fetal genetics to the timing of parturition and spontaneous preterm birth. However, there has been only modest success in the discovery of genes predisposing to preterm birth, despite increasing sophistication of genetic and genomic technology. In contrast, DNA variants associated with other traits/diseases have been identified. For example, there is overwhelming evidence that suggests that the nature and intensity of an inflammatory response in adults and children are under genetic control. Because inflammation is often invoked as an etiologic factor in spontaneous preterm birth, the question of whether spontaneous preterm birth has a genetic predisposition in the case of pathologic inflammation has been of long-standing interest to investigators. Here, we review various genetic approaches used for the discovery of preterm birth genetic variants in the context of inflammation-associated spontaneous preterm birth. Candidate gene studies have sought genetic variants that regulate inflammation in the mother and fetus; however, the promising findings have often not been replicated. Genome-wide association studies, an approach to the identification of chromosomal loci responsible for complex traits, have also not yielded compelling evidence for DNA variants predisposing to preterm birth. A recent genome-wide association study that included a large number of White women (>40,000) revealed that maternal loci contribute to preterm birth. Although none of these loci harbored genes directly related to innate immunity, the results were replicated. Another approach to identify DNA variants predisposing to preterm birth is whole exome sequencing, which examines the DNA sequence of protein-coding regions of the genome. A recent whole exome sequencing study identified rare mutations in genes encoding for proteins involved in the negative regulation (dampening) of the innate immune response (eg, CARD6, CARD8, NLRP10, NLRP12, NOD2, TLR10) and antimicrobial peptide/proteins (eg, DEFB1, MBL2). These findings support the concept that preterm labor, at least in part, has an inflammatory etiology, which can be induced by pathogens (ie, intraamniotic infection) or "danger signals" (alarmins) released during cellular stress or necrosis (ie, sterile intraamniotic inflammation). These findings support the notion that preterm birth has a polygenic basis that involves rare mutations or damaging variants in multiple genes involved in innate immunity and host defense mechanisms against microbes and their noxious products. An overlap among the whole exome sequencing-identified genes and other inflammatory conditions associated with preterm birth, such as periodontal disease and inflammatory bowel disease, was observed, which suggests a shared genetic substrate for these conditions. We propose that whole exome sequencing, as well as whole genome sequencing, is the most promising approach for the identification of functionally significant genetic variants responsible for spontaneous preterm birth, at least in the context of pathologic inflammation. The identification of genes that contribute to preterm birth by whole exome sequencing, or whole genome sequencing, promises to yield valuable population-specific biomarkers to identify the risk for spontaneous preterm birth and potential strategies to mitigate such a risk.
Collapse
Affiliation(s)
- Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA.
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute for 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.
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute for 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 and the Department of Immunology, Microbiology and Biochemistry, Wayne State University School of Medicine, Detroit, MI
| | - Hannah Haymond-Thornburg
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Bhavi P Modi
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Laurel N Pearson
- Department of Anthropology, Pennsylvania State University, University Park, PA
| | - Timothy P York
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Harvey A Schenkein
- Department of Periodontics, Virginia Commonwealth University School of Dentistry, Richmond, VA
| |
Collapse
|
19
|
Wang LK, Huang MC, Liu CC, Chen CP. Second-trimester plasma mannose-binding lectin levels and risk of preterm birth. J Matern Fetal Neonatal Med 2016; 30:678-683. [PMID: 27124763 DOI: 10.1080/14767058.2016.1182978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate whether mannose-binding lectin (MBL) gene polymorphisms and low levels of second-trimester plasma MBL were significant risk factors for preterm birth in Taiwanese women. METHODS We conducted a prospective longitudinal study to explore the associations of MBL2 gene single nucleotide polymorphisms and plasma MBL levels between preterm birth and term controls. Blood samples were collected at 16-23 weeks of gestation, and were divided into 51 mothers with preterm births and 255 term controls after delivery. Blood samples were further collected at delivery from 11 mothers with term delivery and 9 with preterm births. DNA was isolated, and polymorphisms in exon 1, the promoter untranslated regions of MBL2 were determined by polymerase chain reaction. The plasma concentrations of MBL were measured by enzyme-linked immunosorbent assay. RESULTS There is a positive correlation between SNP genotypes and second-trimester plasma MBL levels. Among mothers with preterm births, a higher frequency of specific genotypes with low MBL levels was not observed. The second-trimester plasma MBL levels were not significantly different between mothers with preterm births (N = 51) and term deliveries (N = 255). However, among mothers (N = 11) with term pregnancies, the MBL plasma level significantly increased from the second trimester to delivery, whereas in mothers (N = 9) who developed preterm delivery, the MBL level did not significantly change. CONCLUSION Genotypes associated with low levels of plasma MBL during pregnancy did not increase the risk of preterm births. A low second-trimester plasma MBL level is therefore not a predictor for the development of preterm birth.
Collapse
Affiliation(s)
- Liang-Kai Wang
- a Division of High Risk Pregnancy , MacKay Memorial Hospital , Taipei , Taiwan
| | - Ming-Chao Huang
- b Division of General Obstetrics and Gynecology , MacKay Memorial Hospital , Taipei , Taiwan , and
| | - Chang-Ching Liu
- c Department of Medical Research , MacKay Memorial Hospital , Taipei , Taiwan
| | - Chie-Pein Chen
- a Division of High Risk Pregnancy , MacKay Memorial Hospital , Taipei , Taiwan.,c Department of Medical Research , MacKay Memorial Hospital , Taipei , Taiwan
| |
Collapse
|
20
|
Ramos BRDA, Mendes ND, Tanikawa AA, Amador MAT, dos Santos NPC, dos Santos SEB, Castelli EC, Witkin SS, da Silva MG. Ancestry informative markers and selected single nucleotide polymorphisms in immunoregulatory genes on preterm labor and preterm premature rupture of membranes: a case control study. BMC Pregnancy Childbirth 2016; 16:30. [PMID: 26846412 PMCID: PMC4743423 DOI: 10.1186/s12884-016-0823-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/31/2016] [Indexed: 01/15/2023] Open
Abstract
Background A genetic predisposition to Preterm Labor (PTL) and Preterm Premature Rupture of Membranes (PPROM) has been suggested; however the relevance of polymorphisms and ancestry to susceptibility to PTL and PPROM in different populations remains unclear. The aim of this study was to evaluate the contribution of maternal and fetal SNPs in the IL1B, IL6, IL6R, TNFA, TNFR, IL10, TLR2, TLR4, MMP9, TIMP1 and TIMP2 genes and the influence of ancestry background in the susceptibility to PTL or PPROM in Brazilian women. Methods Case–control study conducted at a tertiary hospital in São Paulo State, Brazil. We included women with PTL or PPROM and their babies (PTL: 136 women and 88 babies; PPROM: 65 women and 44 babies). Control group included 402 mother-babies pairs of term deliveries. Oral swabs were collected for identification of AIMs by fragment analysis and SNPs by Taqman® SNP Genotyping Assays and PCR. Linkage Disequilibrium and Hardy-Weinberg proportions were evaluated using Genepop 3.4. Haplotypes were inferred using the PHASE algorithm. Allele, genotype and haplotype frequencies were compared by Fisher’s exact test or χ2 and Odds Ratio. Logistic regression was performed. Clinical and sociodemographic data were analyzed by Fisher’s exact test and Mann–Whitney. Results PTL was associated with European ancestry and smoking while African ancestry was protective. The fetal alleles IL10-592C (rs800872) and IL10-819C (rs1800871) were also associated with PTL and the maternal haplotype TNFA-308G-238A was protective. Maternal presence of IL10-1082G (rs1800896) and TLR2A (rs4696480) alleles increased the risk for PPROM while TNFA-238A (rs361525) was protective. Family history of PTL/PPROM was higher in cases, and time to delivery was influenced by IL1B-31T (rs1143627) and TLR4-299G (rs4986790). Conclusion There is an association between European ancestry and smoking and PTL in our Brazilian population sample. The presence of maternal or fetal alleles that modify the inflammatory response increase the susceptibility to PTL and PPROM. The family history of PTL/PPROM reinforces a role for genetic polymorphisms in susceptibility to these outcomes.
Collapse
Affiliation(s)
- Bruna Ribeiro de Andrade Ramos
- Department of Pathology, Botucatu Medical School, São Paulo State University - UNESP, Distrito de Rubião Júnior, 18618-970, Botucatu, São Paulo, Brazil.
| | - Niele Dias Mendes
- Department of Pathology, Botucatu Medical School, São Paulo State University - UNESP, Distrito de Rubião Júnior, 18618-970, Botucatu, São Paulo, Brazil.
| | - Aline Aki Tanikawa
- Blood Transfusion Center, Botucatu Medical School, São Paulo State University - UNESP, Botucatu, São Paulo, Brazil.
| | | | | | | | - Erick C Castelli
- Department of Pathology, Botucatu Medical School, São Paulo State University - UNESP, Distrito de Rubião Júnior, 18618-970, Botucatu, São Paulo, Brazil.
| | - Steven S Witkin
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA.
| | - Márcia Guimarães da Silva
- Department of Pathology, Botucatu Medical School, São Paulo State University - UNESP, Distrito de Rubião Júnior, 18618-970, Botucatu, São Paulo, Brazil.
| |
Collapse
|
21
|
Polettini J, Behnia F, Taylor BD, Saade GR, Taylor RN, Menon R. Telomere Fragment Induced Amnion Cell Senescence: A Contributor to Parturition? PLoS One 2015; 10:e0137188. [PMID: 26397719 PMCID: PMC4580414 DOI: 10.1371/journal.pone.0137188] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/13/2015] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress (OS)-induced senescence of the amniochorion has been associated with parturition at term. We investigated whether telomere fragments shed into the amniotic fluid (AF) correlated with labor status and tested if exogenous telomere fragments (T-oligos) could induce human and murine amnion cell senescence. In a cross-sectional clinical study, AF telomere fragment concentrations quantitated by a validated real-time PCR assay were higher in women in labor at term compared to those not in labor. In vitro treatment of primary human amnion epithelial cells with 40 μM T-oligos ([TTAGGG]2) that mimic telomere fragments, activated p38MAPK, produced senescence-associated (SA) β-gal staining and increased interleukin (IL)-6 and IL-8 production compared to cells treated with complementary DNA sequences (Cont-oligos, [AATCCC]2). T-oligos injected into the uteri of pregnant CD1 mice on day 14 of gestation, led to increased p38MAPK, SA-β-gal (SA β-gal) staining in murine amniotic sacs and higher AF IL-8 levels on day 18, compared to saline treated controls. In summary, term labor AF samples had higher telomere fragments than term not in labor AF. In vitro and in situ telomere fragments increased human and murine amnion p38MAPK, senescence and inflammatory cytokines. We propose that telomere fragments released from senescent fetal cells are indicative of fetal cell aging. Based on our data, these telomere fragments cause oxidative stress associated damages to the term amniotic sac and force them to release other DAMPS, which, in turn, provide a sterile immune response that may be one of the many inflammatory signals required to initiate parturition at term.
Collapse
Affiliation(s)
- Jossimara Polettini
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- Department of Pathology, Botucatu Medical School, UNESP–Univ. Estadual Paulista, Botucatu, Sao Paulo, Brazil
| | - Faranak Behnia
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Brandie D. Taylor
- Department of Epidemiology & Biostatistics, Texas A&M University System Health Science Center, College Station, Texas, United States of America
| | - George R. Saade
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
| | - Robert N. Taylor
- Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, North Carolina, United States of America
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
22
|
Liu Y, Yao CJ, Tao FB, Luo CM, Cao Y, Su-Juan Z, Chen YY, Zhu XX, Cao JY. Association between maternal tumor necrosis factor-α G308A polymorphism and interferon-γ A874T polymorphism and risk of preterm birth: a meta-analysis. Eur J Obstet Gynecol Reprod Biol 2015; 190:11-9. [PMID: 25917437 DOI: 10.1016/j.ejogrb.2015.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 03/04/2015] [Accepted: 04/07/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVE(S) This article was undertaken to investigate the association between tumor necrosis factor-α (TNF-α) G308A polymorphism and interferon-γ (INF-γ) A874T polymorphism and risk of preterm birth (PTB) by performing a meta-analysis of available studies. STUDY DESIGN Articles were chosen based on PubMed, EMBASE, Web of science, and China Biology Medicine (CBM) databases with no language restriction from their inceptions to 1 March, 2014. Specific inclusion criteria were used to evaluate articles. Meta-analysis was performed by using a random or fixed effect model with STATA 11.0 software. We estimated the summary odds ratios (ORs) with its corresponding 95% confidence interval (95%CI) to assess the association. RESULTS 21 eligible case-control studies with a total of 2103 cases and 5070 controls were finally included into this meta-analysis. Pooled analysis showed that A allele of TNF-α G308A was not associated with increased PTB risk (OR=0.84, 95%CI: 0.65-1.07, p=0.167 for G vs. A). Stratifying analysis for ethnicity and different definition of PTB also indicated that A allele was not associated with increased PTB risk. However, the meta-analysis showed that INF-γ A874T polymorphism was associated with the increased risk of PTB (OR=1.14, 95%CI: 1.11-1.73, p=0.004 for A vs. T). Stratifying analysis was not performed due to the small sample size. CONCLUSION(S) TNF-α G308A polymorphism was not associated with an increased risk of PTB, but INF-γ A874T polymorphism may contribute to increasing susceptibility to PTB. Detection of polymorphism of INF-γ A874T might be a promising biomarker for the diagnosis and prognosis of preterm delivery.
Collapse
Affiliation(s)
- Ying Liu
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Ci-Jiang Yao
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Fang-Biao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Chun-Miao Luo
- Department of Cardiology, The Second People's Hospital of Hefei, Anhui, China
| | - Yi Cao
- Department of Pharmacy, The Second Affiliated Hospital of Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Zhao Su-Juan
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Yan-Yan Chen
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Xiao-Xia Zhu
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Ji-Yu Cao
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China; The Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
23
|
Menon R, Bhat G, Saade GR, Spratt H. Multivariate adaptive regression splines analysis to predict biomarkers of spontaneous preterm birth. Acta Obstet Gynecol Scand 2014; 93:382-91. [PMID: 24461165 DOI: 10.1111/aogs.12344] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To develop classification models of demographic/clinical factors and biomarker data from spontaneous preterm birth in African Americans and Caucasians. DESIGN Secondary analysis of biomarker data using multivariate adaptive regression splines (MARS), a supervised machine learning algorithm method. SETTING Analysis of data on 36 biomarkers from 191 women was reduced by MARS to develop predictive models for preterm birth in African Americans and Caucasians. SAMPLES Maternal plasma, cord plasma collected at admission for preterm or term labor and amniotic fluid at delivery. METHODS Data were partitioned into training and testing sets. Variable importance, a relative indicator (0-100%) and area under the receiver operating characteristic curve (AUC) characterized results. RESULTS Multivariate adaptive regression splines generated models for combined and racially stratified biomarker data. Clinical and demographic data did not contribute to the model. Racial stratification of data produced distinct models in all three compartments. In African Americans maternal plasma samples IL-1RA, TNF-α, angiopoietin 2, TNFRI, IL-5, MIP1α, IL-1β and TGF-α modeled preterm birth (AUC train: 0.98, AUC test: 0.86). In Caucasians TNFR1, ICAM-1 and IL-1RA contributed to the model (AUC train: 0.84, AUC test: 0.68). African Americans cord plasma samples produced IL-12P70, IL-8 (AUC train: 0.82, AUC test: 0.66). Cord plasma in Caucasians modeled IGFII, PDGFBB, TGF-β1 , IL-12P70, and TIMP1 (AUC train: 0.99, AUC test: 0.82). Amniotic fluid in African Americans modeled FasL, TNFRII, RANTES, KGF, IGFI (AUC train: 0.95, AUC test: 0.89) and in Caucasians, TNF-α, MCP3, TGF-β3 , TNFR1 and angiopoietin 2 (AUC train: 0.94 AUC test: 0.79). CONCLUSIONS Multivariate adaptive regression splines models multiple biomarkers associated with preterm birth and demonstrated racial disparity.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, University of Texas Medical Branch at Galveston, Galveston, Texas, USA; Center for Diseases Control (CDC), Atlanta, Georgia, USA
| | | | | | | |
Collapse
|
24
|
Mitsuya K, Singh N, Sooranna SR, Johnson MR, Myatt L. Epigenetics of human myometrium: DNA methylation of genes encoding contraction-associated proteins in term and preterm labor. Biol Reprod 2014; 90:98. [PMID: 24571989 PMCID: PMC11488454 DOI: 10.1095/biolreprod.113.113209] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 10/16/2013] [Accepted: 02/25/2014] [Indexed: 12/30/2022] Open
Abstract
Preterm birth involves the interaction of societal and environmental factors potentially modulating the length of gestation via the epigenome. An established form of epigenetic regulation is DNA methylation where promoter hypermethylation is associated with gene repression. We hypothesized we would find differences in DNA methylation in the myometrium of women with preterm labor of different phenotypes versus normal term labor. Myometrial tissue was obtained at cesarean section at term with or without labor, preterm without labor, idiopathic preterm labor, and twin gestations with labor. Genomic DNA was isolated, and samples in each group were combined and analyzed on a NimbleGen 2.1M human DNA methylation array. Differences in methylation from -8 to +3 kb of transcription start sites of 22 contraction-associated genes were determined. Cytosine methylation was not present in CpG islands of any gene but was present outside of CpG islands in shores and shelves in 19 genes. No differential methylation was found across the tissue groups for six genes (PTGES3L, PTGER2, PTGER4, PTGFRN, ESR2, and GJA1). For 13 genes, differential methylation occurred in several patterns between tissue groups. We find a correlation between hypomethylation and increased mRNA expression of PTGES/mPGES-1, indicating potential functional relevance of methylation, but no such correlation for PTGS2/COX-2, suggesting other regulatory mechanisms for PTGS2 at labor. The majority of differential DNA methylation of myometrial contraction-associated genes with different labor phenotypes occurs outside of CpG islands in gene promoters, suggesting that the entirety of DNA methylation across the genome should be considered.
Collapse
Affiliation(s)
- Kohzoh Mitsuya
- Center for Pregnancy and Newborn Research, Department of Obstetrics and Gynecology, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | | | | | | | | |
Collapse
|
25
|
Schminkey DL, Groer M. Imitating a stress response: a new hypothesis about the innate immune system's role in pregnancy. Med Hypotheses 2014; 82:721-9. [PMID: 24698849 DOI: 10.1016/j.mehy.2014.03.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 03/08/2014] [Indexed: 11/24/2022]
Abstract
Recent research challenges long-held hypotheses about mechanisms through which pregnancy induces maternal immune suppression or tolerance of the embryo/fetus. It is now understood that normal pregnancy engages the immune system and that the immune milieu changes with advancing gestation. We suggest that pregnancy mimics the innate immune system's response to stress, causing a sterile inflammatory response that is necessary for successful reproduction. The relationship between external stressors and immunomodulation in pregnancy has been acknowledged, but the specific mechanisms are still being explicated. Implantation and the first trimester are times of immune activation and intensive inflammation in the uterine environment. A period of immune quiescence during the second trimester allows for the growth and development of the maturing fetus. Labor is also an inflammatory event. The length of gestation and timing of parturition can be influenced by environmental stressors. These stressors affect pregnancy through neuroendocrine interaction with the immune system, specifically through the hypothalamic-pituitary-adrenal (HPA) axis and the hypothalamic-pituitary-ovarian axis. Trophoblastic cells that constitute the maternal-fetal interface appear to harness the maternal immune system to promote and maximize the reproductive success of the mother and fetus. Pregnancy is a time of upregulated innate immune responses and decreased adaptive, cell-mediated responses. The inflammatory processes of pregnancy resemble an immune response to brief naturalistic stressors: there is a shift from T helper (Th) 1 to T helper (Th) 2 dominant adaptive immunity with a concomitant shift in cytokine production, decreased proliferation of T cells, and decreased cytotoxicity of natural killer (NK) cells. Inclusion of both murine and human studies, allows an exploration of insights into how trophoblasts influence the activity of the maternal innate immune system during gestation.
Collapse
|
26
|
Menon R. Race and genetics in understanding the complexities of preterm birth. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/eog.09.48] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Harmon QE, Engel SM, Olshan AF, Moran T, Stuebe AM, Luo J, Wu MC, Avery CL. Association of polymorphisms in natural killer cell-related genes with preterm birth. Am J Epidemiol 2013; 178:1208-18. [PMID: 23982189 DOI: 10.1093/aje/kwt108] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inflammation is implicated in preterm birth, but genetic studies of inflammatory genes have yielded inconsistent results. Maternal DNA from 1,646 participants in the Pregnancy, Infection, and Nutrition Cohort, enrolled in Orange and Wake counties, North Carolina (1995-2005), were genotyped for 432 tag single-nucleotide polymorphisms (SNPs) in 30 candidate genes. Gene-level and SNP associations were modeled within strata of genetic ancestry. Six genes were associated with preterm birth among European Americans: interleukin 12A (IL12A); colony-stimulating factor 2 (CSF2); interferon γ receptor 2 (IFNGR2); killer cell immunoglobulin-like receptor, three domain, long cytoplasmic tail, 2 (KIR3DL2); interleukin 4 (IL4); and interleukin 13 (IL13). Of these, relatively strong single-SNP associations were seen in IFNGR2 and KIR3DL2. Among the 4 genes related to natural killer cell function, 2 (IL12A and CSF2) were consistently associated with reduced risk of prematurity for both European and African Americans. SNPs tagging a locus control region for IL4 and IL13 were associated with an increased risk of spontaneous preterm birth for European Americans (rs3091307; risk ratio = 1.9; 95% confidence interval: 1.4, 2.5). Although gene-level associations were detected only in European Americans, single-SNP associations among European and African Americans were often similar in direction, though estimated with less precision among African Americans. In conclusion, we identified novel associations between variants in the natural killer cell immune pathway and prematurity in this biracial US population.
Collapse
|
28
|
Bhat G, Williams SM, Saade GR, Menon R. Biomarker interactions are better predictors of spontaneous preterm birth. Reprod Sci 2013; 21:340-50. [PMID: 23885102 DOI: 10.1177/1933719113497285] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The objective of this study was to assess the role of biomarker interactions as predictors of spontaneous preterm birth (PTB) using multifactor dimensionality reduction (MDR) analysis. With MDR, a nonparametric, unsupervised, model-free approach, we tested for biomarker interactions within maternal-fetal compartments in 2 racial groups: African Americans (AA) and Caucasians (C). STUDY DESIGN A total of 36 biomarkers from maternal plasma (MP), cord plasma (CP), and amniotic fluid (AF) were analyzed from 191 patients. The MDR combined attribute selection, construction, and classification to detect biomarker interactions that were assessed for generality and significance using 10× cross-validation and permutation testing. Selected significant interactive models were replicated with additional samples. RESULTS The interactive model containing interleukin (IL)-2, angiopoietin 2 (ANGPT-2), and IL-6 receptor was significant in AA MP. In AA CP, the IL-8 and tumor necrosis factor (TNF) receptor 1 model was significant. In AA AF, the ANGPT-2 and macrophage inflammatory protein 1 alpha model was significant. Replication of the AA MP model using 54 additional AA MP samples confirmed predictability of these biomarkers. In C AF, interaction was observed between ANGPT-2, monocyte chemotactic protein 3, and TNF-α, but no other interactions were significant in C. CONCLUSIONS Using MDR, we identified biomarker interactions that are predictors of PTB even in the absence of a main effect with a single biomarker.
Collapse
Affiliation(s)
- G Bhat
- 1Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, The University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | |
Collapse
|
29
|
Non-invasively collected amniotic fluid as a source of possible biomarkers for premature rupture of membranes investigated by proteomic approach. Arch Gynecol Obstet 2013; 289:299-306. [PMID: 23872981 DOI: 10.1007/s00404-013-2967-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE Preterm delivery is one of the main causes of perinatal morbidity and mortality and it accounts for 75 % of perinatal mortality and more than half of the long-term morbidity. We applied a proteomic approach based on mass spectrometry (MS) for biomarkers discovery of preterm premature rupture of membranes (pPROM) by investigating amniotic fluid (AF) invasively and non-invasively collected. METHODS Amniotic fluid was obtained from vagina of women with pPROM (group 1), PROM at term (group 2) and by genetic amniocentesis (group 3). Pre-fractionated AF proteome was analyzed through matrix assisted laser desorption ionization-time of flight (MALDI-TOF) MS. The characterization of proteins/peptides of interest was obtained by high performance liquid chromatography-electrospray tandem MS. RESULTS Three peptides overexpressed in pPROM and able to discriminate the groups 1 and 2 were detected. One peptide was identified as the fragment Gly452LAVPDGPLGLPPKPro466 of the protein KIAA1522, expressed by fetal brain and liver. This peptide was overexpressed in a patient of the group 3, completely asymptomatic at the time of the amniocentesis, who later developed pPROM. CONCLUSION Amniotic fluid invasively and non-invasively collected can be analyzed by MALDI-TOF MS to obtain proteomic profiles. Proteomic analysis identified a peptide with promising diagnostic capability for pPROM.
Collapse
|
30
|
Bezold KY, Karjalainen MK, Hallman M, Teramo K, Muglia LJ. The genomics of preterm birth: from animal models to human studies. Genome Med 2013; 5:34. [PMID: 23673148 PMCID: PMC3707062 DOI: 10.1186/gm438] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Preterm birth (delivery at less than 37 weeks of gestation) is the leading cause of infant mortality worldwide. So far, the application of animal models to understand human birth timing has not substantially revealed mechanisms that could be used to prevent prematurity. However, with amassing data implicating an important role for genetics in the timing of the onset of human labor, the use of modern genomic approaches, such as genome-wide association studies, rare variant analyses using whole-exome or genome sequencing, and family-based designs, holds enormous potential. Although some progress has been made in the search for causative genes and variants associated with preterm birth, the major genetic determinants remain to be identified. Here, we review insights from and limitations of animal models for understanding the physiology of parturition, recent human genetic and genomic studies to identify genes involved in preterm birth, and emerging areas that are likely to be informative in future investigations. Further advances in understanding fundamental mechanisms, and the development of preventative measures, will depend upon the acquisition of greater numbers of carefully phenotyped pregnancies, large-scale informatics approaches combining genomic information with information on environmental exposures, and new conceptual models for studying the interaction between the maternal and fetal genomes to personalize therapies for mothers and infants. Information emerging from these advances will help us to identify new biomarkers for earlier detection of preterm labor, develop more effective therapeutic agents, and/or promote prophylactic measures even before conception.
Collapse
Affiliation(s)
- Katherine Y Bezold
- Center for Prevention of Preterm Birth and Molecular and Developmental Biology Program, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Minna K Karjalainen
- Department of Pediatrics, Institute of Clinical Medicine, University of Oulu, Oulu, 90014, Finland
| | - Mikko Hallman
- Department of Pediatrics, Institute of Clinical Medicine, University of Oulu, Oulu, 90014, Finland
| | - Kari Teramo
- Department of Obstetrics and Gynecology, University Central Hospital, Helsinki, 00029 Finland
| | - Louis J Muglia
- Center for Prevention of Preterm Birth and Molecular and Developmental Biology Program, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
31
|
Wu W, Clark EAS, Stoddard GJ, Watkins WS, Esplin MS, Manuck TA, Xing J, Varner MW, Jorde LB. Effect of interleukin-6 polymorphism on risk of preterm birth within population strata: a meta-analysis. BMC Genet 2013; 14:30. [PMID: 23617681 PMCID: PMC3639799 DOI: 10.1186/1471-2156-14-30] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 04/12/2013] [Indexed: 11/24/2022] Open
Abstract
Background Because of the role of inflammation in preterm birth (PTB), polymorphisms in and near the interleukin-6 gene (IL6) have been association study targets. Several previous studies have assessed the association between PTB and a single nucleotide polymorphism (SNP), rs1800795, located in the IL6 gene promoter region. Their results have been inconsistent and SNP frequencies have varied strikingly among different populations. We therefore conducted a meta-analysis with subgroup analysis by population strata to: (1) reduce the confounding effect of population structure, (2) increase sample size and statistical power, and (3) elucidate the association between rs1800975 and PTB. Results We reviewed all published papers for PTB phenotype and SNP rs1800795 genotype. Maternal genotype and fetal genotype were analyzed separately and the analyses were stratified by population. The PTB phenotype was defined as gestational age (GA) < 37 weeks, but results from earlier GA were selected when available. All studies were compared by genotype (CC versus CG+GG), based on functional studies. For the maternal genotype analysis, 1,165 PTBs and 3,830 term controls were evaluated. Populations were stratified into women of European descent (for whom the most data were available) and women of heterogeneous origin or admixed populations. All ancestry was self-reported. Women of European descent had a summary odds ratio (OR) of 0.68, (95% confidence interval (CI) 0.51 – 0.91), indicating that the CC genotype is protective against PTB. The result for non-European women was not statistically significant (OR 1.01, 95% CI 0.59 - 1.75). For the fetal genotype analysis, four studies were included; there was no significant association with PTB (OR 0.98, 95% CI 0.72 - 1.33). Sensitivity analysis showed that preterm premature rupture of membrane (PPROM) may be a confounding factor contributing to phenotype heterogeneity. Conclusions IL6 SNP rs1800795 genotype CC is protective against PTB in women of European descent. It is not significant in other heterogeneous or admixed populations, or in fetal genotype analysis. Population structure is an important confounding factor that should be controlled for in studies of PTB.
Collapse
Affiliation(s)
- Wilfred Wu
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Alleman BW, Myking S, Ryckman KK, Myhre R, Feingold E, Feenstra B, Geller F, Boyd HA, Shaffer JR, Zhang Q, Begum F, Crosslin D, Doheny K, Pugh E, Pay ASD, Østensen IH, Morken NH, Magnus P, Marazita ML, Jacobsson B, Melbye M, Murray JC, Gene, Environment Association Studies (GENEVA) Consortium, Norwegian Mother and Child Cohort Study (MoBA) Genome-wide Association Study (GWAS) Group. No observed association for mitochondrial SNPs with preterm delivery and related outcomes. Pediatr Res 2012; 72:539-44. [PMID: 22902432 PMCID: PMC3694399 DOI: 10.1038/pr.2012.112] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Preterm delivery (PTD) is the leading cause of neonatal morbidity and mortality. Epidemiologic studies indicate recurrence of PTD is maternally inherited, creating a strong possibility that mitochondrial variants contribute to its etiology. This study examines the association between mitochondrial genotypes and PTD and related outcomes. METHODS This study combined, through meta-analysis, two case-control, genome-wide association studies: one from the Danish National Birth Cohort Study and one from the Norwegian Mother and Child Cohort Study (MoBa) conducted by the Norwegian Institute of Public Health. The outcomes of PTD (≤36 wk), very PTD (≤32 wk), and preterm prelabor rupture of membranes (PPROM) were examined. A total of 135 individual single-nucleotide polymorphism (SNP) associations were tested using the combined genome from mothers and neonates (case vs. control) in each population and then pooled via meta-analysis. RESULTS After meta-analysis, there were four SNPs for the outcome of PTD below P ≤ 0.10 and two below P ≤ 0.05. For the additional outcomes of very PTD and PPROM, there were three and four SNPs, respectively, below P ≤ 0.10. CONCLUSION Given the number of tests, no single SNP reached study-wide significance (P = 0.0006). Our study does not support the hypothesis that mitochondrial genetics contributes to the maternal transmission of PTD and related outcomes.
Collapse
Affiliation(s)
| | - Solveig Myking
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Kelli K. Ryckman
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242
| | - Ronny Myhre
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260
| | - Bjarke Feenstra
- Department of Epidemiology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Frank Geller
- Department of Epidemiology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Heather A. Boyd
- Department of Epidemiology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - John R. Shaffer
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260
| | - Qi Zhang
- Department of Biostatistics, University of Washington, Seattle, WA 98195
| | - Ferdouse Begum
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260
| | - David Crosslin
- Department of Biostatistics, University of Washington, Seattle, WA 98195
| | - Kim Doheny
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Elizabeth Pugh
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | | | - Ingrid H.G. Østensen
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Nils-Halvdan Morken
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Per Magnus
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Mary L. Marazita
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260
| | - Bo Jacobsson
- Division of Epidemiology, Norwegian Institute of Public Health, N-0403, Oslo, Norway
| | - Mads Melbye
- Department of Epidemiology, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | | | | | | |
Collapse
|
33
|
Kharrazi M, Pearl M, Yang J, DeLorenze GN, Bean CJ, Callaghan WM, Grant A, Lackritz E, Romero R, Satten GA, Simhan H, Torres AR, Westover JB, Yolken R, Williamson DM. California Very Preterm Birth Study: design and characteristics of the population- and biospecimen bank-based nested case-control study. Paediatr Perinat Epidemiol 2012; 26:250-63. [PMID: 22471684 PMCID: PMC3536480 DOI: 10.1111/j.1365-3016.2011.01252.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Very preterm birth (VPTB) is a leading cause of infant mortality, morbidity and racial disparity in the US. The underlying causes of VPTB are multiple and poorly understood. The California Very Preterm Birth Study was conducted to discover maternal and infant genetic and environmental factors associated with VPTB. This paper describes the study design, population, data and specimen collection, laboratory methods and characteristics of the study population. Using a large, population-based cohort created through record linkage of livebirths delivered from 2000 to 2007 in five counties of southern California, and existing data and banked specimens from statewide prenatal and newborn screening, 1100 VPTB cases and 796 control mother-infant pairs were selected for study (385/200 White, 385/253 Hispanic and 330/343 Black cases/controls, respectively). Medical record abstraction of cases was conducted at over 50 hospitals to identify spontaneous VPTB, improve accuracy of gestational age, obtain relevant clinical data and exclude cases that did not meet eligibility criteria. VPTB was defined as birth at <32 weeks in Whites and Hispanics and <34 weeks in Blacks. Approximately 55% of all VPTBs were spontaneous and 45% had medical indications or other exclusions. Of the spontaneous VPTBs, approximately 41% were reported to have chorioamnionitis. While the current focus of the California Very Preterm Birth Study is to assess the role of candidate genetic markers on spontaneous VPTB, its design enables the pursuit of other research opportunities to identify social, clinical and biological determinants of different types of VPTB with the ultimate aim of reducing infant mortality, morbidity and racial disparities in these health outcomes in the US and elsewhere.
Collapse
Affiliation(s)
- Martin Kharrazi
- Genetic Disease Screening Program, California Department of Public Health, 850 Marina Bay Parkway, Richmond, CA 94804, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Menon R, Dunlop AL, Kramer MR, Fortunato SJ, Hogue CJ. An overview of racial disparities in preterm birth rates: caused by infection or inflammatory response? Acta Obstet Gynecol Scand 2011; 90:1325-31. [PMID: 21615712 PMCID: PMC5573126 DOI: 10.1111/j.1600-0412.2011.01135.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection has been hypothesized to be one of the factors associated with spontaneous preterm birth (PTB) and with the racial disparity in rates of PTB between African American and Caucasian women. However, recent findings refute the generalizability of the role of infection and inflammation. African Americans have an increased incidence of PTB in the setting of intraamniotic infection, periodontal disease, and bacterial vaginosis compared to Caucasians. Herein we report variability in infection- and inflammation-related factors based on race/ethnicity. For African American women, an imbalance in the host proinflammatory response seems to contribute to infection-associated PTB, as evidenced by a greater presence of inflammatory mediators with limited or reduced presence of immune balancing factors. This may be attributed to differences in the genetic variants associated with PTB between African Americans and Caucasians. We argue that infection may not be a cause of racial disparity but in association with other risk factors such as stress, nutritional deficiency, and differences in genetic variations in PTB, pathways and their complex interactions may produce differential inflammatory responses that may contribute to racial disparity.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Epidemiology, Women and Children's Center, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA.
| | | | | | | | | |
Collapse
|
35
|
Chu NF, Lin FH, Chin HC, Hong YJ. Association between interleukin-6 receptor gene variations and atherosclerotic lipid profiles among young adolescents in Taiwan. Lipids Health Dis 2011; 10:136. [PMID: 21835044 PMCID: PMC3180403 DOI: 10.1186/1476-511x-10-136] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/12/2011] [Indexed: 11/17/2022] Open
Abstract
Background To analyze the potential genetic associations between four polymorphisms of interleukin-6 receptor (IL-6R) gene and atherosclerotic lipid profiles among young adolescents in Taiwan. Methods Using data from the Taipei Children Heart Study-II - a cross-sectional survey in 2003. After multi-stage sampling, we selected 418 boys and 441 girls with an average age of 13.1 years. We genotyped the subjects for four IL-6R gene polymorphisms (rs4845617 G/A, rs4845623 A/G, rs8192284 A/C, and rs2229238 C/T) using a TaqMan 5' nuclease assay. Lipid profiles, including total cholesterol (CHOL), triglycerides (TG), high density lipoprotein-cholesterol (HDL-C), low density lipoprotein-cholesterol (LDL-C) were measured using standard methods. We also calculated CHOL/HDL-C ratio, LDL-C/HDL-C ratio, and TG/HDL-C ratio as atherosclerotic indexes. Results IL-6R rs8192284 A/C and rs2229238 C/T variants showed strong associations with high TG (additive model, OR = 1.58, 95%CI: 1.05-2.37; OR = 1.55, 95%CI: 1.04-2.29, respectively), low HDL-C (additive model, OR = 1.57, 95%CI: 1.03-2.39; OR = 1.68, 95%CI: 1.12-2.52, respectively), and high CHOL/HDL-C (additive model, OR = 1.68, 95%CI: 1.08-2.61, OR = 1.82, 95%CI: 1.18-2.79, respectively) in girls. We inferred five common haplotypes using rs4845617 G/A, rs4845623 A/G, and rs2229238 C/T (GAC, GAT, GGC, AAC, and AAT). In girls, the AAT haplotype was associated with a significant risk of high TG, low HDL-C, high CHOL/HDL-C, and abnormal lipid levels (high TG or low HDL-C) when compared with the GAC haplotype (OR range = 3.08-4.40, all p < 0.05). Conclusion The IL-6R rs8192284 A/C and rs2229238 C/T variants are associated with dyslipidemia in girls, but not in boys. The AAT haplotype of the IL-6R gene (rs4845617 G/A, rs4845623 A/G, and rs2229238 C/T) may play an important role in the pathogenesis of dyslipidemia and atherosclerosis in girls.
Collapse
Affiliation(s)
- Nain-Feng Chu
- Department of Community Medicine, Shuang-Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan.
| | | | | | | |
Collapse
|
36
|
Menon R, Fortunato SJ, Milne GL, Brou L, Carnevale C, Sanchez SC, Hubbard L, Lappas M, Drobek CO, Taylor RN. Amniotic fluid eicosanoids in preterm and term births: effects of risk factors for spontaneous preterm labor. Obstet Gynecol 2011; 118:121-134. [PMID: 21691170 PMCID: PMC3286836 DOI: 10.1097/aog.0b013e3182204eaa] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To evaluate amniotic fluid arachidonic acid metabolites using enzymatic and nonenzymatic (lipid peroxidation) pathways in spontaneous preterm birth and term births, and to estimate whether prostanoid concentrations correlate with risk factors (race, cigarette smoking, and microbial invasion of amniotic cavity) associated with preterm birth. METHODS In a case-control study, amniotic fluid was collected at the time of labor or during cesarean delivery. Amniotic fluid samples were subjected to gas chromatography, negative ion chemical ionization, and mass spectrometry for prostaglandin (PG) E2, PGF2α, and PGD2 and for 6-keto-PGF1α (thromboxane 2 and F2-isoprostane). Primary analysis examined differences between prostanoid concentrations in preterm birth (n=133) compared with term births (n=189). Secondary stratified analyses (by race, cigarette smoking, and microbial invasion of amniotic cavity) compared eicosanoid concentrations in three epidemiological risk factors. RESULTS Amniotic fluid F2-isoprostane, PGE2, and PGD2 were significantly higher at term than in preterm birth, whereas PGF2α was higher in preterm birth 6-keto-PGF1α and thromboxane 2 concentrations were not different. Data stratified by race (African American or white) showed no significant disparity among prostanoid concentrations. Regardless of gestational age status, F2-isoprostane was threefold higher in smokers, and other eicosanoids were also higher in smokers compared with nonsmokers. Preterm birth with microbial invasion of amniotic cavity had significantly higher F2-isoprostane compared with preterm birth without microbial invasion of amniotic cavity. CONCLUSION Most amniotic fluid eicosanoid concentrations (F2-isoprostane, PGE2, and PGD2), are higher at term than in preterm births. The only amniotic fluid eicosanoid that is not higher at term is PGF2α.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- The Perinatal Research Center, Nashville, TN
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA
| | | | - Ginger L. Milne
- Division of Clinical Pharmacology, Eicosanoid core laboratory, Vanderbilt University, Nashville, TN
| | - Lina Brou
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Claudine Carnevale
- Department of Biostatistics, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Stephanie C. Sanchez
- Division of Clinical Pharmacology, Eicosanoid core laboratory, Vanderbilt University, Nashville, TN
| | | | - Martha Lappas
- Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | | | - Robert N. Taylor
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA
| |
Collapse
|
37
|
Méhats C, Schmitz T, Marcellin L, Breuiller-Fouché M. [Biochemistry of fetal membranes rupture]. ACTA ACUST UNITED AC 2011; 39:365-9. [PMID: 21602079 DOI: 10.1016/j.gyobfe.2011.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 02/07/2011] [Indexed: 11/26/2022]
Abstract
Fetal membranes, amnion and chorion, line up the amniotic cavity and are essential for its integrity towards normal term of pregnancy. They consist of a pluristratified structure whose composition assures their cohesion and elasticity. They firstly function in retaining the fluctuant amniotic fluid in a half-rigid cavity. Their elastic limit depends on the organization of the extracellular matrix and firstly on the collagen type it contains. The compact layer of the amnion, responsible for the elastic limit, contains mainly type I collagen, organized in lattice; this allows elongation or spreading. Underneath, the spongy layer, principally of collagen III, is organized in a loose mesh, enriched in hydrated proteoglycans, which allows the absorption of the shocks and the sliding of the amnion on the chorion. The cascade of events leading to the membrane rupture displays: (i) membranes distension with elasticity loss, (ii) separation of the chorion from the amnion, (iii) chorion fracture, (iv) amnion distension which produces an hernia, (v) amnion rupture. The rupture mechanism was long thought to be a consequence of uterine contractions. However, the observation before labour of a zone of altered morphology, with biochemical variations (modifications of metalloprotease activity and of proteoglycans, apoptosis...) associated with focal physical weakness in the region overlying the cervix suggests programming of the rupture before parturition. A better understanding of the biochemical mechanisms of membranes rupture will provide new insights into how to anticipate and to intervene in the case of risk of premature rupture.
Collapse
Affiliation(s)
- C Méhats
- Inserm U1016, institut Cochin, département génétique et développement, faculté de médecine Cochin, 24, rue du Faubourg-Saint-Jacques, 75014 Paris, France.
| | | | | | | |
Collapse
|
38
|
Gracie SK, Lyon AW, Kehler HL, Pennell CE, Dolan SM, McNeil DA, Siever JE, McDonald SW, Bocking AD, Lye SJ, Hegadoren KM, Olson DM, Tough SC. All Our Babies Cohort Study: recruitment of a cohort to predict women at risk of preterm birth through the examination of gene expression profiles and the environment. BMC Pregnancy Childbirth 2010; 10:87. [PMID: 21192811 PMCID: PMC3022739 DOI: 10.1186/1471-2393-10-87] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 12/30/2010] [Indexed: 11/10/2022] Open
Abstract
Background Preterm birth is the leading cause of perinatal morbidity and mortality. Risk factors for preterm birth include a personal or familial history of preterm delivery, ethnicity and low socioeconomic status yet the ability to predict preterm delivery before the onset of preterm labour evades clinical practice. Evidence suggests that genetics may play a role in the multi-factorial pathophysiology of preterm birth. The All Our Babies Study is an on-going community based longitudinal cohort study that was designed to establish a cohort of women to investigate how a women's genetics and environment contribute to the pathophysiology of preterm birth. Specifically this study will examine the predictive potential of maternal leukocytes for predicting preterm birth in non-labouring women through the examination of gene expression profiles and gene-environment interactions. Methods/Design Collaborations have been established between clinical lab services, the provincial health service provider and researchers to create an interdisciplinary study design for the All Our Babies Study. A birth cohort of 2000 women has been established to address this research question. Women provide informed consent for blood sample collection, linkage to medical records and complete questionnaires related to prenatal health, service utilization, social support, emotional and physical health, demographics, and breast and infant feeding. Maternal blood samples are collected in PAXgene™ RNA tubes between 18-22 and 28-32 weeks gestation for transcriptomic analyses. Discussion The All Our Babies Study is an example of how investment in clinical-academic-community partnerships can improve research efficiency and accelerate the recruitment and data collection phases of a study. Establishing these partnerships during the study design phase and maintaining these relationships through the duration of the study provides the unique opportunity to investigate the multi-causal factors of preterm birth. The overall All Our Babies Study results can potentially lead to healthier pregnancies, mothers, infants and children.
Collapse
Affiliation(s)
- Sara K Gracie
- Department of Paediatrics, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Paraoxonase 1 gene and glutathione S-transferase μ 1 gene interaction with preterm delivery in Korean women. Am J Obstet Gynecol 2010; 203:569.e1-7. [PMID: 21111108 DOI: 10.1016/j.ajog.2010.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 05/13/2010] [Accepted: 07/21/2010] [Indexed: 11/22/2022]
Abstract
OBJECTIVE This study was undertaken to identify the paraoxonase 1 gene and glutathione S-transferase μ 1 gene interaction for the risk of preterm delivery and to determine the serum paraoxonase activity according to paraoxonase 1 genotypes. STUDY DESIGN This case-control study was performed on 162 gravida women with preterm delivery and 306 controls. Serum paraoxonase activity was measured by a ultraviolet spectrophotometer. Logistic regression, 2-way analysis of variance, and multifactor dimensionality reduction analysis were used. RESULTS Gravida women with the QQ and QR genotype of paraoxonase 1 with high body mass index had 6.19- and 4.41-fold increased risks of preterm delivery. The glutathione S-transferase μ 1 null genotype and the interaction between the paraoxonase 1 genotype and glutathione S-transferase μ 1 null type conferred a risk for preterm delivery. Serum paraoxonase activity was significantly different according to paraoxonase 1 genotypes (P < .0001). CONCLUSION The glutathione S-transferase μ 1 null genotype confers a risk for preterm delivery in Korean gravida women independent of and interactive with the paraoxonase 1 genotype.
Collapse
|
40
|
Lin FH, Chu NF, Lee CH, Hung YJ, Wu DM. Combined effect of C-reactive protein gene SNP +2147 A/G and interleukin-6 receptor gene SNP rs2229238 C/T on anthropometric characteristics among school children in Taiwan. Int J Obes (Lond) 2010; 35:587-94. [PMID: 20856253 DOI: 10.1038/ijo.2010.195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The purpose of this study was to explore the combined effect of the C-reactive protein (CRP) +2147 A/G (rs1205) and interleukin (IL)-6R rs2229238 C/T single-nucleotide polymorphisms (SNPs) on the anthropometric variables of school children in Taiwan. SUBJECTS AND DESIGN Cross-sectional analyses were performed using the data from the Taipei Children Heart Study-II. After multi-stage sampling, we selected 430 boys and 463 girls with an average age of 13.1 years. We genotyped these individuals for the CRP +2147 A/G and IL-6R rs2229238 C/T SNPs using a TaqMan 5' nuclease assay. Anthropometric characteristics, which included body height, body weight (BW), body mass index (BMI), waist circumference (WC), hip circumference (HC), body fat percentage (BF), and waist circumference to height ratio (WHtR), were measured/calculated. RESULTS When considering the CRP +2147 A/G polymorphism, GG genotype boys were heavier and had larger BMI, WC, HC, BF and WHtR than A allele carriers. The odds ratio (OR) of larger WHtR in GG genotype boys was 2.14 (95% CI: 1.09-4.21). For the IL-6R rs2229238 C/T polymorphism, T allele carrier girls had larger WC and WHtR than those carrying the CC genotype. The OR of a larger HC for T allele carrier boys was 2.33 (95% CI: 1.16-4.68). Boys with the GG genotype of CRP +2147 A/G and the CC genotype of IL-6R rs2229238 C/T had higher OR for BW, BMI, WC, HC, BF and WHtR than boys who were carriers of the A allele of CRP +2147 A/G and had the CC genotype of IL-6R rs2229238 C/T (OR range=3.86-8.04, all P<0.05). CONCLUSION Boys who carry the GG genotype of CRP +2147 A/G and the CC genotype of IL-6R rs2229238 C/T have a greater risk of having abnormal BW, BMI, WC, HC, BF and WHtR and of developing obesity than individuals who do not have these genotypes.
Collapse
Affiliation(s)
- F-H Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, No. 161 Sec. 6 Min-Chuan East Road, Nei-Hu, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
41
|
Robertson SA, Christiaens I, Dorian CL, Zaragoza DB, Care AS, Banks AM, Olson DM. Interleukin-6 is an essential determinant of on-time parturition in the mouse. Endocrinology 2010; 151:3996-4006. [PMID: 20610570 DOI: 10.1210/en.2010-0063] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-6 abundance in amniotic fluid and uterine tissues increases in late gestation or with infection-associated preterm labor. A role in regulation of labor onset is suggested by observations that IL-6 increases expression of genes controlling prostaglandin synthesis and signaling in isolated uterine cells, but whether IL-6 is essential for normal parturition is unknown. To evaluate the physiological role of IL-6 in parturition in mice, we investigated the effect of Il6 null mutation on the timing of parturition and expression of genes associated with uterine activation. Il6 null mutant mice delivered 24 h later than wild-type mice, although circulating progesterone fell similarly in both genotypes during the prepartal period. Il6 null mutant mice were also refractory to low doses of lipopolysaccharide sufficient to induce preterm delivery in wild-type mice. The characteristic late-gestation elevation in uterine expression of Oxtr mRNA encoding oxytocin receptor, and peripartal increases in Ptgfr and Ptgs2 mRNAs regulating prostaglandin synthesis and signaling were delayed by 24 h in Il6 null mutant mice. Conversely, Ptger4 mRNA encoding the prostaglandin E receptor-4 was abnormally elevated in late-gestation in Il6 null mutant mice. Administration of recombinant IL-6 from d 11.5 postcoitum until term restored the normal timing of delivery and normalized Ptger4 mRNA expression in late gestation. We conclude that IL-6 has a key role in controlling the progression of events culminating in parturition and that it acts downstream of luteolysis in the uterus to regulate genes involved in the prostaglandin-mediated uterine activation cascade.
Collapse
Affiliation(s)
- Sarah A Robertson
- Robinson Institute, School of Pediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.
| | | | | | | | | | | | | |
Collapse
|
42
|
Pander J, Wessels JAM, Mathijssen RHJ, Gelderblom H, Guchelaar HJ. Pharmacogenetics of tomorrow: the 1 + 1 = 3 principle. Pharmacogenomics 2010; 11:1011-7. [DOI: 10.2217/pgs.10.87] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Disappointing results from replicating pharmacogenetic association studies have prompted the search for novel statistical techniques to analyze the data, while taking into account the biological complexity underlying drug response. Two of these techniques – multifactor dimensionality reduction and classification and regression tree analysis – will probably be applied in increasing numbers of future pharmacogenetic studies. In this article, we describe the concepts underlying both techniques and illustrate their application in a recent pharmacogenetic study.
Collapse
Affiliation(s)
- Jan Pander
- Leiden University Medical Center, Department of Clinical Pharmacy & Toxicology, PO Box 9600, 2300RC Leiden, The Netherlands
| | - Judith AM Wessels
- Leiden University Medical Center, Department of Clinical Pharmacy & Toxicology, PO Box 9600, 2300RC Leiden, The Netherlands
| | - Ron HJ Mathijssen
- Erasmus Medical Center Daniel den Hoed Cancer Center, Department of Medical Oncology, PO Box 5201, 3008AE Rotterdam, The Netherlands
| | - Hans Gelderblom
- Leiden University Medical Center, Department of Clinical Oncology, PO Box 9600, 2300RC Leiden, The Netherlands
| | | |
Collapse
|
43
|
Romero R, Velez DR, Kusanovic JP, Hassan SS, Mazaki-Tovi S, Vaisbuch E, Kim CJ, Chaiworapongsa T, Pearce B, Friel LA, Bartlett J, Anant MK, Salisbury BA, Vovis GF, Lee MS, Gomez R, Behnke E, Oyarzun E, Tromp G, Williams SM, Menon R. Identification of fetal and maternal single nucleotide polymorphisms in candidate genes that predispose to spontaneous preterm labor with intact membranes. Am J Obstet Gynecol 2010; 202:431.e1-34. [PMID: 20452482 PMCID: PMC3604889 DOI: 10.1016/j.ajog.2010.03.026] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 01/31/2010] [Accepted: 03/15/2010] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The purpose of this study was to determine whether maternal/fetal single nucleotide polymorphisms (SNPs) in candidate genes are associated with spontaneous preterm labor/delivery. STUDY DESIGN A genetic association study was conducted in 223 mothers and 179 fetuses (preterm labor with intact membranes who delivered <37 weeks of gestation [preterm birth (PTB)]), and 599 mothers and 628 fetuses (normal pregnancy); 190 candidate genes and 775 SNPs were studied. Single locus/haplotype association analyses were performed; the false discovery rate was used to correct for multiple testing. RESULTS The strongest single locus associations with PTB were interleukin-6 receptor 1 (fetus; P=.000148) and tissue inhibitor of metalloproteinase 2 (mother; P=.000197), which remained significant after correction for multiple comparisons. Global haplotype analysis indicated an association between a fetal DNA variant in insulin-like growth factor F2 and maternal alpha 3 type IV collagen isoform 1 (global, P=.004 and .007, respectively). CONCLUSION An SNP involved in controlling fetal inflammation (interleukin-6 receptor 1) and DNA variants in maternal genes encoding for proteins involved in extracellular matrix metabolism approximately doubled the risk of PTB.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Digna R. Velez
- Department of Human Genetics, Dr. John T. Macdonald Foundation, and John P. Hussman Institute of Human Genomics and University of Miami, Miami, Florida, USA
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Brad Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Lara A. Friel
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jacquelaine Bartlett
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | - Min Seob Lee
- Genaissance Pharmaceuticals, Inc., New Haven, Connecticut, USA
| | - Ricardo Gomez
- CEDIP (Center for Perinatal Diagnosis and Research), Department of Obstetrics and Gynecology, Sotero del Rio Hospital, Santiago, Chile
- Department of Obstetrics and Gynecology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ernesto Behnke
- CEDIP (Center for Perinatal Diagnosis and Research), Department of Obstetrics and Gynecology, Sotero del Rio Hospital, Santiago, Chile
| | - Enrique Oyarzun
- Department of Obstetrics and Gynecology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Gerard Tromp
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Scott M. Williams
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ramkumar Menon
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
44
|
Association of combined maternal-fetal TNF-alpha gene G308A genotypes with preterm delivery: a gene-gene interaction study. J Biomed Biotechnol 2010; 2010:396184. [PMID: 20224765 PMCID: PMC2836175 DOI: 10.1155/2010/396184] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 11/05/2009] [Accepted: 01/28/2010] [Indexed: 11/24/2022] Open
Abstract
Preterm delivery (PTD) is a complicated perinatal adverse event. We were interested in association of G308A polymorphism in tumor necrosis factor-α (TNF-α) gene with PTD; so we conducted a genetic epidemiology study in Anqing City, Anhui Province, China. Case families and control families were all collected between July 1999 and June 2002. To control potential population stratification as we could, all eligible subjects were ethnic Han Chinese. 250 case families and 247 control families were included in data analysis. A hybrid design which combines case-parent triads and control parents was employed, to test maternal-fetal genotype (MFG) incompatibility. The method is based on a log-linear modeling approach. In summary, we found that when the mother's or child's genotype was G/A, there was a reduced risk of PTD; however when the mother's or child's genotype was genotype A/A, there was a relatively higher risk of PTD. Combined maternal-fetal genotype GA/GA showed the most reduced risk of PTD. Comparison of the LRTs showed that the model with maternal-fetal genotype effects fits significantly better than the model with only maternal and fetal genotype main effects (log-likelihood = −719.4, P = .023, significant at 0.05 level). That means that the combined maternal-fetal genotype incompatibility was significantly associated with PTD. The model with maternal-fetal genotype effects can be considered a gene-gene interaction model. We claim that both maternal effects and fetal effects should be considered together while investigating genetic factors of certain perinatal diseases.
Collapse
|
45
|
Srinivasan U, Misra D, Marazita ML, Foxman B. Vaginal and oral microbes, host genotype and preterm birth. Med Hypotheses 2009; 73:963-75. [PMID: 19942083 PMCID: PMC4026093 DOI: 10.1016/j.mehy.2009.06.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 06/03/2009] [Accepted: 06/06/2009] [Indexed: 01/08/2023]
Abstract
Preterm birth (PTB) is a leading cause of infant mortality and morbidity in the US and across the globe. Infection and associated inflammation are important initiators for PTB pathways; an estimated 40% of PTBs are attributed to amniochorionic-decidual or systemic inflammation. Historically, intrauterine infections have been implicated in PTB; recent evidence suggests that infections remote from the fetal site may also be causative. There is strong epidemiological evidence that bacterial vaginosis and periodontitis--two syndromes characterized by perturbations in the normal vaginal and oral bacterial microflora, respectively--are linked to infection-associated PTB. Oral and vaginal environments are similar in their bacterial microbiology; identical bacterial species have been independently isolated in periodontitis and bacterial vaginosis. Periodontitis and bacterial vaginosis also share many behavioral and sociodemographic risk factors suggesting a possible common pathophysiology. Genetic polymorphisms in host inflammatory responses to infection are shared between bacterial vaginosis, periodontitis and PTB, suggesting common mechanisms through which host genotype modify the effect of abnormal bacterial colonization on preterm birth. We review the state of knowledge regarding the risk of PTB attributable to perturbations in bacterial flora in oral and vaginal sites and the role of host genetics in modifying the risk of infection-related PTB. We posit that bacterial species that are common in perturbed vaginal and oral sites are associated with PTB through their interaction with the host immune system.
Collapse
Affiliation(s)
- Usha Srinivasan
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | |
Collapse
|
46
|
Plunkett J, Feitosa MF, Trusgnich M, Wangler MF, Palomar L, Kistka ZAF, DeFranco EA, Shen TT, Stormo AE, Puttonen H, Hallman M, Haataja R, Luukkonen A, Fellman V, Peltonen L, Palotie A, Daw EW, An P, Teramo K, Borecki I, Muglia LJ. Mother's genome or maternally-inherited genes acting in the fetus influence gestational age in familial preterm birth. Hum Hered 2009; 68:209-19. [PMID: 19521103 PMCID: PMC2869074 DOI: 10.1159/000224641] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Accepted: 02/26/2009] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE While multiple lines of evidence suggest the importance of genetic contributors to risk of preterm birth, the nature of the genetic component has not been identified. We perform segregation analyses to identify the best fitting genetic model for gestational age, a quantitative proxy for preterm birth. METHODS Because either mother or infant can be considered the proband from a preterm delivery and there is evidence to suggest that genetic factors in either one or both may influence the trait, we performed segregation analysis for gestational age either attributed to the infant (infant's gestational age), or the mother (by averaging the gestational ages at which her children were delivered), using 96 multiplex preterm families. RESULTS These data lend further support to a genetic component contributing to birth timing since sporadic (i.e. no familial resemblance) and nontransmission (i.e. environmental factors alone contribute to gestational age) models are strongly rejected. Analyses of gestational age attributed to the infant support a model in which mother's genome and/or maternally-inherited genes acting in the fetus are largely responsible for birth timing, with a smaller contribution from the paternally-inherited alleles in the fetal genome. CONCLUSION Our findings suggest that genetic influences on birth timing are important and likely complex.
Collapse
Affiliation(s)
- Jevon Plunkett
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
- Human and Statistical Genetics Program, Washington University School of Medicine, St. Louis, Miss., USA
| | - Mary F. Feitosa
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Miss., USA
| | - Michelle Trusgnich
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Miss., USA
| | - Michael F. Wangler
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Lisanne Palomar
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Zachary A.-F. Kistka
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Emily A. DeFranco
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Tammy T. Shen
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Adrienne E.D. Stormo
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
| | - Hilkka Puttonen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | - Mikko Hallman
- Department of Pediatrics, University of Oulu, Oulu, Finland
| | - Ritva Haataja
- Department of Pediatrics, University of Oulu, Oulu, Finland
| | - Aino Luukkonen
- Department of Pediatrics, University of Oulu, Oulu, Finland
| | - Vineta Fellman
- Department of Pediatrics, University of Helsinki, Helsinki, Finland
- Department of Pediatrics, Lund University, Lund, Sweden
| | - Leena Peltonen
- Biomedicum Helsinki Research Program in Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Clinical Chemistry, Helsinki University Central Hospital, Helsinki, Finland
- Department of Molecular Medicine, National Public Health Institute, Helsinki, Finland
- The Broad Institute of MIT and Harvard, Cambridge, Mass., USA
- Wellcome Trust Sanger Institute, Cambridge, UK
| | - Aarno Palotie
- Biomedicum Helsinki Research Program in Molecular Medicine, University of Helsinki, Helsinki, Finland
- The Finnish Genome Center, University of Helsinki, Helsinki, Finland
- Department of Pediatrics, University of Oulu, Oulu, Finland
- The Broad Institute of MIT and Harvard, Cambridge, Mass., USA
- Wellcome Trust Sanger Institute, Cambridge, UK
| | - E. Warwick Daw
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Miss., USA
| | - Ping An
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Miss., USA
| | - Kari Teramo
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | - Ingrid Borecki
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
- Human and Statistical Genetics Program, Washington University School of Medicine, St. Louis, Miss., USA
| | - Louis J. Muglia
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Miss., USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Miss., USA
- Center for Preterm Birth Research, Washington University School of Medicine, St. Louis, Miss., USA
- Human and Statistical Genetics Program, Washington University School of Medicine, St. Louis, Miss., USA
| |
Collapse
|
47
|
Moura E, Mattar R, de Souza E, Torloni MR, Gonçalves-Primo A, Daher S. Inflammatory cytokine gene polymorphisms and spontaneous preterm birth. J Reprod Immunol 2009; 80:115-21. [DOI: 10.1016/j.jri.2008.11.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2008] [Revised: 10/22/2008] [Accepted: 11/21/2008] [Indexed: 11/29/2022]
|
48
|
Velez DR, Fortunato S, Thorsen P, Lombardi SJ, Williams SM, Menon R. Spontaneous preterm birth in African Americans is associated with infection and inflammatory response gene variants. Am J Obstet Gynecol 2009; 200:209.e1-27. [PMID: 19019335 DOI: 10.1016/j.ajog.2008.08.051] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 06/20/2008] [Accepted: 08/20/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The objective of the study was to study the genetic risk factors of spontaneous preterm birth (PTB) in African Americans. STUDY DESIGN Case-control analyses were performed using maternal and fetal deoxyribonucleic acid from 279 African American birth events (82 PTB and 197 term) and 1432 single-nucleotide polymorphisms from 130 candidate genes. Single-locus association and haplotype analyses were performed. RESULTS The most significant associations were in the maternal interleukin (IL)-15 (rs10833, allele P = 2.91 x 10(-4), genotype P = 2.00 x 10(-3)) gene and the fetal IL-2 receptor B (IL-2RB) (rs84460, allele P = 1.37 x 10(-4), genotype P = 6.29 x 10(-4)) gene. The best models for these markers were additive (rs10833, odds ratio [OR], 0.30; 95% confidence interval [CI], 0.14-0.62; P = 1.0 x 10(-3); rs84460, OR, 2.32; 95% CI, 1.47-3.67; P < 1.0 x 10(-3)). The largest number of significant associations was found in genes related to infection and inflammation. There were overall a larger number of significant associations in infants than in mothers. CONCLUSION These results support a strong role for genes involved in infection and inflammation in the pathogenesis of PTB, particularly IL-12 and IL-12RB, and indicate that in African Americans there may be complementarity of maternal and fetal genetic risks for PTB.
Collapse
|
49
|
Himes KP, Simhan HN. Genetic susceptibility to infection-mediated preterm birth. Infect Dis Clin North Am 2009; 22:741-753. [PMID: 18954761 DOI: 10.1016/j.idc.2008.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Preterm birth is a heterogeneous disorder that is responsible for significant neonatal morbidity and mortality. Intrauterine infection is implicated in a significant proportion of preterm birth-particularly in early gestation. Epidemiologic evidence supports a genetic component to infection overall and preterm birth in particular. Furthermore, a number of studies examining genes involved in pathogen recognition and our response to pathogens suggest a genetic susceptibility to infection-mediated preterm birth. On the whole, however, these studies have been difficult to replicate and explain only a small portion of the phenotypic variation in preterm birth. Given this, methodological considerations are emphasized to improve our understanding of the genetic susceptibility to infection-mediated preterm birth.
Collapse
Affiliation(s)
- Katherine P Himes
- Division of Maternal Fetal Medicine, University of Pittsburgh Medical Center, Magee-Womens Hospital, 300 Halket Street, Pittsburgh, PA 15213, USA
| | - Hyagriv N Simhan
- Division of Maternal Fetal Medicine, University of Pittsburgh Medical Center, Magee-Womens Hospital, 300 Halket Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
50
|
Shen TT, DeFranco EA, Stamilio DM, Chang JJ, Muglia LJ. A population-based study of race-specific risk for placental abruption. BMC Pregnancy Childbirth 2008; 8:43. [PMID: 18789147 PMCID: PMC2546363 DOI: 10.1186/1471-2393-8-43] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 09/12/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Efforts to elucidate risk factors for placental abruption are imperative due to the severity of complications it produces for both mother and fetus, and its contribution to preterm birth. Ethnicity-based differences in risk of placental abruption and preterm birth have been reported. We tested the hypotheses that race, after adjusting for other factors, is associated with the risk of placental abruption at specific gestational ages, and that there is a greater contribution of placental abruption to the increased risk of preterm birth in Black mothers, compared to White mothers. METHODS We conducted a population-based cohort study using the Missouri Department of Health's maternally-linked database of all births in Missouri (1989-1997) to assess racial effects on placental abruption and the contribution of placental abruption to preterm birth, at different gestational age categories (n = 664,303). RESULTS Among 108,806 births to Black mothers and 555,497 births to White mothers, 1.02% (95% CI 0.96-1.08) of Black births were complicated by placental abruption, compared to 0.71% (95% CI 0.69-0.73) of White births (aOR 1.32, 95% CI 1.22-1.43). The magnitude of risk of placental abruption for Black mothers, compared to White mothers, increased with younger gestational age categories. The risk of placental abruption resulting in term and extreme preterm births (< 28 weeks) was higher for Black mothers (aOR 1.15, 95% CI 1.02-1.29 and aOR 1.98, 95% CI 1.58-2.48, respectively). Compared to White women delivering in the same gestational age category, there were a significantly higher proportion of placental abruption in Black mothers who delivered at term, and a significantly lower proportion of placental abruption in Black mothers who delivered in all preterm categories (p < 0.05). CONCLUSION Black women have an increased risk of placental abruption compared to White women, even when controlling for known coexisting risk factors. This risk increase is greatest at the earliest preterm gestational ages when outcomes are the poorest. The relative contribution of placental abruption to term births was greater in Black women, whereas the relative contribution of placental abruption to preterm birth was greater in White women.
Collapse
Affiliation(s)
- Tammy T Shen
- Department of Pediatrics, Washington University in St, Louis, St, Louis, Missouri, USA.
| | | | | | | | | |
Collapse
|