1
|
Chen Y, Wang C, Wang Y, Peng X, Li R, Pan F. Association of fetal fraction and cell-free fetal DNA with adverse pregnancy outcomes: A systematic review. Int J Gynaecol Obstet 2024; 167:479-490. [PMID: 38766692 DOI: 10.1002/ijgo.15612] [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: 09/19/2023] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Adverse pregnancy outcomes, which can be caused by multiple factors, present a significant threat to the health of mothers and their babies. Cell-free fetal DNA (cffDNA) from placental trophoblast cells might be able to reflect placental and fetal status. Previous studies have yielded controversial results regarding the association of FF or cffDNA with various adverse pregnancy outcomes. A previous study has attempted to systematically assess the association between low fetal fraction (FF) and adverse pregnancy outcomes, but it failed to perform quantitative analyses due to the few studies included. In the present study, we attempted to quantitatively assess the association of FF (or cffDNA) with adverse pregnancy outcomes and further analyze the causes of heterogeneity. OBJECTIVES To investigate the association of high/low FF or cffDNA with adverse pregnancy outcomes. SEARCH STRATEGY We searched the databases of PubMed, Embase, Cochrane, and Web of Science from January 1, 1990, to June 15, 2022 in this meta-analysis. SELECTION CRITERIA Studies on the relationships of adverse pregnancy outcomes in women with FF or cell free DNA were included. Non-English literature was excluded. DATA COLLECTION AND ANALYSIS Data about pregnancy outcomes and cell free DNA were extracted and meta-analyzed. Subgroup analysis was performed by different outcomes. MAIN RESULTS There were 11 studies included involving 8280 participants. No significant heterogeneity was observed among the studies (I2 = 27%, 25%), and a fixed-effect model was used for weighted quantitative analysis. The results revealed that the FF or cffDNA during pregnancy was significantly associated with adverse pregnancy outcomes in pregnant women (OR = 1.57, 95% CI [1.24, 1.99], P = 0.233). The overall incidence of the maternal adverse outcomes was 8% (95% CI: 5-13). Subgroup analysis of different outcomes showed an evident association between low FF or cffDNA and hypertensive disorders of pregnancy (HDP) (OR = 1.76, 95% CI [1.36, 2.27], P = 0.581). There was no evidence that the occurrence of spontaneous preterm birth (sPTB) and placental abnormality was associated with FF or cffDNA. No association was observed between low FF or cffDNA during pregnancy and adverse outcomes in fetuses (OR = 1.39, 95% CI [0.99, 1.94], P = 0.242). The overall incidence of adverse outcomes in fetuses was 8% (95% CI: 6-11). There were controversies over the association between high FF or cffDNA and HDP, and sPTB and small for gestational age infant, among different studies. CONCLUSIONS Pregnant women with low FF or cffDNA during the first or second trimester of pregnancy have an overall increased risk of adverse pregnancy outcomes, especially HDP. However, the association between FF and various pregnancy outcomes needs to be further explored by more prospective studies.
Collapse
Affiliation(s)
- Yixin Chen
- Department of Blood Transfusion, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| | - Chun Wang
- Department of Blood Transfusion, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| | - Yonghong Wang
- Department of Clinical Laboratory, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| | - Xin Peng
- Department of Clinical Laboratory, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| | - Rujing Li
- Department of Clinical Laboratory, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| | - Feng Pan
- Department of Clinical Laboratory, Qianjiang Central Hospital, Qianjiang Hospital Affiliated to Chongqing University, Chongqing, P.R. China
| |
Collapse
|
2
|
Cochrane E, Doctor T, Tavella N, Dubois B, McCarthy L, Stratis C, Stoffels G, Bianco A, DeBolt C. Examining the predictive value of fetal fraction on hypertensive disorders of pregnancy. Eur J Obstet Gynecol Reprod Biol 2024; 301:77-81. [PMID: 39106618 DOI: 10.1016/j.ejogrb.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Cell-free fetal DNA (cffDNA) screening is routinely performed in pregnancy. Abnormal fetal fraction has been associated with adverse pregnancy outcomes, including hypertensive disorders of pregnancy, which are associated with severe maternal and neonatal morbidity and mortality. OBJECTIVE This study examined whether abnormal fetal fraction, defined in this study as fetal fraction either <6 or >15 on the basis of restricted-cubic-spline-plot within our study population, was associated with HDP in a retrospective sample, as well as whether fetal fraction improves the prediction of hypertensive disorders of pregnancy (HDP). We hypothesized that abnormal fetal fraction would be associated with HDP and that adding fetal fraction to a model would significantly improve its strength to predict HDP. STUDY DESIGN This was a retrospective cohort study of 729 patients delivering singleton, non-anomalous pregnancies with conclusive cffDNA screening. The primary outcome was HDP. Logistic regression models tested associations between fetal fraction and HDP. We evaluated the impact of including fetal fraction on the prediction of hypertensive disorders of pregnancy (HDP) by comparing the area under the receiver operating characteristic (ROC) curve (AUC) between predictive models with and without fetal fraction. RESULTS Among the study sample, there was an HDP rate of 11.5 %. Abnormal fetal fraction was defined as <6 % percentile and >15 %, HDP incidence was significantly higher in patients with fetal fraction <6 % compared to patients with fetal fraction in normal range (fetal fraction 6-15 %) (19.5 % vs 10.7 %, p = 0.006 on post hoc comparison). Model 1 had one predictor (fetal fraction) with an AUC of 0.59, Model 2 had three predictors (BMI, nulliparity, history of HDP) with an AUC of 0.71, and Model 3 had four predictors (BMI, nulliparity, history of HDP, and fetal fraction) with an AUC of 0.73. Models 2 and 3 were not significantly different (p = 0.18). CONCLUSIONS More patients who developed HDP had low fetal fraction and fewer patients who developed HDP had high fetal fraction compared to those patients who did not develop HDP. Based on results from multivariable regression models, we cannot conclude that fetal fraction improves HDP prediction. However, developing standardized values for abnormal fetal fraction may be clinically useful.
Collapse
Affiliation(s)
| | - Tahera Doctor
- Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | | | | | - Lily McCarthy
- Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | | | | | - Angela Bianco
- Icahn School of Medicine at Mount Sinai, NY, NY, USA
| | | |
Collapse
|
3
|
Golbasi H, Bayraktar B, Golbasi C, Omeroglu I, Adiyaman D, Alkan KO, Ozdemir TR, Ozer OK, Ozyilmaz B, Ekin A. Association between fetal fraction of cell-free DNA and adverse pregnancy outcomes. Arch Gynecol Obstet 2024; 310:1037-1048. [PMID: 38441603 DOI: 10.1007/s00404-024-07443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/16/2024] [Indexed: 07/19/2024]
Abstract
PURPOSE To determine the association between fetal fraction (FF) levels in cell-free fetal DNA (cffDNA) testing and adverse pregnancy outcomes. METHODS This retrospective cohort study, conducted at a single center, involved 2063 pregnant women with normal 1st and 2nd trimester non-invasive prenatal test (NIPT) results between 2016 and 2021. Pregnancy outcomes were examined by determining the < 4% and < 5th percentile (3.6%) cut-off values for low fetal fraction (LFF). Pregnancy outcomes were also examined by dividing the FF into population-based quartiles. Adverse pregnancy outcomes were pregnancy-induced hypertensive diseases (PIHD), gestational diabetes mellitus (GDM), spontaneous preterm birth (PTB), intrahepatic cholestasis of pregnancy (ICP), small for gestational age (SGA), large for gestational age (LGA), low birth weight (LBW), macrosomia, and 1st and 5th minutes low APGAR scores (< 7). RESULTS PIHD was significantly higher in LFF (< 4% and < 5th percentile) cases (p = 0.015 and p < 0.001, respectively). However, in population-based quartiles of FF, PIHD did not differ significantly between groups. Composite adverse maternal outcomes were significantly higher in the FF < 4% group (p = 0.042). When analyzes were adjusted for maternal age, BMI, and gestational age at NIPT, significance was maintained at < 4%, < 5th percentile LFF for PIHD, and < 4% LFF for composite adverse maternal outcomes. However, there was no significant relationship between LFF with GDM, ICP and PTB. Additionally, there was no significant association between low APGAR scores, SGA, LGA, LBW, macrosomia, and LFF concerning neonatal outcomes. CONCLUSION Our study showed that LFF in pregnant women with normal NIPT results may be a predictor of subsequent PIHD.
Collapse
Affiliation(s)
- Hakan Golbasi
- Division of Perinatology, Department of Obstetrics and Gynecology, Bakircay University Cigli Education and Research Hospital, Yeni District., 8780/1 St., Cigli, Izmir, Izmir, Türkiye.
- Division of Perinatology, Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye.
| | - Burak Bayraktar
- Division of Perinatology, Department of Obstetrics and Gynecology, University of Health Sciences Ankara Etlik City Hospital, Ankara, Türkiye
- Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Ceren Golbasi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Izmir Tinaztepe University, Izmir, Türkiye
| | - Ibrahim Omeroglu
- Division of Perinatology, Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Duygu Adiyaman
- Division of Perinatology, Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
- Department of Obstetrics and Gynecology, Ulm University Hospital, Ulm, Germany
| | - Kaan Okan Alkan
- Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Taha Resid Ozdemir
- Department of Genetics, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Ozge Kaya Ozer
- Department of Genetics, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Berk Ozyilmaz
- Department of Genetics, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| | - Atalay Ekin
- Division of Perinatology, Department of Obstetrics and Gynecology, University of Health Sciences Tepecik Training and Research Hospital, Izmir, Türkiye
| |
Collapse
|
4
|
Dangat K, Wadhwani N, Randhir K, Poddar A, Gupte S, Wagh G, Lalwani S, Joshi S. Longitudinal profile of high-sensitivity C-reactive protein in women with pre-eclampsia. Am J Reprod Immunol 2023; 90:e13741. [PMID: 37491921 DOI: 10.1111/aji.13741] [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: 04/13/2023] [Revised: 05/26/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM C-reactive protein (CRP) is a marker for inflammation and its role as a possible biomarker for an early prediction of pre-eclampsia (PE) is unclear. The present study investigates the levels of high-sensitivity CRP (hs-CRP) longitudinally across pregnancy in women with PE and compares them to women without PE (non-PE). METHOD OF STUDY A total of 324 pregnant women [216 non-PE and 108 PE women] were included in this study. Maternal blood was taken at four different intervals (V1 = 11-14 weeks, V2 = 18-22 weeks, V3 = 26-28 weeks, and V4 = at delivery). RESULTS Maternal serum hs-CRP levels were higher at V1, V2, and V3 (p < .05 for all) in the PE group compared to the non-PE group. The hs-CRP levels were associated with maternal blood pressure throughout pregnancy. Maternal hs-CRP levels did not differ among early and late onset PE. Higher maternal hs-CRP levels were associated with the increased risk of PE in unadjusted model in early pregnancy. However, there was no significance after adjusting for confounding factors. CONCLUSIONS Our findings suggest although the levels of hs-CRP were higher in PE in early pregnancy, they are not associated with an increased risk of PE.
Collapse
Affiliation(s)
- Kamini Dangat
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Nisha Wadhwani
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Karuna Randhir
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Anupam Poddar
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Sanjay Gupte
- Gupte Hospital and Research Centre, Pune, Maharashtra, India
| | - Girija Wagh
- Department of Obstetrics and Gynaecology, Bharati Medical College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Sanjay Lalwani
- Department of Pediatrics, Bharati Medical College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| |
Collapse
|
5
|
Suresh S, Freedman A, Plunkett BA, Ernst LM. Low first-trimester fetal fraction is associated with chronic inflammation in the placenta. Am J Obstet Gynecol MFM 2023; 5:101012. [PMID: 37169285 DOI: 10.1016/j.ajogmf.2023.101012] [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: 03/23/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Some data suggest an association between abnormal fetal fraction on noninvasive prenatal screening and adverse pregnancy outcomes, including low birthweight, preeclampsia, and preterm birth in the absence of aneuploidy. These findings suggest that abnormal fetal fraction may be associated with placental pathologic processes in early gestation. OBJECTIVE This study aimed to determine the independent association of fetal fraction on genetic noninvasive prenatal screening with histologic placental types. STUDY DESIGN This was a retrospective cohort study at a single institution in the period between January 2017 and March 2021, including live births at ≥24 weeks for which noninvasive prenatal screening was performed and placental pathology results were available. Results were stratified by trimester of noninvasive prenatal screening. Clinical characteristics were compared by quartile of fetal fraction using chi-square tests. Linear regression was used to model continuous fetal fraction as a function of 3 histologic types representing chronic placental injury-chronic inflammation, maternal vascular malperfusion, and fetal vascular malperfusion. Inverse probability weighting was used to account for selection bias in characteristics of patients with placental pathology examination. RESULTS A total of 1374 patients had noninvasive prenatal screening in the first trimester and 262 in the second trimester. Preterm birth and hypertensive disorders of pregnancy were most common in the lowest quartile of fetal fraction. Chronic inflammation was associated with a 0.56 percentage point reduction in fetal fraction (95% confidence interval, -0.95 to -0.16), and maternal vascular malperfusion was associated with a 0.48 percentage point reduction in fetal fraction (95% confidence interval, -0.91 to -0.04) in adjusted models. The association with maternal vascular malperfusion was no longer statistically significant after accounting for selection bias in placentas sent for pathologic examination. Second-trimester fetal fraction was not associated with placental pathology. CONCLUSION Chronic inflammation is associated with lower first-trimester fetal fraction even after accounting for selection bias. Higher fetal fraction in the second trimester was associated with fetal vascular pathology, although this association was no longer statistically significant after inverse probability weighting to account for selection bias. First-trimester fetal fraction may be a biomarker of adverse outcomes associated with chronic inflammation.
Collapse
Affiliation(s)
- Sunitha Suresh
- Division of Maternal-Fetal Medicine, NorthShore University HealthSystem, University of Chicago, Evanston, IL (Dr Suresh and Dr. Plunkett).
| | - Alexa Freedman
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL (Dr Freedman)
| | - Beth A Plunkett
- Division of Maternal-Fetal Medicine, NorthShore University HealthSystem, University of Chicago, Evanston, IL (Dr Suresh and Dr. Plunkett)
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, Evanston, IL (Dr Ernst); Department of Pathology, University of Chicago Pritzker School of Medicine, Chicago, IL (Dr Ernst)
| |
Collapse
|
6
|
Damour G, Baumer K, Legardeur H, Hall D. Early noninvasive prenatal paternity testing by targeted fetal DNA analysis. Sci Rep 2023; 13:12139. [PMID: 37495669 PMCID: PMC10372148 DOI: 10.1038/s41598-023-39367-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/21/2023] [Indexed: 07/28/2023] Open
Abstract
Today the challenge in paternity testing is to provide an accurate noninvasive assay that can be performed early during pregnancy. This requires the use of novel analytical methods capable of detecting the low fraction of circulating fetal DNA in maternal blood. We previously showed that forensic compound markers such as deletion/insertion polymorphisms-short tandem repeats (DIP-STR) can efficiently resolve complex mixed biological evidence including the target analysis of paternally inherited fetal alleles. In this study, we describe for the first time the validation of this type of markers in the first trimester of pregnancies, in addition to defining the statistical framework to evaluate paternity. To do so, we studied 47 DIP-STRs in 87 cases, with blood samples collected throughout gestation starting from the seven weeks of amenorrhea. Fetal DNA detection in the first trimester shows a false negative rate as low as 6%. The combined paternity index (CPI) results indicate that seven markers with fully informative genotypes are sufficient to determine the paternity. This study demonstrates that DIP-STR markers can be used from early pregnancy and that a small set of markers (about 40) is sufficient to address the question of paternity. The novel method offers substantial improvements over similar approaches in terms of reduced number of markers, lower costs and increased accuracy.
Collapse
Affiliation(s)
- Géraldine Damour
- Unité de Génétique Forensique, Centre Universitaire Romand de Médecine Légale, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Ch. de Vulliette 4, 1000, Lausanne, Switzerland
| | - Karine Baumer
- Unité de Génétique Forensique, Centre Universitaire Romand de Médecine Légale, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Ch. de Vulliette 4, 1000, Lausanne, Switzerland
| | - Hélène Legardeur
- Woman-Mother-Child Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Diana Hall
- Unité de Génétique Forensique, Centre Universitaire Romand de Médecine Légale, Centre Hospitalier Universitaire Vaudois et Université de Lausanne, Ch. de Vulliette 4, 1000, Lausanne, Switzerland.
| |
Collapse
|
7
|
Farias-Jofre M, Romero R, Xu Y, Levenson D, Tao L, Kanninen T, Galaz J, Arenas-Hernandez M, Liu Z, Miller D, Bhatti G, Seyerle M, Tarca AL, Gomez-Lopez N. Differential immunophenotype of circulating monocytes from pregnant women in response to viral ligands. BMC Pregnancy Childbirth 2023; 23:323. [PMID: 37149573 PMCID: PMC10163583 DOI: 10.1186/s12884-023-05562-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/30/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Viral infections during pregnancy can have deleterious effects on mothers and their offspring. Monocytes participate in the maternal host defense against invading viruses; however, whether pregnancy alters monocyte responses is still under investigation. Herein, we undertook a comprehensive in vitro study of peripheral monocytes to characterize the differences in phenotype and interferon release driven by viral ligands between pregnant and non-pregnant women. METHODS Peripheral blood was collected from third-trimester pregnant (n = 20) or non-pregnant (n = 20, controls) women. Peripheral blood mononuclear cells were isolated and exposed to R848 (TLR7/TLR8 agonist), Gardiquimod (TLR7 agonist), Poly(I:C) (HMW) VacciGrade™ (TLR3 agonist), Poly(I:C) (HMW) LyoVec™ (RIG-I/MDA-5 agonist), or ODN2216 (TLR9 agonist) for 24 h. Cells and supernatants were collected for monocyte phenotyping and immunoassays to detect specific interferons, respectively. RESULTS The proportions of classical (CD14hiCD16-), intermediate (CD14hiCD16+), non-classical (CD14loCD16+), and CD14loCD16- monocytes were differentially affected between pregnant and non-pregnant women in response to TLR3 stimulation. The proportions of pregnancy-derived monocytes expressing adhesion molecules (Basigin and PSGL-1) or the chemokine receptors CCR5 and CCR2 were diminished in response to TLR7/TLR8 stimulation, while the proportions of CCR5- monocytes were increased. Such differences were found to be primarily driven by TLR8 signaling, rather than TLR7. Moreover, the proportions of monocytes expressing the chemokine receptor CXCR1 were increased during pregnancy in response to poly(I:C) stimulation through TLR3, but not RIG-I/MDA-5. By contrast, pregnancy-specific changes in the monocyte response to TLR9 stimulation were not observed. Notably, the soluble interferon response to viral stimulation by mononuclear cells was not diminished in pregnancy. CONCLUSIONS Our data provide insight into the differential responsiveness of pregnancy-derived monocytes to ssRNA and dsRNA, mainly driven by TLR8 and membrane-bound TLR3, which may help to explain the increased susceptibility of pregnant women to adverse outcomes resulting from viral infection as observed during recent and historic pandemics.
Collapse
Affiliation(s)
- Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330024, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Dustyn Levenson
- Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Li Tao
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330024, Santiago, Chile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Zhenjie Liu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Gaurav Bhatti
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Megan Seyerle
- Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, 48202, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
8
|
Leon-Martinez D, Lynn T, Abrahams VM. Cell-free fetal DNA impairs trophoblast migration in a TLR9-dependent manner and can be reversed by hydroxychloroquine. J Reprod Immunol 2023; 157:103945. [PMID: 37062109 DOI: 10.1016/j.jri.2023.103945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/13/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Growing evidence suggests a relationship between elevated circulating placental-derived cell-free fetal DNA (cffDNA) and preeclampsia. Hypomethylation of CpG motifs, a hallmark of cffDNA, allows it to activate Toll-like receptor 9 (TLR9). Using an in vitro human first trimester extravillous trophoblast cell model, we sought to determine if trophoblast-derived cffDNA and ODN 2216, a synthetic unmethylated CpG oligodeoxynucleotide, directly impacted spontaneous trophoblast migration. The role of the DNA sensors TLR9, AIM2, and cGAS was assessed using the inhibitor A151. To test whether any effects could be reversed by therapeutic agents, trophoblasts were treated with or without cffDNA or ODN 2216 with or without aspirin (ASA; a known cGAS inhibitor), aspirin-triggered lipoxin (ATL), or hydroxychloroquine (HCQ; a known TLR9 inhibitor). Trophoblast-derived cffDNA and ODN 2216 reduced trophoblast migration without affecting cell viability. Reduced trophoblast migration in response to cffDNA or ODN 2216 was reversed by A151. cffDNA inhibition of trophoblast migration was reversed by HCQ, while ASA or ATL had no effect. In contrast ODN 2216 inhibition of trophoblast migration was reversed by ASA, ATL and HCQ. Our findings suggest that cffDNA can exert a local effect on placental function by impairing trophoblast migration through activation of innate immune DNA sensors. HCQ, a known TLR9 inhibitor, reversed the effects of cffDNA on trophoblast migration. Greater insights into the molecular underpinnings of how cffDNA impacts placentation can aid in our understanding of the pathogenesis of preeclampsia, and in the development of novel therapeutic approaches for preeclampsia therapy.
Collapse
|
9
|
The amniotic fluid proteome changes with term labor and informs biomarker discovery in maternal plasma. Sci Rep 2023; 13:3136. [PMID: 36823217 PMCID: PMC9950459 DOI: 10.1038/s41598-023-28157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/13/2023] [Indexed: 02/25/2023] Open
Abstract
The intra-uterine components of labor, namely, myometrial contractility, cervical ripening, and decidua/membrane activation, have been extensively characterized and involve a local pro-inflammatory milieu of cellular and soluble immune mediators. Targeted profiling has demonstrated that such processes extend to the intra-amniotic space, yet unbiased analyses of the proteome of human amniotic fluid during labor are lacking. Herein, we utilized an aptamer-based platform to characterize 1,310 amniotic fluid proteins and found that the proteome undergoes substantial changes with term labor (251 proteins with differential abundance, q < 0.1, and fold change > 1.25). Proteins with increased abundance in labor are enriched for immune and inflammatory processes, consistent with prior reports of labor-associated changes in the intra-uterine space. By integrating the amniotic fluid proteome with previously generated placental-derived single-cell RNA-seq data, we demonstrated the labor-driven upregulation of signatures corresponding to stromal-3 and decidual cells. We also determined that changes in amniotic fluid protein abundance are reflected in the maternal plasma proteome. Collectively, these findings provide novel insights into the amniotic fluid proteome in term labor and support its potential use as a source of biomarkers to distinguish between true and false labor by using maternal blood samples.
Collapse
|
10
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Uterine artery Doppler indices throughout gestation in women with and without previous Cesarean deliveries: a prospective longitudinal case-control study. Sci Rep 2022; 12:20913. [PMID: 36463315 PMCID: PMC9719472 DOI: 10.1038/s41598-022-25232-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022] Open
Abstract
To determine whether a history of previous Cesarean delivery (CD) impacts uterine artery (UtA) Doppler indices throughout pregnancy. Women with and without CD (NCD) were prospectively enrolled for sequential assessments of the UtA mean/median pulsatility index (UtA-PI), resistance index (UtA-RI), and systolic/diastolic ratio (UtA-S/D) at 11-13 + 6, 14-19 + 6, 30-34 + 6, and 35-37 + 6 weeks' gestation. Data from 269/269, 246/257, 237/254, and 219/242 CD/NCD participants from each gestational period were available for analysis. Multiples of the median (MoMs) of UtA Doppler indices showed biphasic temporal (Δ) pattern; with an initial dropping until the second trimester, then a subsequent elevation until late in pregnancy (p < 0.05). The measurements and Δs of the UtA indices between CD and NCD were not different (p > 0.05). Mixed-effects modelling ruled out effects from nulliparity (n = 0 and 167 for CD and NCD, respectively) (p > 0.05). History of CD neither influenced the measurements nor the temporal changes of the UtA Doppler indices throughout pregnancy. The biphasic Δs of UtA Doppler indices added to the longitudinal data pool, and may aid in future development of a more personalized prediction using sequential/contingent methodologies, which may reduce the false results from the current cross-sectional screening.
Collapse
|
12
|
Liehr T, Harutyunyan T, Williams H, Weise A. Non-Invasive Prenatal Testing in Germany. Diagnostics (Basel) 2022; 12:2816. [PMID: 36428876 PMCID: PMC9689121 DOI: 10.3390/diagnostics12112816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
In the short 10 years following the introduction of non-invasive prenatal testing (NIPT), it has been adapted in many countries around the world as a standard screening test. In this review, this development was analyzed with a special focus on Germany. As a result, it can be stated that all known advantages of NIPT apart from "compensating for having no access to centers offering invasive diagnostics" are valid for Germany. In addition, following a review of the international literature, all documented issues with NIPT are also observed in Germany. However, the German Gene Diagnostics Act (GenDG) addresses a number of these issues, for example, the regulations by GenDG hamper induced abortions, based exclusively on an abnormal NIPT result. At the same time, GenDG has created new problems, as a possible collusion between the "right not to know with regard to parts of the examination result" may occur, or that the sex of the fetus must not be reported to the pregnant woman before the 12th week of gestation. Main conclusions drawn are that appropriate training and the continuing education of the physicians providing NIPT-related counseling are needed, as well as the provision of balanced and comprehensive information for the pregnant woman or the couple that is imperative.
Collapse
Affiliation(s)
- Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, 07747 Jena, Germany
| | - Tigran Harutyunyan
- Department of Genetics and Cytology, Yerevan State University, Yerevan 0001, Armenia
| | | | - Anja Weise
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, 07747 Jena, Germany
| |
Collapse
|
13
|
Global Trends in Research on Cell-Free Nucleic Acids in Obstetrics and Gynecology during 2017–2021. J Clin Med 2022; 11:jcm11195545. [PMID: 36233412 PMCID: PMC9572904 DOI: 10.3390/jcm11195545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/14/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Objectives. The objectives of this study were to identify global trends in research on cell-free deoxyribonucleic acid (cfDNA) from a bibliometric perspective and provide researchers with new research hotspots. Methods. In all, we extracted 5038 pieces of literature from PubMed and 527 articles from the Web of Science Core Collection (WoSCC) database related to cfDNA published from 1 January 2017 to 31 December 2021. For PubMed literature, we employed co-word, biclustering, and strategic diagram analysis to describe the trends in research on cfDNA in the said five years. Then, we used VOSviewer analysis for the WoSCC database to display the trends in research on cfDNA in obstetrics and gynecology during 2017–2021. Results. Strategy diagram analysis of 95 major Medical Subject Headings terms extracted from 5038 pieces of literature indicated that cfDNA sequence analysis for non-invasive prenatal and genetic testing and its application in the fields of neoplasm genetics and diagnosis is a newly emerging immature theme of cfDNA. VOSviewer analysis of 527 articles showed the global trends in research on cfDNA in obstetrics and gynecology, for example, in terms of most influential authors, institutions, countries, journals, and five research hotspots: (1) cfDNA application in prenatal screening and prenatal diagnosis, (2) cfDNA application in assisted reproductive technology, (3) cfDNA application in pre-eclampsia, DNA methylation, etc., (4) cfDNA application in placental dysfunction and fetal growth restriction, and (5) cfDNA application in fetal chromosomal abnormalities (fetal aneuploidy). Conclusions. Comprehensive visual analysis provides information regarding authors, organizations, countries/regions, journals, research hotspots, and emerging topics in the field of cfDNA for obstetrics and gynecology research. This comprehensive study could make it easier to find a partner for project development and build a network of knowledge on this emerging topic.
Collapse
|
14
|
Pique-Regi R, Romero R, Garcia-Flores V, Peyvandipour A, Tarca AL, Pusod E, Galaz J, Miller D, Bhatti G, Para R, Kanninen T, Hadaya O, Paredes C, Motomura K, Johnson JR, Jung E, Hsu CD, Berry SM, Gomez-Lopez N. A single-cell atlas of the myometrium in human parturition. JCI Insight 2022; 7:153921. [PMID: 35260533 PMCID: PMC8983148 DOI: 10.1172/jci.insight.153921] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/26/2022] [Indexed: 01/14/2023] Open
Abstract
Parturition is a well-orchestrated process characterized by increased uterine contractility, cervical ripening, and activation of the chorioamniotic membranes; yet, the transition from a quiescent to a contractile myometrium heralds the onset of labor. However, the cellular underpinnings of human parturition in the uterine tissues are still poorly understood. Herein, we performed a comprehensive study of the human myometrium during spontaneous term labor using single-cell RNA sequencing (scRNA-Seq). First, we established a single-cell atlas of the human myometrium and unraveled the cell type–specific transcriptomic activity modulated during labor. Major cell types included distinct subsets of smooth muscle cells, monocytes/macrophages, stromal cells, and endothelial cells, all of which communicated and participated in immune (e.g., inflammation) and nonimmune (e.g., contraction) processes associated with labor. Furthermore, integrating scRNA-Seq and microarray data with deconvolution of bulk gene expression highlighted the contribution of smooth muscle cells to labor-associated contractility and inflammatory processes. Last, myometrium-derived single-cell signatures can be quantified in the maternal whole-blood transcriptome throughout pregnancy and are enriched in women in labor, providing a potential means of noninvasively monitoring pregnancy and its complications. Together, our findings provide insights into the contributions of specific myometrial cell types to the biological processes that take place during term parturition.
Collapse
Affiliation(s)
- Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA.,Detroit Medical Center, Detroit, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Azam Peyvandipour
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Errile Pusod
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Robert Para
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Tomi Kanninen
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Ola Hadaya
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Carmen Paredes
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | | | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Department of Physiology and
| | - Stanley M Berry
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA.,Department of Obstetrics and Gynecology and.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
15
|
Kazemi NY, Fedyshyn B, Yelsa I, Fedyshyn Y, Ruano R, Markovic SN, Chakraborty R, Enninga EAL. Increased cell-free fetal DNA release after apoptosis and sterile inflammation in human trophoblast cells. Am J Reprod Immunol 2021; 86:e13483. [PMID: 34233077 PMCID: PMC8541917 DOI: 10.1111/aji.13483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/02/2021] [Accepted: 07/05/2021] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Cell-free fetal DNA (cffDNA) shed from the placenta can be detected in maternal blood and increases incrementally during gestation. Concentrations are further elevated with pregnancy complications. Specific activators of cffDNA release in such complications have not been identified. Here, we use trophoblast cells from early and term placenta to examine cffDNA release following apoptosis, infection, and sterile inflammatory stress. METHOD OF STUDY HTR8/SVneo cells were used to model first-trimester trophoblasts, and term cytotrophoblasts (CTBs) were isolated from placentae collected after uncomplicated deliveries. Trophoblasts were treated with varying concentrations of doxorubicin (DOX), lipopolysaccharide (LPS), or high-mobility group box protein 1 (HMGB1) for 18 h. Cells or supernatants were quantified for caspase-3/7 cleavage, pro-inflammatory cytokine secretion, and cffDNA release. RESULTS Both HTR8/SVneo and CTBs underwent caspase-3/7 cleavage following DOX treatment, with HTR8/SVneo cells more sensitive to apoptosis than term CTBs. Apoptotic cells released more cffDNA in a dose-dependent manner. Treatment with LPS resulted in an increase in pro-inflammatory IL-6 release, particularly in term CTBs compared to early trophoblasts; however, LPS did not affect cffDNA release. Lastly, while neither cell released more TNF-α following stimulation with HMGB1, both HTR8/SVneo and CTBs released significantly more cffDNA in the presence of HMGB1. CONCLUSIONS These data show that apoptosis and sterile inflammation induced by DOX and HMGB1, respectively, cause an increase in cffDNA concentrations in both first-trimester and term trophoblasts. Understanding physiologic release of cffDNA during healthy and complicated pregnancy can identify new targets for the diagnosis and treatment of gestational complications.
Collapse
Affiliation(s)
| | - Bohdana Fedyshyn
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Isabel Yelsa
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | - Yaroslav Fedyshyn
- Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rodrigo Ruano
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
| | | | - Rana Chakraborty
- Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth Ann L Enninga
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Stupak A, Kwaśniewski W, Goździcka-Józefiak A, Kwaśniewska A. The Influence of Maternal Obesity on Cell-Free Fetal DNA and Blood Pressure Regulation in Pregnancies with Hypertensive Disorders. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:962. [PMID: 34577885 PMCID: PMC8472671 DOI: 10.3390/medicina57090962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022]
Abstract
Background and Objectives: obesity and blood pressure disorders are one of the main risk factors for antenatal, intra, postpartum, and neonatal complications. In preeclampsia (PE), the placental hypoxia leads to vascular endothelium dysfunction, cell necrosis, and apoptosis. This condition is associated with the release of free fetal DNA (cffDNA) circulating in plasma. The disturbance of the efficiency of vasodilatation and blood pressure regulation in PE can be confirmed by analyzing the apelin, salusin, and prosalusin. This study aimed to assess the influence of obesity on cffDNA, and the effectiveness of maintaining normal blood pressure in patients with preeclampsia and gestational hypertension. Material and Methods: the research material was blood serum and oral mucosa swabs, obtained from 168 patients. Pregnant women were divided into the following: a control group (C)-67 women; a gestational hypertension group (GH)-35 patients; a preeclampsia with obesity group (PE + O) (pre-gravid BMI > 30)-23 patients. The rest were lean preeclamptic women (PE)-66 patients-(pre-gravid BMI < 25 in 43 women). Results: the cffDNA was observed in 1.50% of women in the C group, in 2.45% in the GH group, but in 18.18% of lean patients with preeclampsia. The cffDNA was detected in 58% of obese pregnant women with PE. The greater the placental hypoxia was in preeclampsia, the less efficient the hypotensive mechanisms, according to an analysis of the studied adipokines. The prosalusin concentration was significantly lower in the PE group with cffDNA than in the PE group without it (p = 0.008). Apelin was higher in the PE group with cffDNA (p = 0.006) compared to other groups. The same results were also observed in the subgroup with obesity. Conclusion: in preeclamptic women, obesity seems to act as an additive factor of placental damage by means of the dysregulation of hypotensive mechanisms.
Collapse
Affiliation(s)
- Aleksandra Stupak
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Wojciech Kwaśniewski
- Department of Gynecology Oncology and Gynecology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Anna Goździcka-Józefiak
- Department of Molecular Virology, Institute of Experimental Biology, Adam Mickiewicz University in Poznań, 61-614 Poznań, Poland;
| | - Anna Kwaśniewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland;
| |
Collapse
|
17
|
Banerjee S, Huang Z, Wang Z, Nakashima A, Saito S, Sharma S, Cheng S. Etiological Value of Sterile Inflammation in Preeclampsia: Is It a Non-Infectious Pregnancy Complication? Front Cell Infect Microbiol 2021; 11:694298. [PMID: 34485175 PMCID: PMC8415471 DOI: 10.3389/fcimb.2021.694298] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022] Open
Abstract
Understanding of sterile inflammation and its associated biological triggers and diseases is still at the elementary stage. This becomes more warranted in cases where infections are not associated with the pathology. Detrimental effects of bacterial and viral infections on the immune responses at the maternal-fetal interface as well as pregnancy outcomes have been well documented. However, an infection-induced etiology is not thought to be a major contributing component to severe pregnancy complications such as preeclampsia (PE) and gestational diabetes. How is then an inflammatory signal thought to be associated with these pregnancy complications? It is not clear what type of inflammation is involved in the onset of PE-like features. We opine that sterile inflammation regulated by the inflammasome-gasdermins-caspase-1 axis is a contributory factor to the onset of PE. We hypothesize that increased production and release of damage-associated molecular patterns (DAMPs) or Alarmins such as high-mobility group box1 (HMGB1), cell-free fetal DNA, uric acid, the NOD-like receptor pyrin-containing receptor 3 (NLRP3) inflammasome, IL-1β and IL-18 occur in the PE placenta. Some of these molecules have already been observed in the placenta from women with PE. Mechanistically, emerging evidence has demonstrated that excessive placental endoplasmic reticulum (ER) stress, impaired autophagy and gasdermine D (GSDMD)-mediated intrinsic pyroptosis are key events that contribute to systemic sterile inflammation in patients with PE, especially early-onset PE (e-PE). In this review, we highlight the advances on the roles of sterile inflammation and inflammatory signaling cascades involving ER stress, autophagy deficiency and pyroptosis in PE pathophysiology. Deciphering the mechanisms underlying these inflammatory pathways may provide potential diagnostic biomarkers and facilitate the development of therapeutic strategies to treat this devastating disease.
Collapse
Affiliation(s)
- Sayani Banerjee
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Zheping Huang
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Zhengke Wang
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Shigeru Saito
- Department of Obstetrics and Gynecology, University of Toyama, Toyama, Japan
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Shibin Cheng
- Department of Pediatrics, Women and Infants Hospital-Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
18
|
Kolarova TR, Gammill HS, Nelson JL, Lockwood CM, Shree R. At Preeclampsia Diagnosis, Total Cell-Free DNA Concentration is Elevated and Correlates With Disease Severity. J Am Heart Assoc 2021; 10:e021477. [PMID: 34310191 PMCID: PMC8475684 DOI: 10.1161/jaha.121.021477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Placental derived cell‐free DNA (cfDNA), widely utilized for prenatal screening, may serve as a biomarker for preeclampsia. To determine whether cfDNA parameters are altered in preeclampsia, we conducted a case‐control study using prospectively collected maternal plasma (n=20 preeclampsia, n=22 normal) using our in‐house validated prenatal screening assay. Methods and Results Isolated cfDNA was quantified, sequenced using Illumina NextSeq 500, and the placental‐derived fraction was determined. Clinical and test characteristics were compared between preeclampsia and controls, followed by comparisons within the preeclampsia cohort dichotomized by cfDNA concentration. Lastly, cfDNA parameters in preeclampsia were correlated with markers of disease severity. Maternal age, body mass index, gestational age at delivery, cesarean rate, and neonatal birthweight were expectedly different between groups (P≤0.05). The placental‐derived cfDNA fraction did not differ between groups (21.4% versus 16.9%, P=0.06); however, total cfDNA was more than 10 times higher in preeclampsia (1235 versus 106.5 pg/µL, P<0.001). This relationship persisted when controlling for important confounders (OR 1.22, 95% CI 1.04–1.43, P=0.01). The dichotomized preeclampsia group with the highest cfDNA concentration delivered earlier (33.2 versus 36.6 weeks, P=0.02) and had lower placental‐derived fractions (9.1% versus 21.4%, P=0.04). Among preeclampsia cases, higher total cfDNA correlated with earlier gestational age at delivery (P=0.01) and higher maximum systolic blood pressure (P=0.04). Conclusions At diagnosis, total cfDNA is notably higher in preeclampsia, whereas the placental derived fraction remains similar to healthy pregnancies. In preeclampsia, higher total cfDNA correlates with earlier gestational age at delivery and higher systolic blood pressure. These findings may indicate increased release of cfDNA from maternal tissue injury.
Collapse
Affiliation(s)
- Teodora R Kolarova
- Division of Maternal Fetal Medicine Department of Obstetrics and Gynecology University of Washington Seattle WA
| | - Hilary S Gammill
- Division of Maternal Fetal Medicine Department of Obstetrics and Gynecology University of Washington Seattle WA
| | - J Lee Nelson
- Clinical Research Division Fred Hutchinson Cancer Research Center Seattle WA.,Division of Rheumatology Department of Medicine University of Washington Seattle WA
| | | | - Raj Shree
- Division of Maternal Fetal Medicine Department of Obstetrics and Gynecology University of Washington Seattle WA
| |
Collapse
|
19
|
Rosner M, Kolbe T, Hengstschläger M. Fetomaternal microchimerism and genetic diagnosis: On the origins of fetal cells and cell-free fetal DNA in the pregnant woman. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108399. [PMID: 34893150 DOI: 10.1016/j.mrrev.2021.108399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 06/14/2023]
Abstract
During pregnancy several types of fetal cells and fetal stem cells, including pregnancy-associated progenitor cells (PAPCs), traffic into the maternal circulation. Whereas they also migrate to various maternal organs and adopt the phenotype of the target tissues to contribute to regenerative processes, fetal cells also play a role in the pathogenesis of maternal diseases. In addition, cell-free fetal DNA (cffDNA) is detectable in the plasma of pregnant women. Together they constitute the well-known phenomenon of fetomaternal microchimerism, which inspired the concept of non-invasive prenatal testing (NIPT) using maternal blood. An in-depth knowledge concerning the origins of these fetal cells and cffDNA allows a more comprehensive understanding of the biological relevance of fetomaternal microchimerism and has implications for the ongoing expansion of resultant clinical applications.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria; Department IFA Tulln, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
20
|
Hamadeh R, Mohsen A, Kobeissy F, Karouni A, Akoum H. C-Reactive Protein for Prediction or Early Detection of Pre-Eclampsia: A Systematic Review. Gynecol Obstet Invest 2021; 86:13-26. [PMID: 33902044 DOI: 10.1159/000515530] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/28/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Pre-eclampsia (PE) is a serious disease of pregnancy and one of the major causes of morbidity and mortality for both the mother and baby. This systematic review aims to detect the role of high-sensitivity C-reactive protein (CRP) in the detection of PE. METHODS Thirty-four articles published between 2001 and 2019 were included in this review. The articles were extracted from OVID Medline and Embase. The study designs of these articles are randomized controlled, cohort, case-control, and cross-sectional studies evaluating CRP as a marker to predict or early diagnose PE. The quality assessment of these articles is made by the modified Quality Assessment of Diagnostic Accuracy Studies 2 tool. Meta-analysis was not done because of clinical and statistical heterogeneity. RESULTS A positive association between CRP levels and the development of PE was confirmed in 18 studies. This positive effect was addressed in patients with normal BMI (<25 kg/m2) in 3 studies and in overweight patients in 2 studies. One study addressed this positive association in patients with a BMI ranging between 28 and 31 kg/m2. Three studies determined a cutoff level of CRP above which a significant risk of PE development should be suspected. These levels ranged between 7 and 15 mg/L. CONCLUSION CRP is a promising cost-effective biomarker that may be used in the prediction of PE. A CRP level higher than 15 mg/L may suggest initiation of low-dose aspirin in low-risk pregnancies.
Collapse
Affiliation(s)
- Rola Hamadeh
- Ecole Doctorale en Sciences et Technologie (EDST), Lebanese University, Beirut, Lebanon
| | - Amani Mohsen
- Consultant Obstetrics, Palestinian Red Crescent, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ali Karouni
- Department of Engineering, Faculty of Technology, Lebanese University, Beirut, Lebanon
| | - Hikmat Akoum
- Faculty of Public Health, Lebanese University, Saida, Lebanon
| |
Collapse
|
21
|
Hypoxia and oxidative stress induce sterile placental inflammation in vitro. Sci Rep 2021; 11:7281. [PMID: 33790316 PMCID: PMC8012380 DOI: 10.1038/s41598-021-86268-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Fetal growth restriction (FGR) and stillbirth are associated with placental dysfunction and inflammation and hypoxia, oxidative and nitrative stress are implicated in placental damage. Damage-associated molecular patterns (DAMPs) are elevated in pregnancies at increased risk of FGR and stillbirth and are associated with increase in pro-inflammatory placental cytokines. We hypothesised that placental insults lead to release of DAMPs, promoting placental inflammation. Placental tissue from uncomplicated pregnancies was exposed in vitro to hypoxia, oxidative or nitrative stress. Tissue production and release of DAMPs and cytokines was determined. Oxidative stress and hypoxia caused differential release of DAMPs including uric acid, HMGB1, S100A8, cell-free fetal DNA, S100A12 and HSP70. After oxidative stress pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, IL-8, TNFα, CCL2) were increased both within explants and in conditioned culture medium. Hypoxia increased tissue IL-1α/β, IL-6, IL-8 and TNFα levels, and release of IL-1α, IL-6 and IL-8, whereas CCL2 and IL-10 were reduced. IL1 receptor antagonist (IL1Ra) treatment prevented hypoxia- and oxidative stress-induced IL-6 and IL-8 release. These findings provide evidence that relevant stressors induce a sterile inflammatory profile in placental tissue which can be partially blocked by IL1Ra suggesting this agent has translational potential to prevent placental inflammation evident in FGR and stillbirth.
Collapse
|
22
|
Caldwell S, Almasri E, Schmidt L, Xu C, Dyr B, Wardrop J, Cacheris P. Not all low fetal fraction cell-free DNA screening failures are at increased risk for aneuploidy. Prenat Diagn 2021; 41:1372-1379. [PMID: 33682142 PMCID: PMC8596865 DOI: 10.1002/pd.5918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/11/2021] [Accepted: 01/31/2021] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To evaluate cell-free DNA (cfDNA) redraws and pregnancy outcomes following low fetal fraction (FF) cfDNA failures, as it has been suggested that a failed cfDNA screen due to insufficient FF carries increased risk for fetal aneuploidy. METHODS Here >200,000 consecutive samples were reviewed and >1,100 patients were identified with a failed cfDNA due to low FF using genome-wide massively parallel sequencing. Redraw results following the initial low FF failure were analyzed, as well as pregnancy outcomes for patients with repeated low FF failure on redraw. RESULTS Upon redraw 84.2% of samples yielded a reportable result with no enrichment of aneuploidy observed (p = 0.332). Higher maternal weights and multifetal pregnancy rates were observed in samples with insufficient FF. In patients with repeated low FF failure on redraw, almost all pregnancies resulted in apparently healthy liveborns. CONCLUSION Insufficient FF was not an indicator of aneuploidy risk or adverse pregnancy outcomes in this study. Caution should be taken in generalizing aneuploidy risk to all low FF cfDNA failures. Redrawing may be an appropriate next step, as proceeding directly with diagnostic testing for aneuploidy may be unwarranted for most patients.
Collapse
Affiliation(s)
- Samantha Caldwell
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| | - Eyad Almasri
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| | | | - Chen Xu
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| | - Brittany Dyr
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| | - Jenna Wardrop
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| | - Philip Cacheris
- Women's Health and Genetics, Laboratory Corporation of America, La Jolla, California, USA
| |
Collapse
|
23
|
The Amniotic Fluid Cell-Free Transcriptome Provides Novel Information about Fetal Development and Placental Cellular Dynamics. Int J Mol Sci 2021; 22:ijms22052612. [PMID: 33807645 PMCID: PMC7961801 DOI: 10.3390/ijms22052612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
The amniotic fluid (AF) is a complex biofluid that reflects fetal well-being during development. AF con be divided into two fractions, the supernatant and amniocytes. The supernatant contains cell-free components, including placenta-derived microparticles, protein, cell-free fetal DNA, and cell-free fetal RNA from the fetus. Cell-free mRNA (cfRNA) analysis holds a special position among high-throughput analyses, such as transcriptomics, proteomics, and metabolomics, owing to its ease of profiling. The AF cell-free transcriptome differs from the amniocyte transcriptome and alters with the progression of pregnancy and is often associated with the development of various organ systems including the fetal lung, skin, brain, pancreas, adrenal gland, gastrointestinal system, etc. The AF cell-free transcriptome is affected not only by normal physiologies, such as fetal sex, gestational age, and fetal maturity, but also by pathologic mechanisms such as maternal obesity, and genetic syndromes (Down, Edward, Turner, etc.), as well as pregnancy complications (preeclampsia, intrauterine growth restriction, preterm birth, etc.). cfRNA in the amniotic fluid originates from the placenta and fetal organs directly contacting the amniotic fluid as well as from the fetal plasma across the placenta. The AF transcriptome may reflect the fetal and placental development and therefore aid in the monitoring of normal and abnormal development.
Collapse
|
24
|
Wu Y, Werlang A, Cheng W, Lanes A, Wen SW, Walker M. Association between Levels of Total Cell-Free DNA and Development of Preeclampsia-A Literature Review. AJP Rep 2021; 11:e38-e48. [PMID: 33747613 PMCID: PMC7964254 DOI: 10.1055/s-0040-1721674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/24/2020] [Indexed: 11/03/2022] Open
Abstract
Objectives The aim of the study is to synthesize the evidence and evaluate the total cell-free deoxyribonucleic (cfDNA) associated with the prediction of preeclampsia (PE). Total cfDNA is constituted by both cell-free fetal DNA (cffDNA) originated mainly from the placenta, and maternal cfDNA derived from maternal leukocytes. Methods A systematic review was conducted by searching PubMed and Medline. Literature reporting levels of total cfDNA in the development of PE was included. Studies that only reported cffDNA, but no cfDNA concentrations were not included in this review. Results Eight studies were included. Seven reported values of cfDNA in PE patients, regardless of early or late onset PE, six of which demonstrated a significant increase of cfDNA in patients who subsequently developed PE. Seven studies evaluated cfDNA levels in the first trimester, six of which showed significant increase of cfDNA concentrations in women who later developed PE. Five studies investigated cfDNA levels in the second trimester, all presenting increased total cfDNA levels in the PE group compared with normal controls. Conclusion Total cfDNA may play a role as a biochemical marker of PE, compared with fetal cfDNA. Large prospective studies with homogeneous populations and standardized methodology are needed to further confirm its predictive value.
Collapse
Affiliation(s)
- Yi Wu
- OMNI Research Group, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Prenatal Diagnosis Centre, Shanghai JiaoTong University, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai, China
- OHRI, Ottawa Hospital Research Institute, Clinical Epidemiology Program, Ottawa, Canada
| | - Ana Werlang
- Division of Maternal–Fetal Medicine, Department of Obstetrics, Gynecology and Newborn Care, The Ottawa Hospital, Ottawa, Canada
| | - Weiwei Cheng
- Department of Obstetrics, Shanghai JiaoTong University, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai, China
| | - Andrea Lanes
- Better Outcomes and Registry Network, Ottawa, Canada
- CHEO, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Shi Wu Wen
- Department of Epidemiology and Community Medicine, University of Ottawa, Ottawa, Canada
| | - Mark Walker
- OHRI, Ottawa Hospital Research Institute, Clinical Epidemiology Program, Ottawa, Canada
- Division of Maternal–Fetal Medicine, Department of Obstetrics, Gynecology and Newborn Care, The Ottawa Hospital, Ottawa, Canada
- Canadian Institutes of Health Research, Ottawa, Canada
| |
Collapse
|
25
|
Merriel A, Alberry M, Abdel-Fattah S. Implications of non-invasive prenatal testing for identifying and managing high-risk pregnancies. Eur J Obstet Gynecol Reprod Biol 2020; 256:32-39. [PMID: 33166795 DOI: 10.1016/j.ejogrb.2020.10.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 10/23/2022]
Abstract
Non-invasive prenatal testing is regularly used to screen for aneuploidies and Rhesus status of a fetus. Since 1997 when free fetal DNA (ffDNA) in the maternal circulation was first identified, it has been hypothesized that it may be possible to use non-invasive prenatal testing (NIPT) to identify high-risk pregnancies including pre-eclampsia, growth restriction and preterm birth. Since then there has been much interest in this area as a way to identify and understand disease processes. This review presents the current evidence for this approach. For pre-eclampsia the hypothesis is that ffDNA would increase but the evidence for this is heterogenous across studies and trimesters. There is however increasing agreement between studies that by the third trimester ffDNA is more likely to be raised in pre-eclamptic patients than controls. For preterm birth, again, the main hypothesis is that ffDNA should increase. The results are also heterogenous, with some studies finding increased ffDNA prior to preterm birth, and others finding no change. For fetal growth restriction, there are competing theories for reduced and increased ffDNA and some studies suggest that levels are raised and some reduced. There are complexities in interpreting all of this evidence as the studies' designs, patient populations, and especially in the context of growth restriction, the definitions are not clear. Furthermore, authors use different biochemical tests and different units to describe their results, making meta-analysis difficult. All of these issues and conflicting findings lead us to the conclusion that currently there is yet no definitive place in clinical practice for NIPT to support the diagnosis and management of high-risk pregnancies. However, it is vital that this research continues as it could open the door to better understanding of the disease process and novel approaches to management.
Collapse
Affiliation(s)
- Abi Merriel
- Population Health Sciences, Bristol Medical School, University of Bristol, Level 3, Learning and Research Building, Southmead Hospital, Bristol, BS10 5NB, United Kingdom; Department of Women's and Children's Health, North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom.
| | - Medhat Alberry
- Weill Cornell Medicine, Doha, Qatar; Consultant Maternal Fetal Medicine and Obstetrics, Sidra Medicine, Doha, Qatar.
| | - Sherif Abdel-Fattah
- Department of Women's and Children's Health, North Bristol NHS Trust, Southmead Hospital, Bristol, BS10 5NB, United Kingdom.
| |
Collapse
|
26
|
Wertaschnigg D, Lucovnik M, Klieser E, Huber-Katamay J, Moertl MG. Increased Cell-Free Fetal DNA Fraction in the First Trimester: A Sign of Abnormally Invasive Placenta? ULTRASCHALL IN DER MEDIZIN (STUTTGART, GERMANY : 1980) 2020; 41:560-561. [PMID: 30396215 DOI: 10.1055/a-0770-5209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Affiliation(s)
- Dagmar Wertaschnigg
- Department of Obstetrics and Gynecology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Miha Lucovnik
- Department of Perinatology, University-Medical-Centre Ljubljana, Ljubljana, Slovenia
| | - Eckhard Klieser
- Department of Pathology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Judith Huber-Katamay
- Department of Obstetrics and Gynecology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Manfred Georg Moertl
- Department of Obstetrics and Gynecology, Tertiary care center for Perinatology, Medical Center, Klagenfurt, Klagenfurt, Austria
| |
Collapse
|
27
|
Serrano NC, Guio E, Becerra-Bayona SM, Quintero-Lesmes DC, Bautista-Niño PK, Colmenares-Mejía C, Páez MC, Luna ML, Díaz LA, Ortiz R, Beltrán M, Monterrosa Á, Miranda Y, Mesa CM, Saldarriaga W, Casas JP. C-reactive protein, interleukin-6 and pre-eclampsia: large-scale evidence from the GenPE case-control study. Scandinavian Journal of Clinical and Laboratory Investigation 2020; 80:381-387. [PMID: 32400228 DOI: 10.1080/00365513.2020.1747110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multiple small studies have suggested that women with pre-eclampsia present elevated levels of C-reactive protein (CRP) and interleukin-6 (IL-6). However, little is known regarding the source of this CRP and IL-6 increase. Therefore, the aim of this study was to evaluate the relationship between CRP and IL-6 levels with pre-eclampsia considering different confounding factors. Using data from a large Colombian case-control study (3,590 cases of pre-eclampsia and 4,564 normotensive controls), CRP and IL-6 levels were measured in 914 cases and 1297 controls. The association between maternal serum levels of CRP and IL-6 with pre-eclampsia risk was evaluated using adjusted logistic regression models. Pre-eclampsia was defined as presence of blood pressure ≥140/90 mmHg and proteinuria ≥300mg/24 h (or ≥1 + dipstick). There was no evidence of association between high levels of CRP and IL-6 with pre-eclampsia after adjusting for the following factors: maternal and gestational age, ethnicity, place and year of recruitment, multiple-pregnancy, socio-economic position, smoking, and presence of infections during pregnancy. The adjusted OR for 1SD increase in log-CRP and log-IL-6 was 0.96 (95%CI 0.85, 1.08) and 1.09 (95%CI 0.97, 1.22), respectively. Although previous reports have suggested an association between high CRP and IL-6 levels with pre-eclampsia, sample size may lack the sufficient power to draw robust conclusions, and this association is likely to be explained by unaccounted biases. Our results, the largest case-control study reported up to date, demonstrate that there is not a causal association between elevated levels of CRP and IL-6 and the presence of pre-eclampsia.
Collapse
Affiliation(s)
- Norma C Serrano
- Fundación Cardiovascular de Colombia, FCV, Floridablanca, Colombia.,Hospital Internacional de Colombia HIC, Floridablanca, Colombia
| | - Elizabeth Guio
- Fundación Cardiovascular de Colombia, FCV, Floridablanca, Colombia
| | | | | | | | | | - María C Páez
- Universidad Autónoma de Bucaramanga UNAB, Bucaramanga, Colombia
| | - María L Luna
- Universidad Autónoma de Bucaramanga UNAB, Bucaramanga, Colombia
| | - Luis A Díaz
- Universidad Industrial de Santander UIS, Bucaramanga, Colombia
| | - Ricardo Ortiz
- Universidad Autónoma de Bucaramanga UNAB, Bucaramanga, Colombia
| | - Mónica Beltrán
- Universidad Autónoma de Bucaramanga UNAB, Bucaramanga, Colombia.,Universidad Industrial de Santander UIS, Bucaramanga, Colombia
| | | | | | | | | | - Juan P Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
| |
Collapse
|
28
|
Li N, He F, Gao H, Ge Y, Fan X, Zhang J, Qi H, Ren L. Elevated cell-free fetal DNA contributes to placental inflammation and antiangiogenesis via AIM2 and IFI16 during pre-eclampsia. J Cell Physiol 2020; 235:9577-9588. [PMID: 32383175 DOI: 10.1002/jcp.29766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/11/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022]
Abstract
Accumulated evidence has shown that pre-eclampsia (PE) is related to both maternal and utero-placental antiangiogenesis and inflammation. Remarkably, an elevated cell-free fetal DNA (cffDNA) level has been found in maternal circulation; however, it remains unclear whether this DNA can induce activation of cytosolic DNA sensor signaling pathways and lead to the development of PE. In this study, we found that trophoblast cells constitutively expressed the cytosolic DNA sensors, absent in melanoma 2 (AIM2) and interferon-inducible protein 16 (IFI16). The cffDNA and pro-inflammatory and antiangiogenic factors were present at higher concentrations in PE compared with the control group and correlated with the severity of PE. DNA stimulation significantly increased the AIM2 and IFI16 levels, consistent with the elevated AIM2 and IFI16 expression in women with PE, and elicited increased production of AIM2-mediated interleukin IL-8 (IL-8), IL-6 and CC chemokine ligand 2 (CCL2) and IFI16-mediated sEndoglin, sFlt-1 and CXCL10. Furthermore, enhancement of the inflammatory response was found to be induced by DNA exposure, but DNA exposure did not induce PE-like symptoms in pregnant mice. It is possible that elevated cffDNA could reflect the degree of placental damage and trigger cytosolic DNA sensor activation, which disrupts the immunity balance and, consequently, contributes to inflammatory and antiangiogenic responses. In conclusion, the results of this study suggest that circulating cffDNA levels are increased in preeclamptic women and act through AIM2 and IFI16 activation to promote the production of pro-inflammatory and antiangiogenic factors, which correlate with the severity of the disease, and may offer insights into the etiology and pathogenesis of PE.
Collapse
Affiliation(s)
- Ning Li
- Cytotherapy Laboratory, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Fei He
- Cytotherapy Laboratory, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Hang Gao
- The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Ying Ge
- Jilin Gynecology and Obstetrics Hospital, Jilin, China
| | - Xiujun Fan
- Laboratory for Reproductive Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jian Zhang
- Laboratory for Reproductive Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hui Qi
- Cytotherapy Laboratory, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| | - Lili Ren
- Cytotherapy Laboratory, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, China
| |
Collapse
|
29
|
Hopkins MK, Koelper N, Bender W, Durnwald C, Sammel M, Dugoff L. Association between cell-free DNA fetal fraction and gestational diabetes. Prenat Diagn 2020; 40:724-727. [PMID: 32091624 DOI: 10.1002/pd.5671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/27/2020] [Accepted: 02/12/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To determine the association between cell-free DNA (cfDNA) fetal fraction and gestational diabetes (GDM) in a cohort of women presenting for cfDNA screening for fetal aneuploidy. METHODS A retrospective cohort study of women with singleton pregnancies who had cfDNA screening at a single institution at 10 to 20 weeks gestation between October 2011 and October 2017. Fetal fractions were adjusted for gestational age (GA) using multiples of the median (MoM). Multivariable logistic regression was used to estimate the odds ratio (OR) of GDM controlling for potential confounders. RESULTS Two thousand six hundred twenty-three pregnancies met criteria. Women with GDM had a lower fetal fraction (0.93 MoM vs. 1.05 MoM, P = .002). However, the association between fetal fraction and GDM was not significant after adjusting for body mass index (BMI) [OR 0.84, 95% confidence interval (CI) 0.52-1.36; P = .48]. Since insulin resistance increases at later GAs, separate analysis on women with GA 14 to 20 weeks was performed. Again, the association between fetal fraction and GDM was not significant after adjusting for BMI, (OR 0.81, 95% CI 0.31-2.12; P = .67). CONCLUSION Low or high fetal fraction of cfDNA was not associated with GDM. Although fetal fraction was lower among women diagnosed with GDM, this relationship was no longer statistically significant once maternal BMI was taken into account.
Collapse
Affiliation(s)
- Maeve K Hopkins
- Division of Maternal and Fetal Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nathanael Koelper
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Whitney Bender
- Division of Maternal and Fetal Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Celeste Durnwald
- Division of Maternal and Fetal Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mary Sammel
- Department of Obstetrics and Gynecology, Center for Research on Reproduction and Women's Health, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lorraine Dugoff
- Division of Maternal and Fetal Medicine and Reproductive Genetics, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Benn P, Martin K, McKanna T, Valenti E, Billings P, Demko Z. Combining the use of a fetal fraction-based risk algorithm and probability of an informative redraw in noninvasive prenatal testing for fetal aneuploidy. J Genet Couns 2019; 29:800-806. [PMID: 31872514 PMCID: PMC7586960 DOI: 10.1002/jgc4.1208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022]
Abstract
Some women undergoing noninvasive prenatal testing (NIPT) do not receive an informative result due to low fetal fraction (FF). A proportion of these are at increased risk for fetal trisomy 13, 18, or triploidy, while others have no change from their prior risk. Women with an initial uninformative NIPT need to be counseled about any such change in their risk for fetal abnormality and also the probability that a redraw will be informative. To help in the decision making, we reviewed a dataset of single nucleotide polymorphism‐based NIPT with uninformative results where a redraw was received. Risk for trisomy 13, 18, or triploidy was evaluated using a fetal fraction‐based risk (FFBR) algorithm. Risk‐unchanged women were further analyzed using a regression model to determine the likelihood of an informative redraw. Of 2,644 women with an uninformative NIPT and a redraw, 1,147 (43.4%) were high risk for trisomy 13, 18, or triploidy. 1,497 (56.6%) were risk unchanged and, of these, 975 (65.1%) cases had an informative redraw (i.e., risks were available for 2,122 (80%) of those initially classified as uninformative). The regression model for the risk‐unchanged cases provided a new table for predicting an informative redraw. Likelihood of a successful redraw was significantly (p < .001) dependent on the initial FF, maternal weight, and time between blood draws. We conclude that the FFBR algorithm and the predictive model for an informative redraw provide complementary additions in the management of women presented with an initially uninformative SNP‐based NIPT due to low FF. We suggest approaches for the counseling and follow‐up testing for women with an initially uninformative NIPT.
Collapse
|
31
|
Ioannides M, Achilleos A, Kyriakou S, Kypri E, Loizides C, Tsangaras K, Constantinou L, Koumbaris G, Patsalis PC. Development of a new methylation-based fetal fraction estimation assay using multiplex ddPCR. Mol Genet Genomic Med 2019; 8:e1094. [PMID: 31821748 PMCID: PMC7005606 DOI: 10.1002/mgg3.1094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/17/2019] [Indexed: 01/12/2023] Open
Abstract
Background Non‐invasive prenatal testing (NIPT) for fetal aneuploidies has rapidly been incorporated into clinical practice. Current NGS‐based methods can reliably detect fetal aneuploidies non‐invasively with fetal fraction of at least 4%. Inaccurate fetal fraction assessment can compromise the accuracy of the test as affected samples with low fetal fraction have an increased risk for misdiagnosis. Using a novel set of fetal‐specific differentially methylated regions (DMRs) and methylation sensitive restriction digestion (MSRD), we developed a multiplex ddPCR assay for accurate detection of fetal fraction in maternal plasma. Methods We initially performed MSRD followed by methylation DNA immunoprecipitation (MeDIP) and NGS on fetal and non‐pregnant female tissues to identify fetal‐specific DMRs. DMRs with the highest methylation difference between the two tissues were selected for fetal fraction estimation employing MSRD and multiplex ddPCR. Chromosome Y multiplex ddPCR assay (YMM) was used as a reference standard, to develop our fetal fraction estimation model in male pregnancy samples. Additional 123 samples were tested to examine whether the model is sex dependent and/or ploidy dependent. Results In all, 93 DMRs were identified of which seven were selected for fetal fraction estimation. Statistical analysis resulted in the final model which included four DMRs (FFMM). High correlation with YMM‐based fetal fractions was observed using 85 male pregnancies (r = 0.86 95% CI: 0.80–0.91). The model was confirmed using an independent set of 53 male pregnancies. Conclusion By employing a set of well‐characterized DMRs, we developed a SNP‐, sex‐ and ploidy‐independent methylation‐based multiplex ddPCR assay for accurate fetal fraction estimation.
Collapse
|
32
|
Carrara J, Vivanti A, Jani JC, Demain A, Costa JM, Benachi A. Usefulness and reliability of cell free fetal DNA screening for main trisomies in case of atypical profile on first trimester maternal serum screening. J Transl Med 2019; 17:398. [PMID: 31779618 PMCID: PMC6883576 DOI: 10.1186/s12967-019-02152-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/22/2019] [Indexed: 11/20/2022] Open
Abstract
Background Patients with atypical values of HCG and/or PAPP-A are at higher risk of chromosomal abnormality and vascular complications of pregnancy. The performance of cfDNA in this particular population has not yet been evaluated. Objectives The primary objective was to evaluate the usefulness and reliability of cfDNA in screening for trisomy 21, 18 and 13 for patients with HCG < 0.25 multiple of median (MoM), HCG > 5.0 MoM and/or PAPP-A < 0.25 MoM, PAPP-A > 2.5 MoM. The secondary objective was to evaluate the contribution of cfDNA assay for the prediction of pregnancy’s vascular complications. Method Between June 2016 and July 2017, we analysed a women cohort from all over France who had at least one first trimester serum biomarker outside of normal range, in a retrospective, observational and multicentre study. Patients were included if they had a single pregnancy, normal first trimester ultrasound examination, whatever the result of the combined first trimester screening test was. The cfDNA was analysed by massive parallel sequencing technique. The accuracy of cfDNA assay was evaluated by calculation of sensitivity and specificity, and multivariate regression analysis was used to search for predictive factors for pregnancy’s vascular complications. Results Among the 498 patients who underwent a cfDNA assay in this context, twenty-one (4.2%) were excluded because of loss to follow-up. Out of 477, test failure occurred for four patients initially, reduced to two patients (0.4%) after redrawn. CfDNA was positive for Trisomy 21 (n = 19), Trisomy 18 (n = 6) and Trisomy 13 (n = 1) and negative in 449. The sensitivity of cfDNA assay for trisomy 21 screening was 100% (19/19) (IC 95% 82.4–100) and specificity 100% (458/458) (IC 95% 99.2–100). Among the 447 patients included for prediction of vascular complications, there were four cases of pregnancy induced hypertension and 10 cases of preeclampsia, for which no predictive factor was identified. Intra Uterine growth restriction under 5th percentile (n = 44, 9.8%) was significantly associated with a low fetal fraction (OR = 0.87, IC 95% 0.79–0.96, p = 0.006). Conclusion cfDNA assay is an effective and reliable tool for women with atypical profile of first trimester serum biomarkers.
Collapse
Affiliation(s)
- Julie Carrara
- Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, 157 Rue de la Porte de Trivaux, 92140, Clamart, France. .,Université Paris Saclay, 91190, Saint-Aubin, France.
| | - Alexandre Vivanti
- Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, 157 Rue de la Porte de Trivaux, 92140, Clamart, France.,Université Paris Saclay, 91190, Saint-Aubin, France
| | - Jacques C Jani
- Department of Obstetrics and Gynecology, University Hospital Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| | - Adèle Demain
- Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, 157 Rue de la Porte de Trivaux, 92140, Clamart, France
| | | | - Alexandra Benachi
- Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, 157 Rue de la Porte de Trivaux, 92140, Clamart, France.,Université Paris Saclay, 91190, Saint-Aubin, France
| |
Collapse
|
33
|
Novel Epigenetic Biomarkers in Pregnancy-Related Disorders and Cancers. Cells 2019; 8:cells8111459. [PMID: 31752198 PMCID: PMC6912400 DOI: 10.3390/cells8111459] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022] Open
Abstract
As the majority of cancers and gestational diseases are prognostically stage- and grade-dependent, the ultimate goal of ongoing studies in precision medicine is to provide early and timely diagnosis of such disorders. These studies have enabled the development of various new diagnostic biomarkers, such as free circulating nucleic acids, and detection of their epigenetic changes. Recently, extracellular vesicles including exosomes, microvesicles, oncosomes, and apoptotic bodies have been recognized as powerful diagnostic tools. Extracellular vesicles carry specific proteins, lipids, DNAs, mRNAs, and miRNAs of the cells that produced them, thus reflecting the function of these cells. It is believed that exosomes, in particular, may be the optimal biomarkers of pathological pregnancies and cancers, especially those that are frequently diagnosed at an advanced stage, such as ovarian cancer. In the present review, we survey and critically appraise novel epigenetic biomarkers related to free circulating nucleic acids and extracellular vesicles, focusing especially on their status in trophoblasts (pregnancy) and neoplastic cells (cancers).
Collapse
|
34
|
Kumar N, Singh AK. Cell-Free Fetal DNA: A Novel Biomarker for Early Prediction of Pre-eclampsia and Other Obstetric Complications. Curr Hypertens Rev 2019; 15:57-63. [PMID: 29766818 DOI: 10.2174/1573402114666180516131832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/06/2018] [Accepted: 05/11/2018] [Indexed: 11/22/2022]
Abstract
Hypertensive disorder of pregnancy, especially Pre-eclampsia is one of the major causes of increased maternal and perinatal morbidity and mortality all over the world. Early prediction of pre-eclampsia is the need of modern obstetrics, as this can timely prevent the progress of disease as well as related fetal and maternal morbidity and mortality. In addition to the screening of fetal aneuploidies, Rhesus-D status, fetal sex, single gene disorders, the cell-free fetal Deoxyribonucleic acid (DNA) quantification has emerged as a promising biomarker for the prediction of pre-eclampsia. Hence, its use can help in the early prediction of hypertensive disorders of pregnancy, especially pre-eclampsia even before the appearance of symptoms. Furthermore, in future, it can also help in the determination of the complete DNA sequence of every gene of the fetus. The present review focuses on recent literature concerning the use of cell-free fetal DNA in early prediction of preeclampsia as well as for non-invasive prenatal genetic screening of fetus for various disorders. Methods: The recent literature related to cell-free fetal DNA was searched from numerous English language journals and published peer-reviewed articles on Pubmed, Google Scholar, MEDLINE and various government agencies till 2016.
Collapse
Affiliation(s)
- Naina Kumar
- Department of Obstetrics and Gynecology, Maharishi Markandeshwar Institute of Medical Sciences and Research, Mullana-133207, Ambala, Haryana, India, Postal address: House Number 2, "F" Block, MMIMSR Campus, Mullana- 133207, Ambala, Haryana, India
| | - Amit Kant Singh
- Department of Physiology, U.P. University of Medical Sciences, Saifai, Etawah, Uttar Pradesh, India, Postal Address: House No. 168, Kaveri Kunj, Phase II, Kamla Nagar-282005, Agra, Uttar Pradesh, India
| |
Collapse
|
35
|
Zhao Q, HuoJiaBieKe J, Du S. The influence of fetal gender and maternal characteristics on fetal cell-free DNA in maternal plasma. J Gynecol Obstet Hum Reprod 2019; 48:653-656. [PMID: 31276844 DOI: 10.1016/j.jogoh.2019.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/12/2019] [Accepted: 07/01/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To examine the possible effects of fetal gender and maternal characteristics on concentration of fetal cell-free DNA (cfDNA). METHODS Maternal plasma that collected from 2638 singleton pregnancies women were analyzed using non-invasive prenatal testing for aneuploidy by next generation sequence technology. The effects of fetal gender and maternal BMI on fetal cfDNA was measured by Pearson correlation analysis. RESULTS The proportion of fetal cfDNA was positively correlated with gestational age (regression equation: Y=16.2483+6.8856X, r=0.1660, p<0.0001); and negatively correlated with BMI (Body Mass Index) (regression equation: Y=25.6342-19.0065X, r=-0.2146, p<0.0001); Concentration of female fetal cfDNA (mean fetal cfDNA is 13.07%, p<0.0001) is higher than male fetal cfDNA (mean fetal cfDNA is 8.37%, p<0.0001). CONCLUSIONS Fetal cfDNA increases stably between 12 and 20 weeks of gestation, and increases in a higher rate after 20 weeks. The maternal BMI is an important factor affecting fetal cfDNA, should be paid enough attention in clinical application.
Collapse
Affiliation(s)
- Qiongzhen Zhao
- Reproductive Medicine Center, XinJiang JiaYin Hospital, 48 Hubei Road, Shuimogou District, Urumchi, Xinjiang Province, 830000, China
| | - JiaNaGuLi HuoJiaBieKe
- Reproductive Medicine Center, XinJiang JiaYin Hospital, 48 Hubei Road, Shuimogou District, Urumchi, Xinjiang Province, 830000, China.
| | - Shanshan Du
- Reproductive Medicine Center, XinJiang JiaYin Hospital, 48 Hubei Road, Shuimogou District, Urumchi, Xinjiang Province, 830000, China
| |
Collapse
|
36
|
Characteristics, properties, and potential applications of circulating cell-free dna in clinical diagnostics: a focus on transplantation. J Immunol Methods 2018; 463:27-38. [DOI: 10.1016/j.jim.2018.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022]
|
37
|
Brien ME, Baker B, Duval C, Gaudreault V, Jones RL, Girard S. Alarmins at the maternal-fetal interface: involvement of inflammation in placental dysfunction and pregnancy complications 1. Can J Physiol Pharmacol 2018; 97:206-212. [PMID: 30485131 DOI: 10.1139/cjpp-2018-0363] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation is known to be associated with placental dysfunction and pregnancy complications. Infections are well known to be a cause of inflammation but they are frequently undetectable in pregnancy complications. More recently, the focus has been extended to inflammation of noninfectious origin, namely caused by endogenous mediators known as "damage-associated molecular patterns (DAMPs)" or alarmins. In this manuscript, we review the mechanism by which inflammation, sterile or infectious, can alter the placenta and its function. We discuss some classical DAMPs, such as uric acid, high mobility group box 1 (HMGB1), cell-free fetal deoxyribonucleic acid (DNA) (cffDNA), S100 proteins, heat shock protein 70 (HSP70), and adenosine triphosphate (ATP) and their impact on the placenta. We focus on the main placental cells (i.e., trophoblast and Hofbauer cells) and describe the placental response to, and release of, DAMPs. We also covered the current state of knowledge about the role of DAMPs in pregnancy complications including preeclampsia, fetal growth restriction, preterm birth, and stillbirth and possible therapeutic strategies to preserve placental function.
Collapse
Affiliation(s)
- Marie-Eve Brien
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,b Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Bernadette Baker
- c Maternal and Fetal Health Research Centre, University of Manchester, Manchester, M13 9WL, United Kingdom.,d St. Mary's Hospital, Central Manchester University Hospital National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Cyntia Duval
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Virginie Gaudreault
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Rebecca L Jones
- c Maternal and Fetal Health Research Centre, University of Manchester, Manchester, M13 9WL, United Kingdom.,d St. Mary's Hospital, Central Manchester University Hospital National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Sylvie Girard
- a Ste-Justine Hospital Research Center, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC H3T 1J4, Canada.,b Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.,e Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
38
|
Maternal total cell-free DNA in preeclampsia and fetal growth restriction: Evidence of differences in maternal response to abnormal implantation. PLoS One 2018; 13:e0200360. [PMID: 30001403 PMCID: PMC6042756 DOI: 10.1371/journal.pone.0200360] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/24/2018] [Indexed: 01/24/2023] Open
Abstract
Objectives Preeclampsia and fetal growth restriction are obstetrical syndromes associated with abnormal placental implantation and changes in the activation status of maternal leukocytes. This study is aimed to determine by a simple, rapid fluorescent assay the changes in maternal serum total cell-free DNA (t-cfDNA) concentrations in women with preeclampsia and those with fetal growth restriction (FGR). Study design A cross-sectional study was conducted measuring maternal serum t-cfDNA concentrations. Women were classified into the following groups: 1) patients with preeclampsia (n = 21); 2) FGR-estimated fetal weight below the 10thpercentile (n = 28); and 3) normal pregnancy (n = 39). Serum samples were directly assayed for t-cfDNA using a rapid fluorescent SYBR Gold assay. Elevated maternal serum t-cfDNA concentrations were defined as a cutoff>850ng/ml. Nonparametric statistics were used for analysis. Results Women with preeclampsia had a higher median maternal serum concentration (802 ng/ml, 400–2272 ng/ml) than women with a normal pregnancy (499 ng/ml, 0–1892 ng/ml, p = 0.004) and those with FGR (484 ng/ml, 72–2187 ng/ml, p = 0.012). Moreover, even patients with FGR <5th percentile and abnormal Doppler had a lower median maternal serum t-cfDNA than those with preeclampsia (median 487 ng/ml, 144–1971 ng/ml, p = 0.022). The median concentration of t-cfDNA did not differ between women with a normal pregnancy and those with FGR (p = 0.54), as well as those with fetuses <5th percentile and abnormal Doppler (p = 0.7). Women with preeclampsia had a higher proportion of elevated t-cfDNA than those with a normal pregnancy (p = 0.015) and patients with FGR (p = 0.025). Conclusions Preeclampsia is associated with higher maternal serum t-cfDNA concentration than normal pregnancy or FGR. This observation may reflect an increased systemic activation of the maternal inflammation, rather than placental; this assumption is supported by the fact that we did not observe a significant change in the maternal serum t-cfDNA in patients with placental-mediated FGR.
Collapse
|
39
|
Portelli M, Baron B. Clinical Presentation of Preeclampsia and the Diagnostic Value of Proteins and Their Methylation Products as Biomarkers in Pregnant Women with Preeclampsia and Their Newborns. J Pregnancy 2018; 2018:2632637. [PMID: 30050697 PMCID: PMC6046127 DOI: 10.1155/2018/2632637] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Preeclampsia (PE) is a disorder which affects 1-10% of pregnant women worldwide. It is characterised by hypertension and proteinuria in the later stages of gestation and can lead to maternal and perinatal morbidity and mortality. Other than the delivery of the foetus and the removal of the placenta, to date there are no therapeutic approaches to treat or prevent PE. It is thus only possible to reduce PE-related mortality through early detection, careful monitoring, and treatment of the symptoms. For these reasons the search for noninvasive, blood-borne, or urinary biochemical markers that could be used for the screening, presymptomatic diagnosis, and prediction of the development of PE is of great urgency. So far, a number of biomarkers have been proposed for predicting PE, based on pathophysiological observations, but these have mostly proven to be unreliable and inconsistent between different studies. The clinical presentation of PE and data gathered for the biochemical markers placental growth factor (PlGF), soluble Feline McDonough Sarcoma- (fms-) like tyrosine kinase-1 (sFlt-1), asymmetric dimethylarginine (ADMA), and methyl-lysine is being reviewed with the aim of providing both a clinical and biochemical understanding of how these biomarkers might assist in the diagnosis of PE or indicate its severity.
Collapse
Affiliation(s)
- Maria Portelli
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, Msida MSD2080, Malta
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, Msida MSD2080, Malta
| |
Collapse
|
40
|
Cheng SB, Davis S, Sharma S. Maternal-fetal cross talk through cell-free fetal DNA, telomere shortening, microchimerism, and inflammation. Am J Reprod Immunol 2018; 79:e12851. [PMID: 29577468 PMCID: PMC5908740 DOI: 10.1111/aji.12851] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022] Open
Abstract
There exists a strong correlation between unscheduled inflammation at the maternal-fetal interface and the continuum of pregnancy complications. In normal pregnancy, immunological tolerance is established to protect the semi-allogeneic fetus. There has been extensive research on how the immunity, endovascular trophoblast migration, and hormonal nexus are orchestrated during pregnancy at the maternal-fetal interface to program a normal pregnancy outcome. It is not clear what contributes to the plasticity of uterine immune tolerance, fetal survial, and long-term post-partum health of the mother and the offspring. Old and new concepts have reemerged and emerged that include cell-free fetal DNA (cffDNA), telomere shortening, microchimerism involving bidirectional migration of maternal and fetal cells, and pregnancy as a stress factor. The question is how these pathways converge in a gestational age-dependent manner to contribute to the health of the mother and the offspring later in life and respond to an array of inflammatory challenges. In this Review, we provide pertinent discussion on maternal-fetal cross talk through cffDNA, telomere shortening, and microchimerism in the context of inflammatory and anti-inflammatory settings, particularly how these pathways lead to normal and adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Shi-Bin Cheng
- Department of Pediatrics, Women and Infants’ Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Sarah Davis
- Department of Obstetrics and Gynecology, Women and Infants’ Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants’ Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
41
|
Romero R. A Profile of Dennis Lo, DM, DPhil, FRCP, FRCPath, FRS. Am J Obstet Gynecol 2018; 218:371-378. [PMID: 29598980 DOI: 10.1016/j.ajog.2018.01.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 01/12/2023]
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD and Detroit, MI.
| |
Collapse
|
42
|
McCarthy FP, Ryan RM, Chappell LC. Prospective biomarkers in preterm preeclampsia: A review. Pregnancy Hypertens 2018; 14:72-78. [PMID: 30527122 DOI: 10.1016/j.preghy.2018.03.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/28/2023]
Abstract
Preterm pre-eclampsia (prior to 37 weeks' gestation) remains a major cause of maternal and fetal morbidity and mortality particularly in low to middle income countries. Much research has focused on first and second trimester predictors of pre-eclampsia with the aim of allowing stratification of antenatal care and trialling of potential preventative and therapeutic agents. However, none have been shown to be of benefit in randomised controlled trials. In this literature review we critically evaluate predictive and diagnostic tests for preterm pre-eclampsia and discuss their clinical use and potential value in the management of preterm pre-eclampsia. We defined preterm pre-eclampsia as pre-eclampsia occurring prior to 37 weeks' gestation. Substantial progress has been made in the development of predictive screening tests for preterm pre-eclampsia, but further research is needed prior to their introduction and integration into routine clinical practice. The performance of diagnostic tests mainly utilising angiogenic and anti-angiogenic factors for determining time to delivery in later pregnancy currently hold more promise than first trimester predictive tests, possible reflecting the heterogeneity of pre-eclampsia.
Collapse
Affiliation(s)
- Fergus P McCarthy
- The Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Wilton, Ireland; Department of Women and Children's Health, School of Life Course Sciences, King's College London, 10th Floor North Wing, St Thomas' Hospital, London SE1 7EH, UK.
| | - Roisin M Ryan
- London School of Hygiene and Tropical Medicine, London, UK
| | - Lucy C Chappell
- NIHR Research Professor in Obstetrics, King's College London. Honorary Consultant Obstetrician, St Thomas' Hospital, London SE1 7EH, UK
| |
Collapse
|
43
|
Chan N, Smet M, Sandow R, Silva Costa F, McLennan A. Implications of failure to achieve a result from prenatal maternal serum cell‐free
DNA
testing: a historical cohort study. BJOG 2017; 125:848-855. [DOI: 10.1111/1471-0528.15006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2017] [Indexed: 12/17/2022]
Affiliation(s)
- N Chan
- Sydney Medical School University of Sydney Sydney NSW Australia
| | - M‐E Smet
- Sydney Ultrasound for Women Sydney NSW Australia
| | - R Sandow
- Sydney Ultrasound for Women Sydney NSW Australia
| | - F Silva Costa
- Monash Ultrasound for Women Melbourne VIC Australia
- Department of Obstetrics and Gynaecology Monash University Melbourne VICAustralia
| | - A McLennan
- Sydney Ultrasound for Women Sydney NSW Australia
- Discipline of Obstetrics, Gynaecology and Neonatology Faculty of Medicine University of Sydney Sydney NSW Australia
| |
Collapse
|
44
|
Abstract
Hypertensive disorders in pregnancy have been the cause of much clinical dilemma, affecting up to 10 % of all pregnancies. The precise blood pressure to achieve in a pregnant woman is usually a battle between minimizing end organ damage to the mother and providing adequate perfusion to the placenta and the fetus. This predicament is becoming more, not less, frequent as maternal ages increase in high resource nations. Biomarkers to predict preeclampsia, a subcategory of hypertension in pregnancy, have always been elusive. The discovery of angiogenic factors relevant to preeclampsia in the last decade, however, has propelled much needed research, both in the basic science and clinical arenas. In this review, we summarize the latest clinical studies and international guidelines on blood pressure goals in pregnancy, and discuss the most promising of biomarkers to predict or diagnose preeclampsia.
Collapse
|
45
|
Tsui DW, Chiu RW, Lo YD. Epigenetic approaches for the detection of fetal DNA in maternal plasma. CHIMERISM 2017; 1:30-5. [PMID: 21327153 DOI: 10.4161/chim.1.1.12439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 05/20/2010] [Indexed: 12/18/2022]
Abstract
The presence of fetal DNA in the plasma of pregnant women has opened up new possibilities for noninvasive prenatal diagnosis. Over the past decades, different types of fetal markers have been developed, initially based on discriminative genetic markers such as male-specific signals or paternally-inherited polymorphisms, and gradually evolved to the detection of fetal-specific transcripts or epigenetic signatures. This development has extended the coverage of the application of cell-free fetal DNA to essentially all pregnancies, regardless of the gender of the fetus or its polymorphic status. In this review, we present an overview of the development of noninvasive prenatal diagnosis through epigenetics. We introduce the basis of how fetal DNA could be detected from a large background of maternal DNA in maternal plasma based on fetal-specific DNA methylation patterns. We evaluate the methodologies involved and discuss the factors that affect the robustness of the detection. We review the progress in adopting fetal epigenetic markers for noninvasive prenatal assessment of fetal chromosomal aneuploidies and pregnancy-associated disorders. We conclude with comments on the future directions regarding the search for new fetal epigenetic markers and the clinical implementation of epigenetic approaches for noninvasive prenatal diagnosis.
Collapse
Affiliation(s)
- Dana Wy Tsui
- The Centre for Research into Circulating Fetal Nucleic Acids; Li Ka Shing Institute of Health Sciences; and Department of Chemical Pathology; The Chinese University of Hong Kong; Shatin, Hong Kong SAR China
| | | | | |
Collapse
|
46
|
Cell-Free Fetal DNA for the Prediction of Pre-Eclampsia at the First and Second Trimesters: A Systematic Review and Meta-Analysis. Mol Diagn Ther 2016; 21:125-135. [DOI: 10.1007/s40291-016-0245-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
47
|
Thurik FF, Lamain-de Ruiter M, Javadi A, Kwee A, Woortmeijer H, Page-Christiaens GCML, Franx A, van der Schoot CE, Koster MPH. Absolute first trimester cell-free DNA levels and their associations with adverse pregnancy outcomes. Prenat Diagn 2016; 36:1104-1111. [DOI: 10.1002/pd.4940] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/15/2016] [Accepted: 10/09/2016] [Indexed: 01/23/2023]
Affiliation(s)
- Florentine F. Thurik
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
- Division of Women and Baby, Department of Obstetrics; University Medical Centre Utrecht; Utrecht The Netherlands
| | - Marije Lamain-de Ruiter
- Division of Women and Baby, Department of Obstetrics; University Medical Centre Utrecht; Utrecht The Netherlands
| | - Ahmad Javadi
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Anneke Kwee
- Division of Women and Baby, Department of Obstetrics; University Medical Centre Utrecht; Utrecht The Netherlands
| | - Heleen Woortmeijer
- Department of Immunohematology Diagnostics, Sanquin Diagnostic Services; Amsterdam The Netherlands
| | | | - Arie Franx
- Division of Women and Baby, Department of Obstetrics; University Medical Centre Utrecht; Utrecht The Netherlands
| | - C. Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam and Landsteiner Laboratory, Academic Medical Centre; University of Amsterdam; Amsterdam The Netherlands
| | - Maria P. H. Koster
- Division of Women and Baby, Department of Obstetrics; University Medical Centre Utrecht; Utrecht The Netherlands
- Department of Obstetrics and Gynecology, Erasmus MC; University Medical Centre Rotterdam; Rotterdam The Netherlands
| |
Collapse
|
48
|
Erpenbeck L, Chowdhury CS, Zsengellér ZK, Gallant M, Burke SD, Cifuni S, Hahn S, Wagner DD, Karumanchi SA. PAD4 Deficiency Decreases Inflammation and Susceptibility to Pregnancy Loss in a Mouse Model. Biol Reprod 2016; 95:132. [PMID: 28007693 PMCID: PMC5315429 DOI: 10.1095/biolreprod.116.140293] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/21/2016] [Accepted: 10/21/2016] [Indexed: 02/06/2023] Open
Abstract
Inflammation is thought to play a critical role in the pathogenesis of placentation disorders such as recurrent miscarriages, growth restriction, and preeclampsia. Recently, neutrophil extracellular traps (NETs) have emerged as a potential mechanism for promoting inflammation in both infectious and noninfectious disorders. To investigate a pathogenic role for NETs in placentation disorders, we studied a model of antiangiogenic factor-mediated pregnancy loss in wild-type (WT) mice and in mice deficient in peptidylarginine deiminase 4 (Padi4-/-) that are unable to form NETs. Overexpression of soluble fms-like tyrosine kinase 1 (sFlt-1), an antiangiogenic protein that is pathogenically linked with abnormal placentation disorders during early gestation, resulted in pregnancy loss and large accumulation of neutrophils and NETs in WT placentas. Interestingly, sFlt-1 overexpression in Padi4-/- mice resulted in dramatically lower inflammatory and thrombotic response, which was accompanied by significant reduction in pregnancy losses. Inhibition of NETosis may serve as a novel target in disorders of impaired placentation.
Collapse
Affiliation(s)
- Luise Erpenbeck
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Chanchal Sur Chowdhury
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Zsuzsanna K Zsengellér
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Maureen Gallant
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Suzanne D Burke
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Stephen Cifuni
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Sinuhe Hahn
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - S Ananth Karumanchi
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
49
|
Nadeau-Vallée M, Obari D, Palacios J, Brien MÈ, Duval C, Chemtob S, Girard S. Sterile inflammation and pregnancy complications: a review. Reproduction 2016; 152:R277-R292. [PMID: 27679863 DOI: 10.1530/rep-16-0453] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023]
Abstract
Inflammation is essential for successful embryo implantation, pregnancy maintenance and delivery. In the last decade, important advances have been made in regard to endogenous, and therefore non-infectious, initiators of inflammation, which can act through the same receptors as pathogens. These molecules are referred to as damage-associated molecular patterns (DAMPs), and their involvement in reproduction has only recently been unraveled. Even though inflammation is necessary for successful reproduction, untimely activation of inflammatory processes can have devastating effect on pregnancy outcomes. Many DAMPs, such as uric acid, high-mobility group box 1 (HMGB1), interleukin (IL)-1 and cell-free fetal DNA, have been associated with pregnancy complications, such as miscarriages, preeclampsia and preterm birth in preclinical models and in humans. However, the specific contribution of alarmins to these conditions is still under debate, as currently there is lack of information on their mechanism of action. In this review, we discuss the role of sterile inflammation in reproduction, including early implantation and pregnancy complications. Particularly, we focus on major alarmins vastly implicated in numerous sterile inflammatory processes, such as uric acid, HMGB1, IL-1α and cell-free DNA (especially that of fetal origin) while giving an overview of the potential role of other candidate alarmins.
Collapse
Affiliation(s)
- Mathieu Nadeau-Vallée
- Departments of PediatricsOphthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Department of PharmacologyUniversité de Montréal, Montreal, Quebec, Canada
| | - Dima Obari
- Department of PharmacologyUniversité de Montréal, Montreal, Quebec, Canada
| | - Julia Palacios
- Department of Obstetrics & GynecologyCHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Marie-Ève Brien
- Department of Obstetrics & GynecologyCHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Department of MicrobiologyVirology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Cyntia Duval
- Department of Obstetrics & GynecologyCHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Sylvain Chemtob
- Departments of PediatricsOphthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada .,Department of PharmacologyUniversité de Montréal, Montreal, Quebec, Canada
| | - Sylvie Girard
- Department of PharmacologyUniversité de Montréal, Montreal, Quebec, Canada .,Department of Obstetrics & GynecologyCHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,Department of MicrobiologyVirology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
50
|
Epigenetic regulation of STAT5A and its role as fetal DNA epigenetic marker during placental development and dysfunction. Placenta 2016; 44:46-53. [PMID: 27452437 DOI: 10.1016/j.placenta.2016.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 05/19/2016] [Accepted: 06/07/2016] [Indexed: 10/21/2022]
|