1
|
Gallo DM, Fitzgerald W, Romero R, Gomez-Lopez N, Gudicha DW, Than NG, Bosco M, Chaiworapongsa T, Jung E, Meyyazhagan A, Suksai M, Gotsch F, Erez O, Tarca AL, Margolis L. Proteomic profile of extracellular vesicles in maternal plasma of women with fetal death. J Matern Fetal Neonatal Med 2023; 36:2177529. [PMID: 36813269 PMCID: PMC10395052 DOI: 10.1080/14767058.2023.2177529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVES Fetal death is a complication of pregnancy caused by multiple etiologies rather than being the end-result of a single disease process. Many soluble analytes in the maternal circulation, such as hormones and cytokines, have been implicated in its pathophysiology. However, changes in the protein content of extracellular vesicles (EVs), which could provide additional insight into the disease pathways of this obstetrical syndrome, have not been examined. This study aimed to characterize the proteomic profile of EVs in the plasma of pregnant women who experienced fetal death and to evaluate whether such a profile reflected the pathophysiological mechanisms of this obstetrical complication. Moreover, the proteomic results were compared to and integrated with those obtained from the soluble fraction of maternal plasma. METHODS This retrospective case-control study included 47 women who experienced fetal death and 94 matched, healthy, pregnant controls. Proteomic analysis of 82 proteins in the EVs and the soluble fractions of maternal plasma samples was conducted by using a bead-based, multiplexed immunoassay platform. Quantile regression analysis and random forest models were implemented to assess differences in the concentration of proteins in the EV and soluble fractions and to evaluate their combined discriminatory power between clinical groups. Hierarchical cluster analysis was applied to identify subgroups of fetal death cases with similar proteomic profiles. A p-value of <.05 was used to infer significance, unless multiple testing was involved, with the false discovery rate controlled at the 10% level (q < 0.1). All statistical analyses were performed by using the R statistical language and environment-and specialized packages. RESULTS Nineteen proteins (placental growth factor, macrophage migration inhibitory factor, endoglin, regulated upon activation normal T cell expressed and presumably secreted (RANTES), interleukin (IL)-6, macrophage inflammatory protein 1-alpha, urokinase plasminogen activator surface receptor, tissue factor pathway inhibitor, IL-8, E-Selectin, vascular endothelial growth factor receptor 2, pentraxin 3, IL-16, galectin-1, monocyte chemotactic protein 1, disintegrin and metalloproteinase domain-containing protein 12, insulin-like growth factor-binding protein 1, matrix metalloproteinase-1(MMP1), and CD163) were found to have different plasma concentrations (of an EV or a soluble fraction) in women with fetal death compared to controls. There was a similar pattern of change for the dysregulated proteins in the EV and soluble fractions and a positive correlation between the log2-fold changes of proteins significant in either the EV or the soluble fraction (ρ = 0.89, p < .001). The combination of EV and soluble fraction proteins resulted in a good discriminatory model (area under the ROC curve, 82%; sensitivity, 57.5% at a 10% false-positive rate). Unsupervised clustering based on the proteins differentially expressed in either the EV or the soluble fraction of patients with fetal death relative to controls revealed three major clusters of patients. CONCLUSION Pregnant women with fetal death have different concentrations of 19 proteins in the EV and soluble fractions compared to controls, and the direction of changes in concentration was similar between fractions. The combination of EV and soluble protein concentrations revealed three different clusters of fetal death cases with distinct clinical and placental histopathological characteristics.
Collapse
Affiliation(s)
- Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Universidad Del Valle, Cali, Colombia
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, National Institutes of Health, Bethesda, MD, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
- Detroit Medical Center, Detroit, MI, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dereje W Gudicha
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nándor Gábor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Systems, Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Arun Meyyazhagan
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, HaEmek Medical Center, Afula, Israel
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Detroit, MI, USA
- Division of Intramural Research, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Leonid Margolis
- Section on Intercellular Interactions, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Broekhuizen M, Hitzerd E, van den Bosch TPP, Dumas J, Verdijk RM, van Rijn BB, Danser AHJ, van Eijck CHJ, Reiss IKM, Mustafa DAM. The Placental Innate Immune System Is Altered in Early-Onset Preeclampsia, but Not in Late-Onset Preeclampsia. Front Immunol 2022; 12:780043. [PMID: 34992598 PMCID: PMC8724430 DOI: 10.3389/fimmu.2021.780043] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Preeclampsia is a severe placenta-related pregnancy disorder that is generally divided into two subtypes named early-onset preeclampsia (onset <34 weeks of gestation), and late-onset preeclampsia (onset ≥34 weeks of gestation), with distinct pathophysiological origins. Both forms of preeclampsia have been associated with maternal systemic inflammation. However, alterations in the placental immune system have been less well characterized. Here, we studied immunological alterations in early- and late-onset preeclampsia placentas using a targeted expression profile approach. RNA was extracted from snap-frozen placenta samples (healthy n=13, early-onset preeclampsia n=13, and late-onset preeclampsia n=6). The expression of 730 immune-related genes from the Pan Cancer Immune Profiling Panel was measured, and the data were analyzed in the advanced analysis module of nSolver software (NanoString Technology). The results showed that early-onset preeclampsia placentas displayed reduced expression of complement, and toll-like receptor (TLR) associated genes, specifically TLR1 and TLR4. Mast cells and M2 macrophages were also decreased in early-onset preeclampsia compared to healthy placentas. The findings were confirmed by an immunohistochemistry approach using 20 healthy, 19 early-onset preeclampsia, and 10 late-onset preeclampsia placentas. We conclude that the placental innate immune system is altered in early-onset preeclampsia compared to uncomplicated pregnancies. The absence of these alterations in late-onset preeclampsia placentas indicates dissimilar immunological profiles. The study revealed distinct pathophysiological processes in early-onset and late-onset preeclampsia placentas and imply that a tailored treatment to each subtype is desirable.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Emilie Hitzerd
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | - Jasper Dumas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bas B van Rijn
- Department of Obstetrics and Gynecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Casper H J van Eijck
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Dana A M Mustafa
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands.,The Tumor Immuno-Pathology (TIP) Laboratory, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
3
|
Erol Deniz M, Deniz A, Mendilcioglu I, Sanhal CY, Ozdem S, Kucukcetin IO, Kandemir H. Serial measurement of soluble endoglin for risk assessment at the diagnosis of fetal growth restriction. Int J Clin Pract 2021; 75:e14840. [PMID: 34528351 DOI: 10.1111/ijcp.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 11/29/2022] Open
Abstract
AIM In this study, we aimed to investigate the soluble endoglin (sEng) levels in pregnant women with fetal growth restriction (FGR) and to examine the possible relation of the sEng levels with the time remaining to delivery and maternal and fetal complications. METHODS A total of 42 pregnant women diagnosed with FGR were retrospectively reviewed. Using the maternal blood samples it is at the collected 24-37 gestational weeks, the sEng levels were measured. Fetal biometry measurements, umbilical artery, uterine artery, middle cerebral artery Doppler indices were documented. RESULTS Of all patients, 17 (40%) were diagnosed with early-onset FGR, while 25 (60%) were diagnosed with late-onset FGR. Abnormal Doppler findings were present in 25 (60%) patients. Of 42 newborns, 18 (42%) were hospitalised in the neonatal unit. The mean sEng level calculated by taking the average of the first and second blood samples was 63.24 ± 49.83 ng/mL. There was no statistically significant difference in the mean sEng levels between those who gave birth within four, three, and two weeks after the diagnosis of FGR and those who did not. There was a positive significant correlation between the mean sEng levels and systolic blood pressure (r = 0.319, P = .04). CONCLUSIONS We did not find a statistically significant relationship between the sEng level and the time remaining to the time of delivery in pregnant women with FGR. We found no statistically significant difference in sEng level between the groups in pregnant women with fetuses with FGR with or without maternal and fetal complications.
Collapse
Affiliation(s)
- Merve Erol Deniz
- Clinic of Obstetrics and Gynecology, Manavgat State Hospital, Antalya, Turkey
| | - Alparslan Deniz
- Department of Obstetrics and Gynecology, Alanya Alaaddin Keykubat University, Alanya, Turkey
| | - Inanc Mendilcioglu
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| | - Cem Yasar Sanhal
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| | - Sebahat Ozdem
- Department of Biochemistry, Akdeniz University, Antalya, Turkey
| | | | - Hülya Kandemir
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
4
|
M Dave K, Kaur L, Randhir KN, Mehendale SS, Sundrani DP, Chandak GR, Joshi SR. Placental growth factor and Fms related tyrosine kinase-1 are hypomethylated in preeclampsia placentae. Epigenomics 2021; 13:257-269. [PMID: 33471580 DOI: 10.2217/epi-2020-0318] [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] [Indexed: 01/21/2023] Open
Abstract
Aim: This study aims to examine the DNA methylation (DNAm) and expression patterns of genes associated with placental angiogenesis in preeclampsia. Materials & methods: DNAm and expression were examined in normotensive (n = 100) and preeclampsia (n = 100) women using pyrosequencing and quantitative real-time PCR respectively. Results: Hypomethylation at several CpGs was observed in PlGF and FLT-1 in women with preeclampsia compared to normotensive controls. PlGF expression was lower in women with preeclampsia while FLT-1 expression was comparable. DNAm at various CpGs was negatively correlated with expression in both the genes and were associated with maternal blood pressure and birth outcomes. Conclusion: DNAm and expression of angiogenic factors in placentae are differentially regulated in preeclampsia and influence birth outcomes.
Collapse
Affiliation(s)
- Kinjal M Dave
- Mother & Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune 411043, India
| | - Lovejeet Kaur
- Genomic Research on Complex diseases (GRC Group), Council of Scientific and Industrial Research Centre for Cellular & Molecular Biology (CSIR-CCMB), Hyderabad 500007, India
| | - Karuna N Randhir
- Mother & Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune 411043, India
| | - Savita S Mehendale
- Department of Gynecology & Obstetrics, Bharati Vidyapeeth Medical College & Hospital, Pune 411043, India
| | - Deepali P Sundrani
- Mother & Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune 411043, India
| | - Giriraj R Chandak
- Genomic Research on Complex diseases (GRC Group), Council of Scientific and Industrial Research Centre for Cellular & Molecular Biology (CSIR-CCMB), Hyderabad 500007, India
| | - Sadhana R Joshi
- Mother & Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune 411043, India
| |
Collapse
|
5
|
Li W, Chung CYL, Wang CC, Chan TF, Leung MBW, Chan OK, Wu L, Appiah K, Chaemsaithong P, Cheng YKY, Poon LCY, Leung TY. Monochorionic twins with selective fetal growth restriction: insight from placental whole-transcriptome analysis. Am J Obstet Gynecol 2020; 223:749.e1-749.e16. [PMID: 32437666 DOI: 10.1016/j.ajog.2020.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND The underlying pathomechanism in placenta-related selective fetal growth restriction in monochorionic diamniotic twin pregnancy is not known. OBJECTIVE This study aimed to investigate any differences in placental transcriptomic profile between the selectively growth-restricted twins and the normally grown cotwins in monochorionic diamniotic twin pregnancies. STUDY DESIGN This was a prospective study of monochorionic diamniotic twin pregnancies complicated by selective fetal growth restriction. Placental biopsy specimens were obtained from the subjects in the delivery suite. The placental transcriptome of the selectively growth-restricted twin was compared with that of the normally grown cotwin. This study was divided into 2 stages: (1) gene discovery phase in which placental tissues from 5 monochorionic diamniotic twin pregnancies complicated by selective fetal growth restriction plus 2 control twin pregnancies underwent transcriptome profiling, and transcriptome profiling was carried out using whole-genome RNA sequencing; and (2) validation phase in which placental tissues from 13 monochorionic diamniotic twin pregnancies with selective fetal growth restriction underwent RNA and protein validation. RNA and protein expression levels of candidate genes were determined using quantitative real-time polymerase chain reaction and immunohistochemistry staining. RESULTS A total of 1429 transcripts were differentially expressed in the placentae of selectively growth-restricted twin pairs, where 610 were up-regulated and 819 were down-regulated. Endoplasmic reticulum lectin and mannose 6-phosphate receptor were consistently differentially up-regulated in all placentae of selectively growth-restricted twins. Quantitative real-time polymerase chain reaction and immunohistochemistry staining were used to validate the results (P<.05). CONCLUSION The expression of endoplasmic reticulum lectin and mannose 6-phosphate receptor, which are important for angiogenesis and fetal growth, was significantly increased in the placentae of selectively growth-restricted twin of a monochorionic twin pair.
Collapse
Affiliation(s)
- Wei Li
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Claire Yik Lok Chung
- School of Life Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong; Department of Reproduction and Development, Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ting Fung Chan
- School of Life Sciences, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Maran Bo Wah Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Oi Ka Chan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong; Hong Kong Bioinformatics Centre, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ling Wu
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kubi Appiah
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yvonne Kwun Yue Cheng
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Liona Chiu Yee Poon
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tak Yeung Leung
- Department of Obstetrics and Gynaecology, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
6
|
Kreis NN, Ritter A, Louwen F, Yuan J. A Message from the Human Placenta: Structural and Immunomodulatory Defense against SARS-CoV-2. Cells 2020; 9:E1777. [PMID: 32722449 PMCID: PMC7465902 DOI: 10.3390/cells9081777] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/31/2022] Open
Abstract
The outbreak of the coronavirus disease 2019 (COVID-19) pandemic has caused a global public health crisis. Viral infections may predispose pregnant women to a higher rate of pregnancy complications, including preterm births, miscarriage, and stillbirth. Despite reports of neonatal COVID-19, definitive proof of vertical transmission is still lacking. In this review, we summarize studies regarding the potential evidence for transplacental transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), characterize the expression of its receptors and proteases, describe the placental pathology and analyze virus-host interactions at the maternal-fetal interface. We focus on the syncytium, the barrier between mother and fetus, and describe in detail its physical and structural defense against viral infections. We further discuss the potential molecular mechanisms, whereby the placenta serves as a defense front against pathogens by regulating the interferon type III signaling, microRNA-triggered autophagy and the nuclear factor-κB pathway. Based on these data, we conclude that vertical transmission may occur but rare, ascribed to the potent physical barrier, the fine-regulated placental immune defense and modulation strategies. Particularly, immunomodulatory mechanisms employed by the placenta may mitigate violent immune response, maybe soften cytokine storm tightly associated with severely ill COVID-19 patients, possibly minimizing cell and tissue damages, and potentially reducing SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany; (A.R.); (F.L.)
| | | | | | - Juping Yuan
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt, Germany; (A.R.); (F.L.)
| |
Collapse
|
7
|
Pique-Regi R, Romero R, Tarca AL, Luca F, Xu Y, Alazizi A, Leng Y, Hsu CD, Gomez-Lopez N. Does the human placenta express the canonical cell entry mediators for SARS-CoV-2? eLife 2020; 9:e58716. [PMID: 32662421 PMCID: PMC7367681 DOI: 10.7554/elife.58716] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected more than 10 million people, including pregnant women. To date, no consistent evidence for the vertical transmission of SARS-CoV-2 exists. The novel coronavirus canonically utilizes the angiotensin-converting enzyme 2 (ACE2) receptor and the serine protease TMPRSS2 for cell entry. Herein, building upon our previous single-cell study (Pique-Regi et al., 2019), another study, and new single-cell/nuclei RNA-sequencing data, we investigated the expression of ACE2 and TMPRSS2 throughout pregnancy in the placenta as well as in third-trimester chorioamniotic membranes. We report that co-transcription of ACE2 and TMPRSS2 is negligible in the placenta, thus not a likely path of vertical transmission for SARS-CoV-2. By contrast, receptors for Zika virus and cytomegalovirus, which cause congenital infections, are highly expressed by placental cell types. These data show that the placenta minimally expresses the canonical cell-entry mediators for SARS-CoV-2.
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, U.S. Department of Health and Human ServicesDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - 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, U.S. Department of Health and Human ServicesDetroitUnited States
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, University of MichiganAnn ArborUnited States
- Department of Epidemiology and Biostatistics, Michigan State UniversityEast LansingUnited States
- Detroit Medical CenterDetroitUnited States
- Department of Obstetrics and Gynecology, Florida International UniversityMiamiUnited States
| | - 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, U.S. Department of Health and Human ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Computer Science, Wayne State University College of EngineeringDetroitUnited States
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - Adnan Alazizi
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - Yaozhu Leng
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
| | - 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, U.S. Department of Health and Human ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - 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, U.S. Department of Health and Human ServicesDetroitUnited States
- Department of Obstetrics and Gynecology, Wayne State University School of MedicineDetroitUnited States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of MedicineDetroitUnited States
| |
Collapse
|
8
|
Shashar S, Kloog I, Erez O, Shtein A, Yitshak-Sade M, Sarov B, Novack L. Temperature and preeclampsia: Epidemiological evidence that perturbation in maternal heat homeostasis affects pregnancy outcome. PLoS One 2020; 15:e0232877. [PMID: 32421729 PMCID: PMC7234374 DOI: 10.1371/journal.pone.0232877] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/22/2020] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION This study aims to determine the association between temperature and preeclampsia and whether it is affected by seasonality and rural/urban lifestyle. METHODS This cohort study included women who delivered at our medical center from 2004 to 2013 (31,101 women, 64,566 deliveries). Temperature values were obtained from a spatiotemporally resolved estimation model performing predictions at a 1×1km spatial resolution. In "Warm" pregnancies >50% of gestation occurred during the spring-summer period. In cold pregnancies >50% of gestation occurred during the fall and winter. Generalized estimating equation multivariable models were used to estimate the association between temperature and incidence of preeclampsia. RESULTS 1) The incidence of preeclampsia in at least one pregnancy was 7% (2173/64,566); 2) during "warm" pregnancies, an elevation of one IQR of the average temperature in the 1st or the 3rd trimesters was associated with an increased risk to develop preeclampsia [patients with Jewish ethnicity: 1st trimester: relative risk (RR) of 2.38(95%CI 1.50; 3.80), 3rd trimester 1.94(95%CI 1.34;2.81); Bedouins: 1st trimester: RR = 2.91(95%CI 1.98;4.28), 3rd trimester: RR = 2.37(95%CI 1.75;3.20)]; 3) In "cold" pregnancies, an elevation of one IQR of average temperature was associated with a lower risk to develop preeclampsia among patients with Bedouin-Arab ethnicity RR = 0.68 (95% CI 0.49-0.94) for 1st trimester and RR = 0.62 (95% CI 0.44-0.87) for 3rd trimester. CONCLUSIONS 1) Elevated averaged temperature during the 1st or 3rd trimesters in "warm" pregnancies confer an increased risk for the development of preeclampsia, especially in nomadic patients; 2) Of interest, during cold pregnancies, elevated averaged temperature was associated with a lower risk to develop preeclampsia for nomadic patients. 3) These findings suggest temperature might be associated with perturbations in maternal heat homeostasis resulting in reallocation of energy resources and their availability to the fetus that may increase the risk for preeclampsia. This observation is especially relevant in the context of global warming and its effects on maternal/fetal reproductive health.
Collapse
Affiliation(s)
- Sagi Shashar
- Clinical Research Center, Soroka University Medical Center, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Itai Kloog
- Department of Geography and Environmental Development, Faculty of Humanities and Social Sciences, Ben Gurion University, Beersheba, Israel
| | - Offer Erez
- Division of Obstetrics and Gynecology, Maternity Department "D", Soroka University Medical Center, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Alexandra Shtein
- Department of Geography and Environmental Development, Faculty of Humanities and Social Sciences, Ben Gurion University, Beersheba, Israel
| | - Maayan Yitshak-Sade
- Department of Environmental Health Exposure, Epidemiology, and Risk Program Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Batia Sarov
- Department of Public Health, Faculty of Heath Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Lena Novack
- Clinical Research Center, Soroka University Medical Center, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
9
|
Gene Expression Profiling of Placenta from Normal to Pathological Pregnancies. Placenta 2018. [DOI: 10.5772/intechopen.80551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register]
|
10
|
Romero R, Erez O, Hüttemann M, Maymon E, Panaitescu B, Conde-Agudelo A, Pacora P, Yoon BH, Grossman LI. Metformin, the aspirin of the 21st century: its role in gestational diabetes mellitus, prevention of preeclampsia and cancer, and the promotion of longevity. Am J Obstet Gynecol 2017; 217:282-302. [PMID: 28619690 DOI: 10.1016/j.ajog.2017.06.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 05/30/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022]
Abstract
Metformin is everywhere. Originally introduced in clinical practice as an antidiabetic agent, its role as a therapeutic agent is expanding to include treatment of prediabetes mellitus, gestational diabetes mellitus, and polycystic ovarian disease; more recently, experimental studies and observations in randomized clinical trials suggest that metformin could have a place in the treatment or prevention of preeclampsia. This article provides a brief overview of the history of metformin in the treatment of diabetes mellitus and reviews the results of metaanalyses of metformin in gestational diabetes mellitus as well as the treatment of obese, non-diabetic, pregnant women to prevent macrosomia. We highlight the results of a randomized clinical trial in which metformin administration in early pregnancy did not reduce the frequency of large-for-gestational-age infants (the primary endpoint) but did decrease the frequency of preeclampsia (a secondary endpoint). The mechanisms by which metformin may prevent preeclampsia include a reduction in the production of antiangiogenic factors (soluble vascular endothelial growth factor receptor-1 and soluble endoglin) and the improvement of endothelial dysfunction, probably through an effect on the mitochondria. Another potential mechanism whereby metformin may play a role in the prevention of preeclampsia is its ability to modify cellular homeostasis and energy disposition, mediated by rapamycin, a mechanistic target. Metformin has a molecular weight of 129 Daltons and therefore readily crosses the placenta. There is considerable evidence to suggest that this agent is safe during pregnancy. New literature on the role of metformin as a chemotherapeutic adjuvant in the prevention of cancer and in prolonging life and protecting against aging is reviewed briefly. Herein, we discuss the mechanisms of action and potential benefits of metformin.
Collapse
|
11
|
Nikuei P, Rajaei M, Malekzadeh K, Nejatizadeh A, Mohseni F, Pourdarvishi F, ghashghaeezadeh N, Mohtarami M. Expression of placental growth factor mRNA in preeclampsia. Int J Reprod Biomed 2017. [DOI: 10.29252/ijrm.15.3.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
12
|
Miyazaki J, Nishizawa H, Kambayashi A, Ito M, Noda Y, Terasawa S, Kato T, Miyamura H, Shiogama K, Sekiya T, Kurahashi H, Fujii T. Increased levels of soluble corin in pre-eclampsia and fetal growth restriction. Placenta 2016; 48:20-25. [DOI: 10.1016/j.placenta.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 09/12/2016] [Accepted: 10/03/2016] [Indexed: 10/20/2022]
|
13
|
Serebrova VN, Trifonova EA, Gabidulina TV, Bukharina IY, Agarkova TA, Evtushenko ID, Maksimova NR, Stepanov VA. Detection of novel genetic markers of susceptibility to preeclampsia based on an analysis of the regulatory genes in the placental tissue. Mol Biol 2016. [DOI: 10.1134/s0026893316050162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
14
|
Vinograd A, Wainstock T, Mazor M, Mastrolia SA, Beer-Weisel R, Klaitman V, Dukler D, Hamou B, Benshalom-Tirosh N, Vinograd O, Erez O. A prior placenta accreta is an independent risk factor for post-partum hemorrhage in subsequent gestations. Eur J Obstet Gynecol Reprod Biol 2015; 187:20-4. [DOI: 10.1016/j.ejogrb.2015.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/04/2015] [Accepted: 01/13/2015] [Indexed: 10/24/2022]
|
15
|
Perimenis P, Bouckenooghe T, Delplanque J, Moitrot E, Eury E, Lobbens S, Gosset P, Devisme L, Duvillie B, Abderrahmani A, Storme L, Fontaine P, Froguel P, Vambergue A. Placental antiangiogenic prolactin fragments are increased in human and rat maternal diabetes. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1783-93. [DOI: 10.1016/j.bbadis.2014.06.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 06/20/2014] [Accepted: 06/23/2014] [Indexed: 12/17/2022]
|
16
|
Stevens A, Bonshek C, Whatmore A, Butcher I, Hanson D, De Leonibus C, Shaikh G, Brown M, O'Shea E, Victor S, Powell P, Settle P, Padmakumar B, Tan A, Odeka E, Cooper C, Birch J, Shenoy A, Westwood M, Patel L, Dunn BW, Clayton P. Insights into the pathophysiology of catch-up compared with non-catch-up growth in children born small for gestational age: an integrated analysis of metabolic and transcriptomic data. THE PHARMACOGENOMICS JOURNAL 2014; 14:376-84. [PMID: 24614687 DOI: 10.1038/tpj.2014.4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 12/07/2013] [Accepted: 01/09/2014] [Indexed: 12/11/2022]
Abstract
Small for gestational age (SGA) children exhibiting catch-up (CU) growth have a greater risk of cardiometabolic diseases in later life compared with non-catch-up (NCU) SGA children. The aim of this study was to establish differences in metabolism and gene expression profiles between CU and NCU at age 4-9 years. CU children (n=22) had greater height, weight and body mass index standard deviation scores along with insulin-like growth factor-I (IGF-I) and fasting glucose levels but lower adiponectin values than NCU children (n=11; all P<0.05). Metabolic profiling demonstrated a fourfold decrease of urine myo-inositol in CU compared with NCU (P<0.05). There were 1558 genes differentially expressed in peripheral blood mononuclear cells between the groups (P<0.05). Integrated analysis of data identified myo-inositol related to gene clusters associated with an increase in insulin, growth factor and IGF-I signalling in CU children (P<0.05). Metabolic and transcriptomic profiles in CU SGA children showed changes that may relate to cardiometabolic risk.
Collapse
Affiliation(s)
- A Stevens
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - C Bonshek
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - A Whatmore
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - I Butcher
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - D Hanson
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - C De Leonibus
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - G Shaikh
- Yorkhill Children's Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - M Brown
- 1] Centre for Endocrinology and Diabetes, Institute of Human Development, The University of Manchester, Manchester, UK [2] Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - E O'Shea
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - S Victor
- St Mary's Hospital, CMFT, Manchester, UK
| | - P Powell
- Royal Bolton Hospital, Royal Bolton Hospital NHS Foundation Trust, Manchester, UK
| | - P Settle
- Hope Hospital, Salford Royal NHS Foundation Trust, Salford, UK
| | - B Padmakumar
- North Manchester General Hospital, Pennine Acute Hospitals NHS Trust, Crumpsall, UK
| | - A Tan
- North Manchester General Hospital, Pennine Acute Hospitals NHS Trust, Crumpsall, UK
| | - E Odeka
- North Manchester General Hospital, Pennine Acute Hospitals NHS Trust, Crumpsall, UK
| | - C Cooper
- Stepping Hill Hospital, Stockport NHS Foundation Trust, Manchester, UK
| | - J Birch
- Tameside General Hospital, Tameside Hospital NHS Foundation Trust, Manchester, UK
| | - A Shenoy
- Royal Albert Edward Infirmary, Wrightington, Wigan and Leigh NHS Foundation Trust, Wigan, UK
| | - M Westwood
- Maternal and Fetal Health Research Centre, University of Manchester and St Mary's Hospital, CMFT, MAHSC, Manchester, UK
| | - L Patel
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| | - B W Dunn
- 1] Centre for Endocrinology and Diabetes, Institute of Human Development, The University of Manchester, Manchester, UK [2] Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - P Clayton
- 1] Royal Manchester Children's Hospital (RMCH), Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester Academic Health Science Centre (MAHSC), Manchester, UK [2] Centre for Paediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Sabri A, Lai D, D'Silva A, Seeho S, Kaur J, Ng C, Hyett J. Differential placental gene expression in term pregnancies affected by fetal growth restriction and macrosomia. Fetal Diagn Ther 2014; 36:173-80. [PMID: 24685769 DOI: 10.1159/000360535] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/08/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Extremes of fetal growth are associated with increased perinatal mortality and morbidity and a higher prevalence of cardiovascular disease, obesity and diabetes in later life. We aimed to identify changes in placental gene expression in pregnancies with evidence of growth dysfunction and candidate genes that may be used to identify abnormal patterns of growth prior to delivery. METHODS Growth-restricted (n = 4), macrosomic (n = 6) and normal term (n = 5) placentas were selected from a banked series (n = 200) collected immediately after caesarean section. RNA was extracted prior to microarray analysis using Affymetrix HG-U219 arrays to determine variation in gene expression. Genes of interest were confirmed using qRT-PCR. RESULTS 338 genes in the growth-restricted and 41 genes in the macrosomic group were identified to be significantly dysregulated (>2-fold change; p < 0.05). CPXM2 and CLDN1 were upregulated and TXNDC5 and LRP2 downregulated in fetal growth restriction. In macrosomia, PHLDB2 and CLDN1 were upregulated and LEP and GCH1 were downregulated. DISCUSSION Dysfunctional growth is associated with differential placental gene expression and affects genes with a whole spectrum of developmental and cellular functions. Better elucidation of these pathways may allow the development of biomarkers to identify growth abnormalities and effective prenatal intervention.
Collapse
Affiliation(s)
- Amin Sabri
- Department of Obstetrics, Gynaecology and Neonatology, Queen Elizabeth II Research Institute for Mothers and Infants, The University of Sydney, Sydney, N.S.W., Australia
| | | | | | | | | | | | | |
Collapse
|
18
|
Romero R, Tarca AL, Chaemsaithong P, Miranda J, Chaiworapongsa T, Jia H, Hassan SS, Kalita CA, Cai J, Yeo L, Lipovich L. Transcriptome interrogation of human myometrium identifies differentially expressed sense-antisense pairs of protein-coding and long non-coding RNA genes in spontaneous labor at term. J Matern Fetal Neonatal Med 2014; 27:1397-408. [PMID: 24168098 DOI: 10.3109/14767058.2013.860963] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To identify differentially expressed long non-coding RNA (lncRNA) genes in human myometrium in women with spontaneous labor at term. MATERIALS AND METHODS Myometrium was obtained from women undergoing cesarean deliveries who were not in labor (n = 19) and women in spontaneous labor at term (n = 20). RNA was extracted and profiled using an Illumina® microarray platform. We have used computational approaches to bound the extent of long non-coding RNA representation on this platform, and to identify co-differentially expressed and correlated pairs of long non-coding RNA genes and protein-coding genes sharing the same genomic loci. RESULTS We identified co-differential expression and correlation at two genomic loci that contain coding-lncRNA gene pairs: SOCS2-AK054607 and LMCD1-NR_024065 in women in spontaneous labor at term. This co-differential expression and correlation was validated by qRT-PCR, an experimental method completely independent of the microarray analysis. Intriguingly, one of the two lncRNA genes differentially expressed in term labor had a key genomic structure element, a splice site, that lacked evolutionary conservation beyond primates. CONCLUSIONS We provide, for the first time, evidence for coordinated differential expression and correlation of cis-encoded antisense lncRNAs and protein-coding genes with known as well as novel roles in pregnancy in the myometrium of women in spontaneous labor at term.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, Program for Perinatal Research and Obstetrics, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH , Bethesda, MD and Detroit, MI , USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cheng MH, Wang PH. Placentation abnormalities in the pathophysiology of preeclampsia. Expert Rev Mol Diagn 2014; 9:37-49. [DOI: 10.1586/14737159.9.1.37] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
20
|
Madan I, Than NG, Romero R, Chaemsaithong P, Miranda J, Tarca AL, Bhatti G, Draghici S, Yeo L, Mazor M, Hassan SS, Chaiworapongsa T. The peripheral whole-blood transcriptome of acute pyelonephritis in human pregnancya. J Perinat Med 2014; 42:31-53. [PMID: 24293448 PMCID: PMC5881913 DOI: 10.1515/jpm-2013-0085] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Human pregnancy is characterized by activation of the innate immune response and suppression of adaptive immunity. The former is thought to provide protection against infection for the mother, and the latter, tolerance against paternal antigens expressed in fetal cells. Acute pyelonephritis is associated with an increased risk of acute respiratory distress syndrome and sepsis in pregnant (vs. nonpregnant) women. The objective of this study was to describe the gene expression profile (transcriptome) of maternal whole blood in acute pyelonephritis. METHOD A case-control study was conducted to include pregnant women with acute pyelonephritis (n=15) and women with a normal pregnancy (n=34). Affymetrix HG-U133 Plus 2.0 arrays (Affymetrix, Santa Clara, CA, USA) were used for gene expression profiling. A linear model was used to test the association between the presence of pyelonephritis and gene expression levels while controlling for white blood cell count and gestational age. A fold change of 1.5 was considered significant at a false discovery rate of 0.1. A subset of differentially expressed genes (n=56) was tested with real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) (cases, n=19; controls, n=59). Gene ontology and pathway analyses were applied. RESULTS A total of 983 genes were differentially expressed in acute pyelonephritis: 457 were upregulated and 526 were downregulated. Significant enrichment of 300 biological processes and 63 molecular functions was found in pyelonephritis. Significantly impacted pathways in pyelonephritis included (a) cytokine-cytokine receptor interaction, (b) T-cell receptor signaling, (c) Jak-STAT signaling, and (d) complement and coagulation cascades. Of 56 genes tested by qRT-PCR, 48 (85.7%) had confirmation of differential expression. CONCLUSION This is the first study of the transcriptomic signature of whole blood in pregnant women with acute pyelonephritis. Acute infection during pregnancy is associated with the increased expression of genes involved in innate immunity and the decreased expression of genes involved in lymphocyte function.
Collapse
Affiliation(s)
- Ichchha Madan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jezid Miranda
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L. Tarca
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Moshe Mazor
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI, USA,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
21
|
An integrative view on the physiology of human early placental villi. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 114:33-48. [PMID: 24291663 DOI: 10.1016/j.pbiomolbio.2013.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/20/2013] [Indexed: 01/08/2023]
Abstract
The placenta is an indispensable organ for intrauterine protection, development and growth of the embryo and fetus. It provides tight contact between mother and conceptus, enabling the exchange of gas, nutrients and waste products. The human placenta is discoidal in shape, and bears a hemo-monochorial interface as well as villous materno-fetal interdigitations. Since Peter Medawar's astonishment to the paradoxical nature of the mother-fetus relationship in 1953, substantial knowledge in the domain of placental physiology has been gathered. In the present essay, an attempt has been made to build an integrated understanding of morphological dynamics, cell biology, and functional aspects of genomic and proteomic expression of human early placental villous trophoblast cells followed by a commentary on the future directions of research in this field.
Collapse
|
22
|
Chaiworapongsa T, Romero R, Whitten A, Tarca AL, Bhatti G, Draghici S, Chaemsaithong P, Miranda J, Kim CJ, Hassan SS. Differences and similarities in the transcriptional profile of peripheral whole blood in early and late-onset preeclampsia: insights into the molecular basis of the phenotype of preeclampsiaa. J Perinat Med 2013; 41:485-504. [PMID: 23793063 PMCID: PMC4164302 DOI: 10.1515/jpm-2013-0082] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 05/15/2013] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Preeclampsia (PE) can be sub-divided into early- and late-onset phenotypes. The pathogenesis of these two phenotypes has not been elucidated. To gain insight into the mechanisms of disease, the transcriptional profiles of whole blood from women with early- and late-onset PE were examined. METHODS A cross-sectional study was conducted to include women with: i) early-onset PE (diagnosed prior to 34 weeks, n=25); ii) late-onset PE (after 34 weeks, n=47); and iii) uncomplicated pregnancy (n=61). Microarray analysis of mRNA expression in peripheral whole blood was undertaken using Affymetrix microarrays. Differential gene expression was evaluated using a moderated t-test (false discovery rate <0.1 and fold change >1.5), adjusting for maternal white blood cell count and gestational age. Validation by real-time qRT-PCR was performed in a larger sample size [early PE (n=31), late PE (n=72) and controls (n=99)] in all differentially expressed genes. Gene ontology analysis and pathway analysis were performed. RESULTS i) 43 and 28 genes were differentially expressed in early- and late-onset PE compared to the control group, respectively; ii) qRT-PCR confirmed the microarray results for early and late-onset PE in 77% (33/43) and 71% (20/28) of genes, respectively; iii) 20 genes that are involved in coagulation (SERPINI2), immune regulation (VSIG4, CD24), developmental process (H19) and inflammation (S100A10) were differentially expressed in early-onset PE alone. In contrast, only seven genes that encoded proteins involved in innate immunity (LTF, ELANE) and cell-to-cell recognition in the nervous system (CNTNAP3) were differentially expressed in late-onset PE alone. Thirteen genes that encode proteins involved in host defense (DEFA4, BPI, CTSG, LCN2), tight junctions in blood-brain barrier (EMP1) and liver regeneration (ECT2) were differentially expressed in both early- and late-onset PE. CONCLUSION Early- and late-onset PE are characterized by a common signature in the transcriptional profile of whole blood. A small set of genes were differentially regulated in early- and late-onset PE. Future studies of the biological function, expression timetable and protein expression of these genes may provide insight into the pathophysiology of PE.
Collapse
Affiliation(s)
| | - Roberto Romero
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA
| | - Amy Whitten
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Gaurav Bhatti
- Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, MI, USA
| | - Piya Chaemsaithong
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Jezid Miranda
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Chong Jai Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sonia S Hassan
- Perinatology Research Branch, NICHD, NIH, DHHS, Detroit, MI, and Bethesda, Maryland, USA,Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
23
|
Sundrani DP, Reddy US, Joshi AA, Mehendale SS, Chavan-Gautam PM, Hardikar AA, Chandak GR, Joshi SR. Differential placental methylation and expression of VEGF, FLT-1 and KDR genes in human term and preterm preeclampsia. Clin Epigenetics 2013; 5:6. [PMID: 23621880 PMCID: PMC3640948 DOI: 10.1186/1868-7083-5-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 04/05/2013] [Indexed: 12/27/2022] Open
Abstract
Background Preeclampsia, a pregnancy complication of placental origin is associated with altered expression of angiogenic factors and their receptors. Recently, there is considerable interest in understanding the role of adverse intrauterine conditions in placental dysfunction and adverse pregnancy outcomes. Since we have observed changes in placental global DNA methylation levels in preeclampsia, this study was undertaken to examine gene promoter CpG methylation and expression of several angiogenic genes. We recruited 139 women comprising, 46 normotensive women with term delivery (≥37 weeks), 45 women with preeclampsia delivering preterm (<37 weeks) and 48 women with preeclampsia delivering at term. Expression levels and promoter CpG methylation of VEGF, FLT-1 and KDR genes in placentae from respective groups were determined by Taqman-based quantitative real time PCR and by the Sequenom® EpiTYPER™ technology respectively. Results We observed several differentially methylated CpG sites in the promoter regions of VEGF, FLT-1 and KDR between the normotensive and preeclampsia groups. We specifically observed hypomethylated CpGs in the promoter region and an increased expression of VEGF gene between term and preterm preeclampsia. However, mean promoter CpG methylation could not account for the higher expression of FLT-1 and KDR in preterm preeclampsia as compared to normotensive group. Conclusions Our data indicates altered DNA methylation patterns in the VEGF, FLT-1 and KDR genes in preeclampsia as compared to the normotensive group, which could be involved in the pathophysiology of preeclampsia. Hypomethylation of VEGF promoter and consequent upregulation of VEGF mRNA levels could be a compensatory mechanism to restore normal angiogenesis and blood flow in preterm preeclampsia. This study suggests a role of altered DNA methylation in placental angiogenesis and in determining adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Deepali P Sundrani
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| | - Umakar S Reddy
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research (CSIR), Hyderabad, 500007, India
| | - Asmita A Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| | - Savita S Mehendale
- Department of Obstetrics and Gynecology, Bharati Medical College and Hospital, Bharati Vidyapeeth University, Pune, 411043, India
| | - Preeti M Chavan-Gautam
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| | - Anandwardhan A Hardikar
- Diabetes and Islet-biology Group, NHMRC Clinical Trials Centre, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Giriraj R Chandak
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research (CSIR), Hyderabad, 500007, India
| | - Sadhana R Joshi
- Department of Nutritional Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth University, Pune, 411043, India
| |
Collapse
|
24
|
Xiang Y, Cheng Y, Li X, Li Q, Xu J, Zhang J, Liu Y, Xing Q, Wang L, He L, Zhao X. Up-regulated expression and aberrant DNA methylation of LEP and SH3PXD2A in pre-eclampsia. PLoS One 2013; 8:e59753. [PMID: 23544093 PMCID: PMC3609796 DOI: 10.1371/journal.pone.0059753] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/18/2013] [Indexed: 12/31/2022] Open
Abstract
The primary mechanism underlying pre-eclampsia (PE) remains one of the most burning problems in the obstetrics and gynecology. In this study, we performed an expression profiling screen and detected 1312 genes that were differentially expressed (p<0.05 and fold change >1.5) in PE placentas, including LEP and SH3PXD2A. After validating the microarray results, we conducted the quantitative methylation analysis of LEP and SH3PXD2A in preeclamptic (n = 16) versus normal placentas (n = 16). Our results showed that many CpG sites close to the transcriptional start site (TSS) of LEP gene were hypomethylated in placentas from pregnancies with PE compared with those of in controls, including the TSS position (p = 0.001), the binding sites of Sp1 (p = 1.57×10−4), LP1 (p = 0.023) and CEBPα (p = 0.031). Luciferase reporter analysis confirmed the aberrant methylation of LEP promoter and CEBPα co-transfection had a role in the regulation of gene expression. Our results indicated the aberrant LEP promoter methylation was involved in the development of PE. We did not find a significant methylation differences between groups in the promoter region of SH3PXD2A, however, a CGI region in the gene body (CGI34) presented a higher methylation in preeclamptic placentas (p = 1.57×10−4), which might promote the efficiency of gene transcription. We speculated that SH3PXD2A may take part in the pathogenesis of PE through its role in the regulation of trophoblast cell invasion in the period of placenta formation.
Collapse
Affiliation(s)
- Yuqian Xiang
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Cheng
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaotian Li
- Obstetrics & Gynecology Hospital of Fudan University, Shanghai, China
| | - Qiaoli Li
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jiawei Xu
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Junyu Zhang
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yun Liu
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qinghe Xing
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Lin He
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (LH); (XZ)
| | - Xinzhi Zhao
- Children’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- * E-mail: (LH); (XZ)
| |
Collapse
|
25
|
Abstract
Tumor progression and pregnancy share many common features, such as immune tolerance and invasion. The invasion of trophoblasts in the placenta into the uterine wall is essential for fetal development, and is thus precisely regulated. Its deregulation has been implicated in preeclampsia, a leading cause for maternal and perinatal mortality and morbidity. Pathogenesis of preeclampsia remains to be defined. Microarray-based gene profiling has been widely used for identifying genes responsible for preeclampsia. In this review, we have summarized the recent data from the microarray studies with preeclamptic placentas. Despite the complex of gene signatures, suggestive of the heterogeneity of preeclampsia, these studies identified a number of differentially expressed genes associated with preeclampsia. Interestingly, most of them have been reported to be tightly involved in tumor progression. We have discussed these interesting genes and analyzed their potential molecular functions in preeclampsia, compared with their roles in malignancy development. Further investigations are warranted to explore the involvement in molecular network of each identified gene, which may provide not only novel strategies for prevention and therapy for preeclampsia but also a better understanding of cancer cells. The trophoblastic cells, with their capacity for proliferation and differentiation, apoptosis and survival, migration, angiogenesis and immune modulation by exploiting similar molecular pathways, make them a compelling model for cancer research.
Collapse
|
26
|
Schmidt S, Stahl F, Mutz KO, Scheper T, Hahn A, Schuchardt JP. Transcriptome-based identification of antioxidative gene expression after fish oil supplementation in normo- and dyslipidemic men. Nutr Metab (Lond) 2012; 9:45. [PMID: 22621246 PMCID: PMC3408332 DOI: 10.1186/1743-7075-9-45] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/23/2012] [Indexed: 12/31/2022] Open
Abstract
Background The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs), especially in dyslipidemic subjects with a high risk of cardiovascular disease, are widely described in the literature. A lot of effects of n-3 PUFAs and their oxidized metabolites are triggered by regulating the expression of genes. Currently, it is uncertain if the administration of n-3 PUFAs results in different expression changes of genes related to antioxidative mechanisms in normo- and dyslipidemic subjects, which may partly explain their cardioprotective effects. The aim of this study was to investigate the effects of n-3 PUFA supplementation on expression changes of genes involved in oxidative processes. Methods Ten normo- and ten dyslipidemic men were supplemented for twelve weeks with fish oil capsules, providing 1.14 g docosahexaenoic acid and 1.56 g eicosapentaenoic acid. Gene expression levels were determined by whole genome microarray analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Results Using microarrays, we discovered an increased expression of antioxidative enzymes and a decreased expression of pro-oxidative and tissue enzymes, such as cytochrome P450 enzymes and matrix metalloproteinases, in both normo- and dyslipidemic men. An up-regulation of catalase and heme oxigenase 2 in both normo- and dyslipidemic subjects and an up-regulation of cytochrome P450 enzyme 1A2 only in dyslipidemic subjects could be observed by qRT-PCR analysis. Conclusions Supplementation of normo- and dyslipidemic subjects with n-3 PUFAs changed the expression of genes related to oxidative processes, which may suggest antioxidative and potential cardioprotective effects of n-3 PUFAs. Further studies combining genetic and metabolic endpoints are needed to verify the regulative effects of n-3 PUFAs in antioxidative gene expression to better understand their beneficial effects in health and disease prevention. Trial registration ClinicalTrials.gov (ID: NCT01089231)
Collapse
Affiliation(s)
- Simone Schmidt
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Frank Stahl
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Kai-Oliver Mutz
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Thomas Scheper
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Andreas Hahn
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Jan Philipp Schuchardt
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| |
Collapse
|
27
|
Abstract
The root cause of preeclampsia is the placenta. Preeclampsia begins to abate with the delivery of the placenta and can occur in the absence of a fetus but with the presence of trophoblast tissue with hydatidiform moles. In view of this, study of the placenta should provide insight into the pathophysiology of preeclampsia. In this presentation we examine placental pathological and pathophysiological changes with preeclampsia and fetal growth restriction (FGR). It would seem that this comparison should be illuminating as both conditions are associated with similarly abnormal placentation yet only in preeclampsia is there a maternal pathophysiological syndrome. Similar insights about early and late onset preeclampsia should also be provided by such information.We report that the placental abnormalities in preeclampsia are what would be predicted in a setting of reduced perfusion and oxidative stress. However, the differences from FGR are inconsistent. The most striking differences between the two conditions are found in areas that have been the least studied. There are differences between the placental findings in early and late onset preeclampsia but whether these are qualitative, indicating different diseases, or simply quantitative differences within the same disease is difficult to determine.We attempt to decipher the true differences, seek an explanation for the disparate results and provide recommendations that we hope may help resolve these issues in future studies.
Collapse
Affiliation(s)
- James M Roberts
- Magee Women Research Institute, Department of Obstetrics and Gynecology, Epidemiology and Clinical and Translational Research, University of Pittsburgh, USA
| | | |
Collapse
|
28
|
Ouhilal S, Vuguin P, Cui L, Du XQ, Gelling RW, Reznik SE, Russell R, Parlow AF, Karpovsky C, Santoro N, Charron MJ. Hypoglycemia, hyperglucagonemia, and fetoplacental defects in glucagon receptor knockout mice: a role for glucagon action in pregnancy maintenance. Am J Physiol Endocrinol Metab 2012; 302:E522-31. [PMID: 22167521 PMCID: PMC3311287 DOI: 10.1152/ajpendo.00420.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Alterations in insulin signaling as well as insulin action predispose to infertility as well as adverse pregnancy outcomes; however, little is known about the role of glucagon signaling in reproduction. The glucagon receptor knockout (Gcgr(-/-)) mouse created by our laboratory was used to define the role of glucagon signaling in maintaining normal reproduction. In this mouse model, lack of glucagon signaling did not alter the hypothalamic-pituitary-ovarian axis. Pregnant Gcgr(-/-) female mice displayed persistent hypoglycemia and hyperglucagonemia. Gcgr(-/-) pregnancies were associated with decreased fetal weight, increased late-gestation fetal demise, and significant abnormalities of placentation. Gcgr(-/-) placentas contained areas of extensive mineralization, fibrinoid necrosis, narrowing of the vascular channels, and a thickened interstitium associated with trophoblast hyperplasia. Absent glucagon signaling did not alter glycogen content in Gcgr(-/-) placentas but significantly downregulated genes that control growth, adrenergic signaling, vascularization, oxidative stress, and G protein-coupled receptors. Our data suggest that, similarly to insulin, glucagon action contributes to normal female reproductive function.
Collapse
Affiliation(s)
- Sophia Ouhilal
- Department of Obstetrics and Gynecology, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chang SD, Chao AS, Peng HH, Chang YL, Wang CN, Cheng PJ, Lee YS, Chao A, Wang TH. Analyses of placental gene expression in pregnancy-related hypertensive disorders. Taiwan J Obstet Gynecol 2012; 50:283-91. [PMID: 22030040 DOI: 10.1016/j.tjog.2011.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2011] [Indexed: 10/15/2022] Open
Abstract
OBJECTIVE To explore the changes in placental gene expression between women with preeclampsia and those with superimposed preeclampsia on chronic hypertension. MATERIALS AND METHODS In Taiwanese population, we compared gene expression between the placentas from preeclamptic patients and those with superimposed preeclampsia on chronic hypertension. RESULTS Although top-ranked activated genes between preeclampsia and superimposed preeclampsia on chronic hypertension were different, functional network analyses indicate that these genes are mainly involved in the regulation of cell death and apoptosis. These results suggest that apoptosis and other types of cell death in the placenta are common consequences of both diseases. However, placental endoglin (ENG) was expressed at a significantly higher level in preeclampsia than in superimposed preeclampsia. Results of functional network analysis indicated that ENG may play a role in the pathogenesis of preeclampsia through its interference with the endothelial nitric oxide synthase-regulated vasodilation. CONCLUSION Our results support the fact that ENG is the culprit for the development of preeclampsia. In addition, this study identifies several other genes in the placenta, which are transcriptionally regulated in pregnancy-related hypertensions.
Collapse
Affiliation(s)
- Shuenn-Dyh Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Lin-Kou Medical Center, Chang Gung University, Tao-Yuan, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
OBJECTIVE To synthesize scientific literature that addresses the role of endoglin (ENG) gene expression in preeclampsia (PE). DATA SOURCES A literature search of PubMed and Ovid MEDLINE was conducted using the keywords endoglin, gene, and preeclampsia. Restrictions included English language and humans. Additional articles were identified/selected for evaluation via PubMed e-mail updates (keywords: endoglin and preeclampsia) and review of article reference lists obtained from the search. STUDY SELECTION The initial 14 abstracts retrieved from the literature search were reviewed and 9 studies were selected for evaluation. Review articles and studies not addressing ENG expression (messenger RNA [mRNA] level) in the context of PE were excluded. An additional six articles were selected from PubMed e-mail updates and reference lists. DATA EXTRACTION Data related to study objective, design, setting, subject information, phenotype, tissue type, data collection method, statistics, and results were extracted. DATA SYNTHESIS Regardless of PE definition, ancestral background, gene expression analysis method, tissue type, and time of specimen collection, endoglin appears to play a role in PE development. Moreover, results suggest that a variety of biological mechanisms have the ability to modulate ENG expression in PE, demonstrating the potential complexity associated with endoglin's role in PE. CONCLUSIONS This review article is the first to systematically synthesize evidence related to ENG expression in PE. Findings can be utilized to design future studies that (a) address methodological limitations observed in the reviewed studies and (b) specifically examine why ENG expression levels are altered and address mechanisms explaining how these alterations are involved in PE development.
Collapse
|
31
|
Nishizawa H, Ota S, Suzuki M, Kato T, Sekiya T, Kurahashi H, Udagawa Y. Comparative gene expression profiling of placentas from patients with severe pre-eclampsia and unexplained fetal growth restriction. Reprod Biol Endocrinol 2011; 9:107. [PMID: 21810232 PMCID: PMC3199758 DOI: 10.1186/1477-7827-9-107] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/02/2011] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND It has been well documented that pre-eclampsia and unexplained fetal growth restriction (FGR) have a common etiological background, but little is known about their linkage at the molecular level. The aim of this study was to further investigate the mechanisms underlying pre-eclampsia and unexplained FGR. METHODS We analyzed differentially expressed genes in placental tissue from severe pre-eclamptic pregnancies (n = 8) and normotensive pregnancies with or (n = 8) without FGR (n = 8) using a microarray method. RESULTS A subset of the FGR samples showed a high correlation coefficient overall in the microarray data from the pre-eclampsia samples. Many genes that are known to be up-regulated in pre-eclampsia are also up-regulated in FGR, including the anti-angiogenic factors, FLT1 and ENG, believed to be associated with the onset of maternal symptoms of pre-eclampsia. A total of 62 genes were found to be differentially expressed in both disorders. However, gene set enrichment analysis for these differentially expressed genes further revealed higher expression of TP53-downstream genes in pre-eclampsia compared with FGR. TP53-downstream apoptosis-related genes, such as BCL6 and BAX, were found to be significantly more up-regulated in pre-eclampsia than in FGR, although the caspases are expressed at equivalent levels. CONCLUSIONS Our current data indicate a common pathophysiology for FGR and pre-eclampsia, leading to an up-regulation of placental anti-angiogenic factors. However, our findings also suggest that it may possibly be the excretion of these factors into the maternal circulation through the TP53-mediated early-stage apoptosis of trophoblasts that leads to the maternal symptoms of pre-eclampsia.
Collapse
Affiliation(s)
- Haruki Nishizawa
- Department of Obstetrics and Gynecology, Fujita Health University School of Medicine, Fujita Health University, Toyoake, Japan
| | - Sayuri Ota
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Machiko Suzuki
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Takema Kato
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Takao Sekiya
- Department of Obstetrics and Gynecology, Fujita Health University School of Medicine, Fujita Health University, Toyoake, Japan
| | - Hiroki Kurahashi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yasuhiro Udagawa
- Department of Obstetrics and Gynecology, Fujita Health University School of Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
32
|
Laskowska M, Laskowska K, Oleszczuk J. Endoglin in pregnancy complicated by fetal intrauterine growth restriction in normotensive and preeclamptic pregnant women: a comparison between preeclamptic patients with appropriate-for-gestational-age weight infants and healthy pregnant women. J Matern Fetal Neonatal Med 2011; 25:806-11. [DOI: 10.3109/14767058.2011.595852] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
SenthamaraiKannan P, Sartor MA, O'Connor KT, Neumann JC, Klyza JP, Succop PA, Wagner BD, Karyala S, Medvedovic M, Menon AG. Identification of maternally regulated fetal gene networks in the placenta with a novel embryo transfer system in mice. Physiol Genomics 2011; 43:317-24. [PMID: 21205871 PMCID: PMC3092334 DOI: 10.1152/physiolgenomics.00078.2010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 01/03/2011] [Indexed: 11/22/2022] Open
Abstract
The mechanisms for provisioning maternal resources to offspring in placental mammals involve complex interactions between maternally regulated and fetally regulated gene networks in the placenta, a tissue that is derived from the zygote and therefore of fetal origin. Here we describe a novel use of an embryo transfer system in mice to identify gene networks in the placenta that are regulated by the mother. Mouse embryos from the same strain of inbred mice were transferred into a surrogate mother either of the same strain or from a different strain, allowing maternal and fetal effects on the placenta to be separated. After correction for sex and litter size, maternal strain overrode fetal strain as the key determinant of fetal weight (P < 0.0001). Computational filtering of the placental transcriptome revealed a group of 81 genes whose expression was solely dependent on the maternal strain [P < 0.05, false discovery rate (FDR) < 0.10]. Network analysis of this group of genes yielded highest statistical significance for pathways involved in the regulation of cell growth (such as insulin-like growth factors) as well as those involved in regulating lipid metabolism [such as the low-density lipoprotein receptor-related protein 1 (LRP1), LDL, and HDL], both of which are known to play a role in fetal development. This novel technique may be generally applied to identify regulatory networks involved in maternal-fetal interaction and eventually help identify molecular targets in disorders of fetal growth.
Collapse
|
34
|
Edwards DRV, Romero R, Kusanovic JP, Hassan SS, Mazaki-Tovi S, Vaisbuch E, Kim CJ, Erez O, Chaiworapongsa T, Pearce BD, Bartlett J, Friel LA, Salisbury BA, Anant MK, Vovis GF, Lee MS, Gomez R, Behnke E, Oyarzun E, Tromp G, Menon R, Williams SM. Polymorphisms in maternal and fetal genes encoding for proteins involved in extracellular matrix metabolism alter the risk for small-for-gestational-age. J Matern Fetal Neonatal Med 2011; 24:362-80. [PMID: 20617897 PMCID: PMC3104673 DOI: 10.3109/14767058.2010.497572] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To examine the association between maternal and fetal genetic variants and small-for-gestational-age (SGA). METHODS A case-control study was conducted in patients with SGA neonates (530 maternal and 436 fetal) and controls (599 maternal and 628 fetal); 190 candidate genes and 775 SNPs were studied. Single-locus, multi-locus and haplotype association analyses were performed on maternal and fetal data with logistic regression, multifactor dimensionality reduction (MDR) analysis, and haplotype-based association with 2 and 3 marker sliding windows, respectively. Ingenuity pathway analysis (IPA) software was used to assess pathways that associate with SGA. RESULTS The most significant single-locus association in maternal data was with a SNP in tissue inhibitor of metalloproteinase 2 (TIMP2) (rs2277698 OR = 1.71, 95% CI [1.26-2.32], p = 0.0006) while in the fetus it was with a SNP in fibronectin 1 isoform 3 preproprotein (FN1) (rs3796123, OR = 1.46, 95% CI [1.20-1.78], p = 0.0001). Both SNPs were adjusted for potential confounders (maternal body mass index and fetal sex). Haplotype analyses resulted in associations in α 1 type I collagen preproprotein (COL1A1, rs1007086-rs2141279-rs17639446, global p = 0.006) in mothers and FN1 (rs2304573-rs1250204-rs1250215, global p = 0.045) in fetuses. Multi-locus analyses with MDR identified a two SNP model with maternal variants collagen type V α 2 (COL5A2) and plasminogen activator urokinase (PLAU) predicting SGA outcome correctly 59% of the time (p = 0.035). CONCLUSIONS Genetic variants in extracellular matrix-related genes showed significant single-locus association with SGA. These data are consistent with other studies that have observed elevated circulating fibronectin concentrations in association with increased risk of SGA. The present study supports the hypothesis that DNA variants can partially explain the risk of SGA in a cohort of Hispanic women.
Collapse
Affiliation(s)
- Digna R. Velez Edwards
- Vanderbilt Epidemiology Center, Institute of Medicine and Public Health, Department of Obstetrics and Gynecology, Vanderbilt University, Nashville, Tennessee, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Wayne State University, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Brad D. Pearce
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Jacquelaine Bartlett
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
| | - Lara A. Friel
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | | - Ricardo Gomez
- CEDIP (Center for Perinatal Diagnosis and Research), Department of Obstetrics and Gynecology, Sotero del Rio Hospital, Santiago, Chile
- Department of Obstetrics and Gynecology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Ernesto Behnke
- CEDIP (Center for Perinatal Diagnosis and Research), Department of Obstetrics and Gynecology, Sotero del Rio Hospital, Santiago, Chile
| | - Enrique Oyarzun
- Department of Obstetrics and Gynecology, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Gerard Tromp
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Ramkumar Menon
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Scott M. Williams
- Center for Human Genetics Research, Vanderbilt University, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
35
|
Romero R, Mazaki-Tovi S, Vaisbuch E, Kusanovic JP, Chaiworapongsa T, Gomez R, Nien JK, Yoon BH, Mazor M, Luo J, Banks D, Ryals J, Beecher C. Metabolomics in premature labor: a novel approach to identify patients at risk for preterm delivery. J Matern Fetal Neonatal Med 2010; 23:1344-59. [PMID: 20504069 PMCID: PMC3440243 DOI: 10.3109/14767058.2010.482618] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Biomarkers for preterm labor (PTL) and delivery can be discovered through the analysis of the transcriptome (transcriptomics) and protein composition (proteomics). Characterization of the global changes in low-molecular weight compounds which constitute the 'metabolic network' of cells (metabolome) is now possible by using a 'metabolomics' approach. Metabolomic profiling has special advantages over transcriptomics and proteomics since the metabolic network is downstream from gene expression and protein synthesis, and thus more closely reflects cell activity at a functional level. This study was conducted to determine if metabolomic profiling of the amniotic fluid can identify women with spontaneous PTL at risk for preterm delivery, regardless of the presence or absence of intraamniotic infection/inflammation (IAI). STUDY DESIGN Two retrospective cross-sectional studies were conducted, including three groups of pregnant women with spontaneous PTL and intact membranes: (1) PTL who delivered at term; (2) PTL without IAI who delivered preterm; and (3) PTL with IAI who delivered preterm. The first was an exploratory study that included 16, 19, and 20 patients in groups 1, 2, and 3, respectively. The second study included 40, 33, and 40 patients in groups 1, 2, and 3, respectively. Amniotic fluid metabolic profiling was performed by combining chemical separation (with gas and liquid chromatography) and mass spectrometry. Compounds were identified using authentic standards. The data were analyzed using discriminant analysis for the first study and Random Forest for the second. RESULTS (1) In the first study, metabolomic profiling of the amniotic fluid was able to identify patients as belonging to the correct clinical group with an overall 96.3% (53/55) accuracy; 15 of 16 patients with PTL who delivered at term were correctly classified; all patients with PTL without IAI who delivered preterm neonates were correctly identified as such (19/19), while 19/20 patients with PTL and IAI were correctly classified. (2) In the second study, metabolomic profiling was able to identify patients as belonging to the correct clinical group with an accuracy of 88.5% (100/113); 39 of 40 patients with PTL who delivered at term were correctly classified; 29 of 33 patients with PTL without IAI who delivered preterm neonates were correctly classified. Among patients with PTL and IAI, 32/40 were correctly classified. The metabolites responsible for the classification of patients in different clinical groups were identified. A preliminary draft of the human amniotic fluid metabolome was generated and found to contain products of the intermediate metabolism of mammalian cells and xenobiotic compounds (e.g. bacterial products and Salicylamide). CONCLUSION Among patients with spontaneous PTL with intact membranes, metabolic profiling of the amniotic fluid can be used to assess the risk of preterm delivery in the presence or absence of infection/inflammation.
Collapse
Affiliation(s)
- Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
BACKGROUND Research into cell-free fetal (cff) nucleic acids has primarily focused on maternal plasma; however, cff DNA and RNA are also detectable in other body fluids such as amniotic fluid (AF). In AF, cff DNA is present in much greater concentrations than in maternal plasma and represents a pure fetal sample uncontaminated by maternal- and trophoblast-derived nucleic acids. The aim of this review was to summarize the current knowledge on cff nucleic acids in AF and to outline future research directions. METHODS MEDLINE and PREMEDLINE were searched up to August 2010 for original investigations of cell-free RNA or DNA in AF. Sixteen studies were included in the review. RESULTS AF cff DNA represents a physiologically separate pool from cff DNA in maternal plasma. The placenta is not a major source of nucleic acids in AF. It is feasible to isolate cff nucleic acids from small volumes of discarded AF supernatant in sufficient quality and quantity to perform microarray studies and downstream applications such as pathway analysis. This 'discovery-driven approach' has resulted in new information on the pathogenesis of Down syndrome and polyhydramnios. There is otherwise a paucity of information relating to the basic biology and clinical applications of cff nucleic acids in AF. CONCLUSIONS AF supernatant is a valuable and widely available but under-utilized biological resource. Further studies of cff nucleic acids in AF may lead to new insights into human fetal development and ultimately new approaches to antenatal treatment of human disease.
Collapse
Affiliation(s)
- L Hui
- Mother Infant Research Institute at Tufts Medical Center, Boston, MA 02111, USA.
| | | |
Collapse
|
37
|
DiGiulio DB, Gervasi M, Romero R, Mazaki-Tovi S, Vaisbuch E, Kusanovic JP, Seok KS, Gómez R, Mittal P, Gotsch F, Chaiworapongsa T, Oyarzún E, Kim CJ, Relman DA. Microbial invasion of the amniotic cavity in preeclampsia as assessed by cultivation and sequence-based methods. J Perinat Med 2010; 38:503-13. [PMID: 20482470 PMCID: PMC3325506 DOI: 10.1515/jpm.2010.078] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Infection has been implicated in the pathogenesis of preeclampsia, yet the association between microbial invasion of the amniotic cavity (MIAC) and preeclampsia has not been determined. The aim of this study was to determine the prevalence, and microbial diversity associated with MIAC, as well as the nature of the host response to MIAC in patients with preeclampsia. METHOD OF STUDY Amniotic fluid (AF) from 62 subjects with preeclampsia, not in labor, was analyzed with both cultivation and molecular methods. Broad-range and group-specific PCR assays targeting small subunit ribosomal DNA, or other gene sequences, from bacteria, fungi and archaea were used. Results were correlated with measurements of host inflammatory response, including AF white blood cell count and AF concentrations of glucose, interleukin-6 (IL-6) and MMP-8. RESULTS 1) The rate of MIAC in preeclampsia was 1.6% (1/62) based on cultivation techniques, 8% (5/62) based on PCR, and 9.6% (6/62) based on the combined results of both methods; 2) among the six patients diagnosed with MIAC, three had a positive PCR for Sneathia/Leptotrichia spp.; and 3) patients with MIAC were more likely to have evidence of an inflammatory response in the amniotic cavity than those without MIAC, as determined by a higher median AF IL-6 [1.65 ng/mL interquartile range (IQR): 0.35-4.62 vs. 0.22 ng/mL IQR: 0.12-0.51; P=0.002). CONCLUSION The prevalence of MIAC in preeclampsia is low, suggesting that intra-amniotic infection plays only a limited role in preeclampsia. However, the unexpectedly high number of positive AF specimens for Sneathia/Leptotrichia warrants further investigation.
Collapse
MESH Headings
- Adult
- Amnion/microbiology
- Amniotic Fluid/immunology
- Amniotic Fluid/metabolism
- Amniotic Fluid/microbiology
- Base Sequence
- Chorioamnionitis/immunology
- Chorioamnionitis/metabolism
- Chorioamnionitis/microbiology
- Cohort Studies
- DNA Primers/genetics
- DNA, Archaeal/genetics
- DNA, Archaeal/isolation & purification
- DNA, Bacterial/genetics
- DNA, Bacterial/isolation & purification
- DNA, Fungal/genetics
- DNA, Fungal/isolation & purification
- Female
- Humans
- Infant, Newborn
- Inflammation Mediators/metabolism
- Interleukin-6/metabolism
- Matrix Metalloproteinase 8/metabolism
- Microbiological Techniques
- Polymerase Chain Reaction
- Pre-Eclampsia/immunology
- Pre-Eclampsia/microbiology
- Pregnancy
- Pregnancy Complications, Infectious/immunology
- Pregnancy Complications, Infectious/metabolism
- Pregnancy Complications, Infectious/microbiology
- Pregnancy Outcome
- Retrospective Studies
- Young Adult
Collapse
Affiliation(s)
- Daniel B. DiGiulio
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - MariaTeresa Gervasi
- Department of Obstetrics and Gynecology, Azienda Ospedaliera of Padova, Padova, Italy
| | - Roberto Romero
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
| | | | - Ricardo Gómez
- CEDIP (Center for Perinatal Diagnosis and Research), Department of Obstetrics and Gynecology, Sotero del Rio Hospital, Santiago, Chile
- Department of Obstetrics and Gynecology, P. Universidad Católica de Chile, Santiago, Chile
| | - Pooja Mittal
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Hutzel Women’s Hospital, Detroit Medical Center, Detroit, MI, USA
| | - Enrique Oyarzún
- Department of Obstetrics and Gynecology, P. Universidad Católica de Chile, Santiago, Chile
| | - Chong Jai Kim
- Perinatology Research Branch, NICHD, NIH, Bethesda, MD, and Detroit, MI, USA
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - David A. Relman
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| |
Collapse
|
38
|
Chaiworapongsa T, Romero R, Tarca AL, Kusanovic JP, Gotsch F, Mittal P, Kim SK, Vaisbuch E, Mazaki-Tovi S, Erez O, Dong Z, Kim CJ, Yeo L, Hassan SS. A decrease in maternal plasma concentrations of sVEGFR-2 precedes the clinical diagnosis of preeclampsia. Am J Obstet Gynecol 2010; 202:550.e1-10. [PMID: 20510958 PMCID: PMC3437767 DOI: 10.1016/j.ajog.2010.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 05/27/2009] [Accepted: 04/05/2010] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The aim of this study was to examine if maternal plasma concentrations of soluble vascular endothelial growth factor receptor (sVEGFR)-2 change prior to the diagnosis of preeclampsia. STUDY DESIGN A longitudinal study was conducted in normal pregnant women (n = 160) and patients with preeclampsia (n = 40). Blood samples were collected at 7 gestational age intervals from 6 weeks to term. Plasma concentrations of sVEGFR-2 were determined by enzyme-linked immunosorbent assay. Analysis was performed with cross-sectional and longitudinal (mixed effects model) approaches. RESULTS Mothers destined to develop preeclampsia have lower plasma sVEGFR-2 concentrations than those who will have a normal pregnancy (longitudinal approach; P < .05). Cross-sectional analysis suggested that the median plasma sVEGFR-2 concentration in women destined to develop preeclampsia was significantly lower than that in normal pregnant women from 28-31 weeks of gestation (P = .001) or 6-10 weeks prior to the diagnosis (P < .001). CONCLUSION A lower maternal plasma sVEGFR-2 concentration precedes the development of preeclampsia, both term and preterm.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- Perinatology Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chatterjee A, Mirer PL, Zaldivar Santamaria E, Klapperich C, Sharon A, Sauer-Budge AF. RNA Isolation from Mammalian Cells Using Porous Polymer Monoliths: An Approach for High-Throughput Automation. Anal Chem 2010; 82:4344-56. [DOI: 10.1021/ac100063f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Anirban Chatterjee
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| | - Paul L. Mirer
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| | - Elvira Zaldivar Santamaria
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| | - Catherine Klapperich
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| | - Andre Sharon
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| | - Alexis F. Sauer-Budge
- Departments of Mechanical Engineering and Biomedical Engineering, Boston University, Boston, Massachusetts 02215, and Center for Manufacturing Innovation, Fraunhofer USA, Brookline, Massachusetts 02446
| |
Collapse
|