1
|
Devor EJ, Santillan DA, Warrier A, Scroggins SM, Santillan MK. Placenta-specific protein 1 (PLAC1) expression is significantly down-regulated in preeclampsia via a hypoxia-mediated mechanism. J Matern Fetal Neonatal Med 2022; 35:8419-8425. [PMID: 34565269 PMCID: PMC8959068 DOI: 10.1080/14767058.2021.1977792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Examine a mechanism of PLAC1 regulation and its potential role in preeclampsia (PE). MATERIALS AND METHODS Placental tissue samples and detailed clinical information were obtained through the University of Iowa Maternal Fetal Tissue Bank (IRB# 200910784) from gestational and maternal age-matched control (n = 17) and PE affected pregnancies (n = 12). PLAC1 and PLAC1 promoter-specific expression was measured using quantitative polymerase chain reaction (qPCR) and differences were assessed via the standard ΔΔCt method. In addition, the role of hypoxia in PLAC1 transcription was investigated through the exposure of HTR8/SVneo human trophoblast cells to the hypoxia mimic dimethyloxaloylglycine (DMOG). RESULTS PLAC1 expression is seen to be 8.9-fold lower in human placentas affected by preeclampsia in comparison with controls (p < .05). Further, this decrease is paralleled by a significantly lower expression of the P2 or proximal PLAC1 promoter (p < .05). Expression of mediator complex subunit 1 (MED1), a known hypoxia-sensitive transcription coactivator and PLAC1 effector, is significantly correlated with PLAC 1 expression (r2 = 0.607, p < .001). These data suggest that PLAC1 expression is significantly down-regulated in preeclampsia at least in part via a MED1 hypoxia-mediated mechanism. CONCLUSIONS We confirm that PLAC1 transcription is suppressed in the placentae of women affected by preeclampsia. We further demonstrate that this suppression is driven through the P2 or proximal PLAC1 promoter. This demonstration led to the identification of the MED1-TRAP cofactor complex as the hypoxia-sensitive driver.
Collapse
Affiliation(s)
- Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Akshaya Warrier
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sabrina M. Scroggins
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Barrett ES, Corsetti M, Day D, Thurston SW, Loftus CT, Karr CJ, Kannan K, LeWinn KZ, Smith AK, Smith R, Tylavsky FA, Bush NR, Sathyanarayana S. Prenatal phthalate exposure in relation to placental corticotropin releasing hormone (pCRH) in the CANDLE cohort. ENVIRONMENT INTERNATIONAL 2022; 160:107078. [PMID: 35007898 PMCID: PMC8821329 DOI: 10.1016/j.envint.2022.107078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 01/02/2022] [Indexed: 06/05/2023]
Abstract
CONTEXT Phthalates may disrupt maternal-fetal-placental endocrine pathways, affecting pregnancy outcomes and child development. Placental corticotropin releasing hormone (pCRH) is critical for healthy pregnancy and child development, but understudied as a target of endocrine disruption. OBJECTIVE To examine phthalate metabolite concentrations (as mixtures and individually) in relation to pCRH. DESIGN Secondary data analysis from a prospective cohort study. SETTING Prenatal clinics in Tennessee, USA. PATIENTS 1018 pregnant women (61.4% non-Hispanic Black, 32% non-Hispanic White, 6.6% other) participated in the CANDLE study and provided data. Inclusion criteria included: low-medical-risk singleton pregnancy, age 16-40, and gestational weeks 16-29. INTERVENTION None. MAIN OUTCOME MEASURES Plasma pCRH at two visits (mean gestational ages 23.0 and 31.8 weeks) and change in pCRH between visits (ΔpCRH). RESULTS In weighted quantile sums (WQS) regression models, phthalate mixtures were associated with higher pCRH at Visit 1 (β = 0.07, 95 %CI: 0.02, 0.11) but lower pCRH at Visit 2 (β = -0.08, 95 %CI: -0.14, -0.02). In stratified analyses, among women with gestational diabetes (n = 59), phthalate mixtures were associated with lower pCRH at Visit 1 (β = -0.17, 95 %CI: -0.35, 0.0006) and Visit 2 (β = -0.35, 95 %CI: -0.50, -0.19), as well as greater ΔpCRH (β = 0.16, 95 %CI: 0.07, 0.25). Among women with gestational hypertension (n = 102), phthalate mixtures were associated with higher pCRH at Visit 1 (β = 0.20, 95 %CI: 0.03, 0.36) and Visit 2 (β = 0.42; 95 %CI: 0.19, 0.64) and lower ΔpCRH (β = -0.17, 95 %CI: -0.29, -0.06). Significant interactions between individual phthalate metabolites and pregnancy complications were observed. CONCLUSIONS Phthalates may impact placental CRH secretion, with differing effects across pregnancy. Differences in results between women with and without gestational diabetes and gestational hypertension suggest a need for further research examining whether women with pregnancy complications may be more vulnerable to endocrine-disrupting effects of phthalates.
Collapse
Affiliation(s)
- Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA.
| | - Matthew Corsetti
- Department of Biostatistics and Computational Biology, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Drew Day
- Seattle Children's Research Institute, University of Washington, Seattle, WA 98101, USA
| | - Sally W Thurston
- Department of Biostatistics and Computational Biology, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Catherine J Karr
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98104, USA
| | - Kurunthachalam Kannan
- Department of Pediatrics and Department of Environmental Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Roger Smith
- Hunter Medical Research Institute, University of Newcastle, Newcastle 2300, Australia
| | - Frances A Tylavsky
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Sheela Sathyanarayana
- Seattle Children's Research Institute, University of Washington, Seattle, WA 98101, USA; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|
3
|
Devor EJ, Schickling BM, Lapierre JR, Bender DP, Gonzalez-Bosquet J, Leslie KK. The Synthetic Curcumin Analog HO-3867 Rescues Suppression of PLAC1 Expression in Ovarian Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14090942. [PMID: 34577642 PMCID: PMC8465575 DOI: 10.3390/ph14090942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/06/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
Elevated expression of placenta-specific protein 1 (PLAC1) is associated with the increased proliferation and invasiveness of a variety of human cancers, including ovarian cancer. Recent studies have shown that the tumor suppressor p53 directly suppresses PLAC1 transcription. However, mutations in p53 lead to the loss of PLAC1 transcriptional suppression. Small molecules that structurally convert mutant p53 proteins to wild-type conformations are emerging. Our objective was to determine whether the restoration of the wild-type function of mutated p53 could rescue PLAC1 transcriptional suppression in tumors harboring certain TP53 mutations. Ovarian cancer cells OVCAR3 and ES-2, both harboring TP53 missense mutations, were treated with the p53 reactivator HO-3867. Treatment with HO-3867 successfully rescued PLAC1 transcriptional suppression. In addition, cell proliferation was inhibited and cell death through apoptosis was increased in both cell lines. We conclude that the use of HO-3867 as an adjuvant to conventional therapeutics in ovarian cancers harboring TP53 missense mutations could improve patient outcomes. Validation of this conclusion must, however, come from an appropriately designed clinical trial.
Collapse
Affiliation(s)
- Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Correspondence:
| | - Brandon M. Schickling
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
| | - Jace R. Lapierre
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
| | - David P. Bender
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Jesus Gonzalez-Bosquet
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Kimberly K. Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA; (B.M.S.); (J.R.L.); (D.P.B.); (J.G.-B.); (K.K.L.)
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
4
|
Integrated bioinformatics analysis reveals novel key biomarkers and potential candidate small molecule drugs in gestational diabetes mellitus. Biosci Rep 2021; 41:228450. [PMID: 33890634 PMCID: PMC8145272 DOI: 10.1042/bsr20210617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is the metabolic disorder that appears during pregnancy. The current investigation aimed to identify central differentially expressed genes (DEGs) in GDM. The transcription profiling by array data (E-MTAB-6418) was obtained from the ArrayExpress database. The DEGs between GDM samples and non-GDM samples were analyzed. Functional enrichment analysis were performed using ToppGene. Then we constructed the protein–protein interaction (PPI) network of DEGs by the Search Tool for the Retrieval of Interacting Genes database (STRING) and module analysis was performed. Subsequently, we constructed the miRNA–hub gene network and TF–hub gene regulatory network. The validation of hub genes was performed through receiver operating characteristic curve (ROC). Finally, the candidate small molecules as potential drugs to treat GDM were predicted by using molecular docking. Through transcription profiling by array data, a total of 869 DEGs were detected including 439 up-regulated and 430 down-regulated genes. Functional enrichment analysis showed these DEGs were mainly enriched in reproduction, cell adhesion, cell surface interactions at the vascular wall and extracellular matrix organization. Ten genes, HSP90AA1, EGFR, RPS13, RBX1, PAK1, FYN, ABL1, SMAD3, STAT3 and PRKCA were associated with GDM, according to ROC analysis. Finally, the most significant small molecules were predicted based on molecular docking. This investigation identified hub genes, signal pathways and therapeutic agents, which might help us, enhance our understanding of the mechanisms of GDM and find some novel therapeutic agents for GDM.
Collapse
|
5
|
Yilmaz N, Timur H, Ugurlu EN, Yilmaz S, Ozgu-Erdinc AS, Erkilinc S, Inal HA. Placenta specific protein-1 in recurrent pregnancy loss and in In Vitro Fertilisation failure: a prospective observational case-control study. J OBSTET GYNAECOL 2019; 40:843-848. [PMID: 31791163 DOI: 10.1080/01443615.2019.1674263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Observations from studies have provided evidence that Placenta-specific protein1 (PLAC1) is important for the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. The aim of this study is to investigate whether maternal serum PLAC1 levels have any impact on etiopathogenesis of recurrent pregnancy loss (RPL) and repeated implantation failure after In Vitro Fertilisation (RIF). We conducted a prospective observational case-control study in a Research Hospital. Twenty-eight patients with RPL (group 1), 30 patients with unexplained infertility and RIF (group 2), 29 fertile patients (group 3) were included. The demographic features and serum PLAC1 levels were compared. There was a significant difference in PLAC1 levels between the groups (group 1 = 19.71 + 16.55 ng/ml; group 2 = 4.82 + 1.44 ng/ml; group 3 = 0.89 + 0.62 ng/ml, respectively) (p=.001). Positive correlation was found between serum PLAC1 levels and abortion rates (r = 0.64; p=.001), a negative correlation was found between serum PLAC1 levels and live birth rates (r = -0.69; p=.001). PLAC1 might have a negative effect on implantation in RPL and RIF. There may be a subgroup of PLAC with different bioactivity. There are no relevant studies conducted among these populations, further large-scale studies are needed to assess the molecular role of PLAC1 on implantation.IMPACT STATEMENTWhat is already known about this subject? PLAC1 (placenta-specific protein-1) gene is located on the X chromosome which encodes for a protein that is thought to be important for placental development although its role has not been clearly defined. Studies in the literature have provided evidence that PLAC1 has an important role in the establishment and maintenance of pregnancy and suggest it as a potential biomarker for gestational pathologies. Several reports over the past few years have demonstrated PLAC1 expression in a variety of human tumours including lung cancers, breast cancer, hepatocellular and colorectal cancers, gastric cancers and uterine cancers.What do the results of this study add? There have been no previous studies conducted among patients with recurrent pregnancy loss (RPL) or repeated implantation failure after In Vitro Fertilisation (RIF) that have searched for any association between PLAC1 levels and implantation failure. This study has demonstrated higher PLAC1 levels in infertile women with RIF and RPL for the first time; suggesting that it could have a negative effect on implantation in these populations. PLAC1 could be detected in the serum as a biomarker that is associated with RIF and RPL. What are the implications of these findings for clinical practice and/or further research? Defining the precise role of PLAC1 during implantation will provide new insight into understanding of poor reproductive outcomes such as RIF and RPL and help in developing treatment strategies. Further large-scale studies with more patients are needed to uncover the clinical value of PLAC1 as a biomarker to predict repeated implantation failure and RPL.
Collapse
Affiliation(s)
- Nafiye Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hakan Timur
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Evin Nil Ugurlu
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Saynur Yilmaz
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - A Seval Ozgu-Erdinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Selcuk Erkilinc
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| | - Hasan Ali Inal
- Dr. Zekai Tahir Burak Women's Health Research and Education Hospital, Ankara, Turkey
| |
Collapse
|
6
|
Wan L, Sun D, Xie J, Du M, Wang P, Wang M, Lei Y, Wang H, Wang H, Dong M. Declined placental PLAC1 expression is involved in preeclampsia. Medicine (Baltimore) 2019; 98:e17676. [PMID: 31689783 PMCID: PMC6946281 DOI: 10.1097/md.0000000000017676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND This study aimed to clarify the change of the expression of placenta-specific 1 (PLAC1) in the placenta of preeclamptic women and to explore the regulatory effects on thophoblast by PLAC1. METHODS Nineteen women with preeclampsia and 19 with normal pregnancies were recruited, and then we determined the expression of PLAC1 by immunohistochemistry (IHC) and Western blotting. To observe the effect of hypoxia on the expression of PLAC1, trophoblasts were cultured at the normoxia or hypoxia condition. Small interference of ribonucleic acid (siRNA) was used to silence PLAC1. The proliferation, migration and invasion of trophoblasts were evaluated with cell counting kit-8 and transwell analysis, and the apoptosis of trophoblast was evaluated by flow cytometry with FITC and PI staining. RESULTS Placental PLAC1 expression was significantly decreased in severe preeclampsia compared with control (P < .001). The expression of PLAC1 in trophoblasts was significantly decreased after treated with low oxygen concentration (P = .018). PLAC1 siRNA significantly inhibited the proliferation (P < .001), the migration (P < .001) and invasion (P < .001) of trophoblasts, but increased the apoptosis (P = .004 for Swan-71; P = .031 for Jar). CONCLUSIONS The expression of PLAC1 was declined in preeclampsia and this inhibited the function of trophoblast, suggesting PLAC1 may play a role in the development of preeclampsia.
Collapse
Affiliation(s)
- Liuxia Wan
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Dandan Sun
- Women's Hospital, School of Medicine, Zhejiang University
- Jiaxing Maternal and Child Health-Care Center
| | - Jiamin Xie
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Mengkai Du
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Peng Wang
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Miaomiao Wang
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Yu Lei
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Huihua Wang
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of women's Reproductive Health of Zhejiang Province
- The First People's Hospital of Tongxiang City, China
| | - Hanzhi Wang
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| | - Minyue Dong
- Women's Hospital, School of Medicine, Zhejiang University
- Key Laboratory of Reproductive Genetics, Ministry of Education
- Key Laboratory of women's Reproductive Health of Zhejiang Province
| |
Collapse
|
7
|
Mahmoudian J, Ghods R, Nazari M, Jeddi-Tehrani M, Ghahremani MH, Ghaffari-Tabrizi-Wizsy N, Ostad SN, Zarnani AH. PLAC1: biology and potential application in cancer immunotherapy. Cancer Immunol Immunother 2019; 68:1039-1058. [PMID: 31165204 PMCID: PMC11028298 DOI: 10.1007/s00262-019-02350-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/24/2019] [Indexed: 12/29/2022]
Abstract
The emergence of immunotherapy has revolutionized medical oncology with unprecedented advances in cancer treatment over the past two decades. However, a major obstacle in cancer immunotherapy is identifying appropriate tumor-specific antigens to make targeted therapy achievable with fewer normal cells being impaired. The similarity between placentation and tumor development and growth has inspired many investigators to discover antigens for effective immunotherapy of cancers. Placenta-specific 1 (PLAC1) is one of the recently discovered placental antigens with limited normal tissue expression and fundamental roles in placental function and development. There is a growing body of evidence showing that PLAC1 is frequently activated in a wide variety of cancer types and promotes cancer progression. Based on the restricted expression of PLAC1 in testis, placenta and a wide variety of cancers, we have designated this molecule with new terminology, cancer-testis-placenta (CTP) antigen, a feature that PLAC1 shares with many other cancer testis antigens. Recent reports from our lab provide compelling evidence on the preferential expression of PLAC1 in prostate cancer and its potential utility in prostate cancer immunotherapy. PLAC1 may be regarded as a potential CTP antigen for targeted cancer immunotherapy based on the available data on its promoting function in cancer development and also its expression in cancers of different histological origin. In this review, we will summarize current data on PLAC1 with emphasis on its association with cancer development and immunotherapy.
Collapse
Affiliation(s)
- Jafar Mahmoudian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran
| | | | - Seyed Nasser Ostad
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Pharmacology Building, Enghelab St., Tehran, 1417614411, Iran.
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Nafisi Building, Enghelab St., Tehran, 1417613151, Iran.
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Etesami E, Nikukar H, Ramezanali F, Ghotbizadeh Vahdani F, Shahhoseini M, Favaedi R, Ghaheri A, Seydabadi S, Zamanian M. Gene expression analysis of MMPs in women with preeclampsia using cell-free fetal RNA in maternal plasma. Pregnancy Hypertens 2019; 17:261-268. [PMID: 31487650 DOI: 10.1016/j.preghy.2019.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Nucleic acids released from the placenta into the mother's blood circulation system provide a valuable source of potential biomarkers for early detection of pregnancy complications such as preeclampsia (PE). PE affects nearly 5-10% of pregnancies worldwide and is a major contributor to the maternal and neonatal mortality and morbidity. It is known that altered placental expression of matrix metalloproteinases (MMPs) may cause shallow cytotrophoblastic invasion and ultimately lead to preeclampsia. The present study aimed to evaluate pattern of placental/fetal expression of the MMP family (MMP-2, MMP-9, MMP-14, MMP-15 and MMP-26) in preeclamptic women and compare it to normal pregnancies, using cell free fetal RNA (cff-RNA). METHODS Blood samples were obtained from 20 pregnant women diagnosed with severe PE (28-32 weeks) and 40 control healthy pregnant women in two groups of either matched gestational age (N = 20) or 14 and 28 weeks pregnancies (each 10). cff-RNA was extracted from plasma, followed by reverse transcription of cff-RNA. Expression of MMP genes was measured using quantitative reverse transcription PCR (qRT-PCR). RESULTS The expression levels of MMP-2, MMP-9 and MMP-15 were significantly increased, while MMP-14 expression level was significantly reduced and the expression of MMP-26 showed a relative increase in PE pregnancies compared to the control group. Additionally, increased level of MMPs expression was observed by comparing 14 and 28 weeks gestation age in normal pregnancy. CONCLUSION Using cff-RNA, circulatory expression level of MMP-2, MMP-9, MMP-14 and MMP-15 were significantly altered in preeclampsia compared to normal pregnancies.
Collapse
Affiliation(s)
- Elham Etesami
- Department of Stem Cell Biology, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Habib Nikukar
- Department of Stem Cell Biology, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fariba Ramezanali
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Fahimeh Ghotbizadeh Vahdani
- Obstetrics and Gynecology Department, Imam Khomeini Medical Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shahhoseini
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Raha Favaedi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Azadeh Ghaheri
- Department of Epidemiology and Reproductive Health, Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Sara Seydabadi
- Department of Stem Cell Biology, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohammadreza Zamanian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
9
|
Shi LY, Ma Y, Zhu GY, Liu JW, Zhou CX, Chen LJ, Wang Y, Li RC, Yang ZX, Zhang D. Placenta‐specific 1 regulates oocyte meiosis and fertilization through furin. FASEB J 2018; 32:5483-5494. [DOI: 10.1096/fj.201700922rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Li-Ya Shi
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Yang Ma
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Gang-Yi Zhu
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Jin-Wei Liu
- Department of GynecologyZhejiang Provincial People's HospitalHangzhouChina
| | - Chun-Xiang Zhou
- Prenatal Diagnosis Center of Jiangsu ProvinceAffiliated Drum Tower Hospital, Nanjing University Medical SchoolNanjingChina
| | - Liang-Jian Chen
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Yang Wang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | | | - Zhi-Xia Yang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Dong Zhang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
10
|
Sifakis S, Androutsopoulos VP, Pontikaki A, Velegrakis A, Papaioannou GI, Koukoura O, Spandidos DA, Papantoniou N. Placental expression of PAPPA, PAPPA-2 and PLAC-1 in pregnacies is associated with FGR. Mol Med Rep 2018. [PMID: 29532882 PMCID: PMC5928614 DOI: 10.3892/mmr.2018.8721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Fetal growth restriction (FGR) is a gynecological disorder of varying etiology. In the present study, an expression analysis of pregnancy-associated plasma protein A (PAPPA), pregnancy-associated plasma protein A2 (PAPPA2) and placenta-specific-1 (PLAC-1) was conducted in pregnancies with FGR and control pregnancies. Placental tissues were collected from pregnancies with FGR (n=16) and control pregnancies (n=16) and the expression of the genes of interest was examined by qPCR. The mean expression levels of PAPPA and PAPPA2 were significantly lower (P<0.001) in placental tissues from FGR pregnancies compared with tissues from healthy subjects, whereas the opposite pattern was observed for PLAC-1 (P<0.001). PAPPA and PLAC-1 expression in FGR and control subjects correlated with birth weight (P<0.001). The findings suggest a possible pathophysiological link between the development of FGR and the expression of PAPPA, PAPPA2 and PLAC-1.
Collapse
Affiliation(s)
- Stavros Sifakis
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | | | - Artemis Pontikaki
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Alexis Velegrakis
- Department of Obstetrics and Gynecology, Venizeleion Hospital, 71409 Heraklion, Greece
| | - George I Papaioannou
- Department of Obstetrics and Gynecology, Attikon University Hospital, University of Athens, 12462 Athens, Greece
| | - Ourania Koukoura
- Department of Obstetrics and Gynecology, University of Thessalia, 41110 Larissa, Greece
| | - Demetrios A Spandidos
- Department of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Nikos Papantoniou
- Department of Obstetrics and Gynecology, Attikon University Hospital, University of Athens, 12462 Athens, Greece
| |
Collapse
|
11
|
Fallen S, Baxter D, Wu X, Kim TK, Shynlova O, Lee MY, Scherler K, Lye S, Hood L, Wang K. Extracellular vesicle RNAs reflect placenta dysfunction and are a biomarker source for preterm labour. J Cell Mol Med 2018. [PMID: 29516617 PMCID: PMC5908130 DOI: 10.1111/jcmm.13570] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Preterm birth (PTB) can lead to lifelong complications and challenges. Identifying and monitoring molecular signals in easily accessible biological samples that can diagnose or predict the risk of preterm labour (PTL) in pregnant women will reduce or prevent PTBs. A number of studies identified putative biomarkers for PTL including protein, miRNA and hormones from various body fluids. However, biomarkers identified from these studies usually lack consistency and reproducibility. Extracellular vesicles (EVs) in circulation have gained significant interest in recent years as these vesicles may be involved in cell‐cell communication. We have used an improved small RNA library construction protocol and a newly developed size exclusion chromatography (SEC)‐based EV purification method to gain a comprehensive view of circulating RNA in plasma and its distribution by analysing RNAs in whole plasma and EV‐associated and EV‐depleted plasma. We identified a number of miRNAs in EVs that can be used as biomarkers for PTL, and these miRNAs may reflect the pathological changes of the placenta during the development of PTL. To our knowledge, this is the first study to report a comprehensive picture of circulating RNA, including RNA in whole plasma, EV and EV‐depleted plasma, in PTL and reveal the usefulness of EV‐associated RNAs in disease diagnosis.
Collapse
Affiliation(s)
| | | | - Xiaogang Wu
- Institute for Systems Biology, Seattle, WA, USA
| | | | - Oksana Shynlova
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | | | - Stephen Lye
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Obstetrics & Gynecology, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA, USA
| |
Collapse
|
12
|
Abstract
Hypertensive disorders in pregnancy have been the cause of much clinical dilemma, affecting up to 10 % of all pregnancies. The precise blood pressure to achieve in a pregnant woman is usually a battle between minimizing end organ damage to the mother and providing adequate perfusion to the placenta and the fetus. This predicament is becoming more, not less, frequent as maternal ages increase in high resource nations. Biomarkers to predict preeclampsia, a subcategory of hypertension in pregnancy, have always been elusive. The discovery of angiogenic factors relevant to preeclampsia in the last decade, however, has propelled much needed research, both in the basic science and clinical arenas. In this review, we summarize the latest clinical studies and international guidelines on blood pressure goals in pregnancy, and discuss the most promising of biomarkers to predict or diagnose preeclampsia.
Collapse
|
13
|
Measuring circulating placental RNAs to non-invasively assess the placental transcriptome and to predict pregnancy complications. Prenat Diagn 2016; 36:997-1008. [DOI: 10.1002/pd.4934] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/08/2016] [Accepted: 09/30/2016] [Indexed: 11/07/2022]
|
14
|
Chang WL, Wang H, Cui L, Peng NN, Fan X, Xue LQ, Yang Q. PLAC1 is involved in human trophoblast syncytialization. Reprod Biol 2016; 16:218-224. [PMID: 27692364 DOI: 10.1016/j.repbio.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022]
Abstract
Placenta specific protein 1 (PLAC1) is thought to be important for murine and human placentation because of its abundant expression in placenta; however, the trophoblast subtypes that express PLAC1 at the fetomaternal interface and the major role of PLAC1 in placentation are still unclear. This study investigated the expression pattern of PLAC1 at the human fetomaternal interface and its involvement in trophoblast syncytialization. Localization of PLAC1 at the fetomaternal interface was studied using in situ hybridization (ISH) and immunohistochemistry (IHC) assays. Real time RT-PCR and Western Blot were employed to exhibit the expression pattern of PLAC1 during human spontaneous syncytialization of term primary cytotrophoblast cells (CTBs). Spontaneous syncytialization of a primary term CTBs model transfected with siRNA specific to PLAC1 was used to investigate the role of PLAC1 during human trophoblast syncytialization. The results showed that PLAC1 was mainly expressed in the human villous syncytiotrophoblast (STB) layer throughout gestation, and the expression level of PLAC1 was significantly elevated during human trophoblast syncytialization. Down-regulation of PLAC1 via specific PLAC1 siRNA transfection attenuated spontaneous syncytialization of primary term CTBs (p<0.05) as indicated by cell fusion index and the expression patterns of the corresponding markers. These data demonstrate the facilitative role of PLAC1 in normal human trophoblast syncytialization.
Collapse
Affiliation(s)
- Wen-Lin Chang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China; State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKUHKUST Medical Center, Shenzhen, China
| | - Huiying Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Lina Cui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Graduate School of Chinese Academy of Sciences, Beijing, China
| | - Nan-Ni Peng
- Reproductive Medical Center of Luohu Hospital Shenzhen, Shenzhen, Guangdong, China
| | - Xiujun Fan
- Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Li-Qun Xue
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| | - Qing Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
15
|
Manokhina I, Wilson SL, Robinson WP. Noninvasive nucleic acid-based approaches to monitor placental health and predict pregnancy-related complications. Am J Obstet Gynecol 2015; 213:S197-206. [PMID: 26428499 DOI: 10.1016/j.ajog.2015.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/11/2015] [Accepted: 07/13/2015] [Indexed: 12/18/2022]
Abstract
During pregnancy, the placenta releases a variety of nucleic acids (including deoxyribonucleic acid, messenger ribonucleic acid, or microribonucleic acids) either as a result of cell turnover or as an active messaging system between the placenta and cells in the maternal body. The profile of released nucleic acids changes with the gestational age and has been associated with maternal and fetal parameters. It also can directly reflect pathological changes in the placenta. Nucleic acids may therefore provide a rich source of novel biomarkers for the prediction of pregnancy complications. However, their utility in the clinical setting depends, first, on overcoming some technical considerations in their quantification, and, second, on developing a better understanding of the factors that influence their function and abundance.
Collapse
Affiliation(s)
- Irina Manokhina
- Child and Family Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Samantha L Wilson
- Child and Family Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- Child and Family Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
16
|
Song J, Li Y, An RF. Identification of Early-Onset Preeclampsia-Related Genes and MicroRNAs by Bioinformatics Approaches. Reprod Sci 2015; 22:954-63. [PMID: 25717061 DOI: 10.1177/1933719115570898] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Early-onset preeclampsia (EOPET), resulting in intrauterine growth restriction, has serious impact on maternal, perinatal, and neonatal livability worldwide. The current study conducted bioinformatics analyses to screen key genes and microRNAs (miRNAs) involved in EOPET and thus to explore the molecular mechanisms of EOPET. METHODS The microarray data set GSE44711 containing 8 EOPET placentas and 8 gestational age-matched controls was obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened and subjected to gene ontology functional enrichment analysis. Then, Human Protein Reference Database was used to construct protein-protein interaction (PPI) network of DEGs. Besides, we predicted EOPET-associated miRNAs and built the miRNA regulatory network. RESULTS A total of 150 DEGs including 26 upregulated and 124 downregulated genes were obtained. Corticotropin releasing hormone (CRH) and vitronectin (VTN) was the most significantly upregulated and downregulated genes, respectively. The DEGs were mainly related to the biological process (BP) of pregnancy, hormone-involved process, and formation of extracellular region. Analysis of PPI network revealed that fibronectin 1(FN1), FBJ murine osteosarcoma viral oncogene homolog (FOS), CD247 molecule (CD247), VTN, and interleukin 2 receptor, beta (IL2RB) were the top 5 DEGs with highest node degree. Furthermore, many EOPET-associated miRNAs were identified and miR-142-3p was the most significant one. Additionally, multiple miRNAs, such as miR-200b/c and miR-27a/b, were predicted to regulate the expression of several key DEGs. CONCLUSION The current study identified several regulators in EOPET, which may contribute to interpret the molecular mechanism of EOPET and develop novel biomarkers and therapeutic targets for EOPET.
Collapse
Affiliation(s)
- Jing Song
- Obstetrics & Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shan xi, China Obstetrics & Gynecology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Li
- Obstetrics & Gynecology, the Hospital of Heilongjiang Province, Harbin, People's Republic of China
| | - Rui Fang An
- Obstetrics & Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shan xi, China
| |
Collapse
|
17
|
Zhao L, Blackburn J, Brosseau CL. Quantitative Detection of Uric Acid by Electrochemical-Surface Enhanced Raman Spectroscopy Using a Multilayered Au/Ag Substrate. Anal Chem 2014; 87:441-7. [DOI: 10.1021/ac503967s] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Lili Zhao
- Department
of Chemistry, Saint Mary’s University, Halifax, Nova Scotia B3H 3C3, Canada
| | - Jonathan Blackburn
- Institute
of Infectious Disease and
Molecular Medicine and Division of Medical Biochemistry, Faculty of
Health Sciences, University of Cape Town, Cape Town, 7925 South Africa
| | - Christa L. Brosseau
- Department
of Chemistry, Saint Mary’s University, Halifax, Nova Scotia B3H 3C3, Canada
| |
Collapse
|
18
|
Chaiworapongsa T, Chaemsaithong P, Korzeniewski SJ, Yeo L, Romero R. Pre-eclampsia part 2: prediction, prevention and management. Nat Rev Nephrol 2014; 10:531-40. [PMID: 25003612 PMCID: PMC5898797 DOI: 10.1038/nrneph.2014.103] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An antiangiogenic state might constitute a terminal pathway for the multiple aetiologies of pre-eclampsia, especially those resulting from placental abnormalities. The levels of angiogenic and antiangiogenic proteins in maternal blood change prior to a diagnosis of pre-eclampsia, correlate with disease severity and have prognostic value in identifying women who will develop maternal and/or perinatal complications. Potential interventions exist to ameliorate the imbalance of angiogenesis and, hence, might provide opportunities to improve maternal and/or perinatal outcomes in pre-eclampsia. Current strategies for managing pre-eclampsia consist of controlling hypertension, preventing seizures and timely delivery of the fetus. Prediction of pre-eclampsia in the first trimester is of great interest, as early administration of aspirin might reduce the risk of pre-eclampsia, albeit modestly. Combinations of biomarkers typically predict pre-eclampsia better than single biomarkers; however, the encouraging initial results of biomarker studies require external validation in other populations before they can be used to facilitate intervention in patients identified as at increased risk. Angiogenic and antiangiogenic factors might also be useful in triage of symptomatic patients with suspected pre-eclampsia, differentiating pre-eclampsia from exacerbations of pre-existing medical conditions and performing risk assessment in asymptomatic women. This Review article discusses the performance of predictive and prognostic biomarkers for pre-eclampsia, current strategies for preventing and managing the condition and its long-term consequences.
Collapse
Affiliation(s)
- Tinnakorn Chaiworapongsa
- 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, 31 Center Drive, Bethesda, MD 20892, USA and 3990 John R Street, Detroit, MI 48201, USA
| | - Piya Chaemsaithong
- 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, 31 Center Drive, Bethesda, MD 20892, USA and 3990 John R Street, Detroit, MI 48201, USA
| | - Steven J Korzeniewski
- 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, 31 Center Drive, Bethesda, MD 20892, USA and 3990 John R Street, Detroit, MI 48201, USA
| | - Lami Yeo
- 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, 31 Center Drive, Bethesda, MD 20892, USA and 3990 John R Street, Detroit, MI 48201, USA
| | - 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, 31 Center Drive, Bethesda, MD 20892, USA and 3990 John R Street, Detroit, MI 48201, USA
| |
Collapse
|
19
|
Chang WL, Yang Q, Zhang H, Lin HY, Zhou Z, Lu X, Zhu C, Xue LQ, Wang H. Role of placenta-specific protein 1 in trophoblast invasion and migration. Reproduction 2014; 148:343-52. [PMID: 24989904 DOI: 10.1530/rep-14-0052] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Placenta-specific protein 1 (PLAC1), a placenta-specific gene, is known to be involved in the development of placenta in both humans and mice. However, the precise role of PLAC1 in placental trophoblast function remains unclear. In this study, the localization of PLAC1 in human placental tissues and its physiological significance in trophoblast invasion and migration are investigated by technical studies including real-time RT-PCR, in situ hybridization, immunohistochemistry, and functional studies by utilizing cell invasion and migration assays in the trophoblast cell line HTR8/SVneo as well as the primary inducing extravillous trophoblasts (EVTs). The results show that PLAC1 is mainly detected in the trophoblast columns and syncytiotrophoblast of the first-trimester human placental villi, as well as in the EVTs that invade into the maternal decidua. Knockdown of PLAC1 by RNA interference significantly suppresses the invasion and migration of HTR8/SVneo cells and shortens the distance of the outgrowth of the induced EVTs from the cytotrophoblast column of the explants. All the above data suggests that PLAC1 plays an important role in human placental trophoblast invasion and migration.
Collapse
Affiliation(s)
- Wen-Lin Chang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Qing Yang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hui Zhang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hai-Yan Lin
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Zhi Zhou
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Xiaoyin Lu
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Cheng Zhu
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Li-Qun Xue
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| | - Hongmei Wang
- College of Veterinary MedicineHunan Agricultural University, Changsha 410128, ChinaState Key Laboratory of Reproductive BiologyInstitute of Zoology, Chinese Academy of Sciences, Beijing 100101, ChinaBeijing Obstetrics and Gynecology HospitalCapital Medical University, Beijing 100069, ChinaGraduate School of Chinese Academy of SciencesBeijing 100039, China
| |
Collapse
|
20
|
Farina A. The Role of RNAs and microRNAs in Non-Invasive Prenatal Diagnosis. J Clin Med 2014; 3:440-52. [PMID: 26237384 PMCID: PMC4449680 DOI: 10.3390/jcm3020440] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 02/17/2014] [Accepted: 03/10/2014] [Indexed: 01/11/2023] Open
Abstract
In this paper, all possible clinical applications of circulating mRNA and miRNA for non-invasive prenatal diagnosis appearing in the medical literature so far are described. Data from the literature have also been reported and commented on along with some possible future applications.
Collapse
Affiliation(s)
- Antonio Farina
- Department of Medicine and Surgery (DIMEC) Division of Prenatal Medicine, University of Bologna, Bologna 40138, Italy.
| |
Collapse
|
21
|
Noninvasive in vivo monitoring of tissue-specific global gene expression in humans. Proc Natl Acad Sci U S A 2014; 111:7361-6. [PMID: 24799715 DOI: 10.1073/pnas.1405528111] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Circulating cell-free RNA in the blood provides a potential window into the health, phenotype, and developmental programs of a variety of human organs. We used high-throughput methods of RNA analysis such as microarrays and next-generation sequencing to characterize the global landscape circulating RNA in a cohort of human subjects. By focusing on genes whose expression is highly specific to certain tissues, we were able to identify the relative contributions of these tissues to circulating RNA and to monitor changes in tissue development and health. As one application of this approach, we performed a longitudinal study on pregnant women and analyzed their combined cell-free RNA transcriptomes across all three trimesters of pregnancy and after delivery. In addition to the analysis of mRNA, we observed and characterized noncoding species such as long noncoding RNA and circular RNA transcripts whose presence had not been previously observed in human plasma. We demonstrate that it is possible to track specific longitudinal phenotypic changes in both the mother and the fetus and that it is possible to directly measure transcripts from a variety of fetal tissues in the maternal blood sample. We also studied the role of neuron-specific transcripts in the blood of healthy adults and those suffering from the neurodegenerative disorder Alzheimer's disease and showed that disease specific neural transcripts are present at increased levels in the blood of affected individuals. Characterization of the cell-free transcriptome in its entirety may thus provide broad insights into human health and development without the need for invasive tissue sampling.
Collapse
|
22
|
Inversetti A, Smid M, Candiani M, Ferrari M, Galbiati S. Predictive biomarkers of pre-eclampsia and effectiveness of preventative interventions for the disease. Expert Opin Biol Ther 2014; 14:1161-73. [PMID: 24766211 DOI: 10.1517/14712598.2014.912271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Pre-eclampsia (PE) is one of the most common pregnancy complication characterized by placental and maternal vascular dysfunction. It affects about 3 - 8% of women during the second half of pregnancy and represents one of the major causes of neonatal morbidity and mortality. The etiology of PE largely remains unknown. AREAS COVERED PE is considered a syndrome with multisystem involvement, so the ideal predictive test for it should utilize a combination of many predictors. Measurement in early pregnancy of a variety of biophysical and biochemical markers implicated in the pathophysiology of PE associated with clinical risk factors has been proposed to predict the development of the syndrome, thereby mitigating an adverse outcome. EXPERT OPINION The identification of reliable indicators is a clinically relevant issue that could result in early therapeutic intervention and leading to the prevention of maternal and fetal injuries before the manifestation of clinical signs. Many factors complicate the prevention of PE cases. Most are attributed to unknown etiology, the low predictive value of current screening tests and the several presentations of the disease. Although preventative treatments have been studied extensively, an effective intervention to avoid the development of PE has not yet been discovered.
Collapse
Affiliation(s)
- Annalisa Inversetti
- San Raffaele Hospital, Department of Obstetrics and Gynecology , Via Olgettina 60, Milan 20132 , Italy
| | | | | | | | | |
Collapse
|
23
|
Placenta-specific protein 1: a potential key to many oncofetal-placental OB/GYN research questions. Obstet Gynecol Int 2014; 2014:678984. [PMID: 24757447 PMCID: PMC3976915 DOI: 10.1155/2014/678984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/23/2014] [Accepted: 02/10/2014] [Indexed: 12/19/2022] Open
Abstract
Placenta-specific protein 1 (PLAC1) is a secreted protein found in trophoblasts. Several reports implicate a central role for PLAC1 in establishment and maintenance of the placenta. In addition to placentae PLAC1 is expressed in a variety of solids including breast, endometrial, and ovarian cancers. In order to show that PLAC1 is potentially relevant to a number of research questions in OB/GYN, we report on PLAC1 expression in a selected panel that includes two choriocarcinoma cell lines, normal placental tissues, and endometrial and ovarian tumors. We report for the first time that PLAC1 is also expressed in human fetal tissues. PLAC1 is transcriptionally heterogeneous with one promoter (P1) generating two transcripts with alternately spliced 5' UTRs and the other promoter (P2) generating a third transcript. Placental tissues favor P2 transcripts, while P1 is favored in most of the other cells. Mechanisms determining multiple PLAC1 transcripts and promoter preferences are as yet unknown, but it is clear that this protein is likely to be important in a variety of phenomena relevant to both gynecologic oncology and maternal-fetal medicine.
Collapse
|
24
|
Fant ME, Fuentes J, Kong X, Jackman S. The nexus of prematurity, birth defects, and intrauterine growth restriction: a role for plac1-regulated pathways. Front Pediatr 2014; 2:8. [PMID: 24600606 PMCID: PMC3930911 DOI: 10.3389/fped.2014.00008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/24/2014] [Indexed: 12/28/2022] Open
Abstract
Epidemiological studies have demonstrated an increased prevalence of birth defects and intrauterine growth restriction (IUGR) among infants born prematurely suggesting they share common biological determinants. The identification of key regulatory pathways contributing to this nexus is essential to ongoing efforts to develop effective intervention strategies. Plac1 is a paternally imprinted and X-linked gene that conforms to this paradigm. Examination of a mutant mouse model has confirmed that Plac1 is essential for normal placental development and function. Moreover, it is expressed throughout the developing embryo indicating that it also has broad relevance to embryogenesis. Most notably, its absence in the developing embryo is associated with abnormal brain development and an increased risk of lethal, postnatal hydrocephalus identifying it as a novel, X-linked determinant of brain development. The essential and non-redundant roles of Plac1 in placental and neurological development represent a novel regulatory paradigm for embryonic growth and pregnancy maintenance. Regulatory pathways influenced, in part, by Plac1 are likely to contribute to the observed nexus of IUGR, prematurity, and birth defects.
Collapse
Affiliation(s)
- Michael E Fant
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA ; Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida , Tampa, FL , USA ; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Juan Fuentes
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Xiaoyuan Kong
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| | - Suzanne Jackman
- Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, FL , USA
| |
Collapse
|
25
|
Rodriguez-Prado YM, Kong X, Fant ME. PLAC1 Expression Decreases in Chorionic Villi in Response to Labor. ISRN OBSTETRICS AND GYNECOLOGY 2013; 2013:704252. [PMID: 23840959 PMCID: PMC3693165 DOI: 10.1155/2013/704252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/26/2013] [Indexed: 11/17/2022]
Abstract
PLAC1 (Placenta-Specific 1) is a recently described, trophoblast-expressed gene essential for normal placental development. The protein localizes to the microvillus membrane surface of the syncytiotrophoblast in direct proximity to the maternal compartment. Although its role has not been defined, increased circulating levels of human PLAC1 mRNA in maternal blood are associated with preeclampsia. Furthermore, PLAC1-null mice exhibit decreased viability in the peripartum period suggesting a role in pregnancy maintenance late in gestation. We examined PLAC1 gene expression in the human placenta during normal pregnancy and pregnancies associated with maternal diabetes and preeclampsia using quantitative, real time PCR (q-RT-PCR). Although there was no apparent difference in PLAC1 gene expression among human pregnancies complicated by diabetes or preeclampsia, an unexpected effect of labor was noted at term. PLAC1 expression in placentae delivered vaginally following induced or spontaneous labor was significantly reduced compared to placentae not exposed to labor making it one of only a few placental genes influenced by labor. The significance of this finding is unknown. Viewed in the context of its importance in placental development, however, these findings are consistent with a role for PLAC1 in the maintenance of the maternal-fetal interface.
Collapse
Affiliation(s)
- Yahdira M Rodriguez-Prado
- Department of Pediatrics, University of South Florida Morsani College of Medicine, Tampa, FL 33606, USA
| | | | | |
Collapse
|
26
|
Matteo M, Greco P, Levi Setti P, Morenghi E, De Rosario F, Massenzio F, Albani E, Totaro P, Liso A. Preliminary evidence for high anti-PLAC1 antibody levels in infertile patients with repeated unexplained implantation failure. Placenta 2013; 34:335-9. [DOI: 10.1016/j.placenta.2013.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 01/12/2013] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
|
27
|
Bianchi DW. From prenatal genomic diagnosis to fetal personalized medicine: progress and challenges. Nat Med 2012; 18:1041-51. [PMID: 22772565 DOI: 10.1038/nm.2829] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Thus far, the focus of personalized medicine has been the prevention and treatment of conditions that affect adults. Although advances in genetic technology have been applied more frequently to prenatal diagnosis than to fetal treatment, genetic and genomic information is beginning to influence pregnancy management. Recent developments in sequencing the fetal genome combined with progress in understanding fetal physiology using gene expression arrays indicate that we could have the technical capabilities to apply an individualized medicine approach to the fetus. Here I review recent advances in prenatal genetic diagnostics, the challenges associated with these new technologies and how the information derived from them can be used to advance fetal care. Historically, the goal of prenatal diagnosis has been to provide an informed choice to prospective parents. We are now at a point where that goal can and should be expanded to incorporate genetic, genomic and transcriptomic data to develop new approaches to fetal treatment.
Collapse
Affiliation(s)
- Diana W Bianchi
- The Mother Infant Research Institute at Tufts Medical Center and the Division of Genetics, Department of Pediatrics, Floating Hospital for Children, Boston, Massachusetts, USA.
| |
Collapse
|
28
|
Edlow AG, Bianchi DW. Tracking fetal development through molecular analysis of maternal biofluids. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1970-80. [PMID: 22542507 DOI: 10.1016/j.bbadis.2012.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 02/23/2012] [Accepted: 04/10/2012] [Indexed: 10/28/2022]
Abstract
Current monitoring of fetal development includes fetal ultrasonography, chorionic villus sampling or amniocentesis for chromosome analysis, and maternal serum biochemical screening for analytes associated with aneuploidy and open neural tube defects. Over the last 15 years, significant advances in noninvasive prenatal diagnosis (NIPD) via cell-free fetal (cff) nucleic acids in maternal plasma have resulted in the ability to determine fetal sex, RhD genotype, and aneuploidy. Cff nucleic acids in the maternal circulation originate primarily from the placenta. This contrasts with cff nucleic acids in amniotic fluid, which derive from the fetus, and are present in significantly higher concentrations than in maternal blood. The fetal origin of cff nucleic acids in the amniotic fluid permits the acquisition of real-time information about fetal development and gene expression. This review seeks to provide a comprehensive summary of the molecular analysis of cff nucleic acids in maternal biofluids to elucidate mechanisms of fetal development, physiology, and pathology. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.
Collapse
Affiliation(s)
- Andrea G Edlow
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA.
| | | |
Collapse
|
29
|
Hromadnikova I. Extracellular nucleic acids in maternal circulation as potential biomarkers for placental insufficiency. DNA Cell Biol 2012; 31:1221-32. [PMID: 22364204 DOI: 10.1089/dna.2011.1530] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the placenta is being continuously remodeled during normal placental development, extracellular nucleic acids of both fetal and placental origin, packed into either trophoblast-derived apoptotic bodies or shedding syncytiotrophoblast microparticles, may be detected in maternal circulation during the course of normal gestation. Placental-insufficiency-related pregnancy complications have been shown to be associated with excessive placental trophoblast apoptosis and shedding of placenta debris. Recent advances in the field are reviewed with a focus on the diagnostic potential of particular molecular biomarkers and their eventual implementation in the currently used predictive and diagnostic algorithms for placental-insufficiency-related pregnancy complications.
Collapse
Affiliation(s)
- Ilona Hromadnikova
- Department of Molecular Biology and Cell Pathology, Third Faculty of Medicine, Charles University in Prague, Prague 10, Czech Republic.
| |
Collapse
|
30
|
Cell-free fetal nucleic acid testing: a review of the technology and its applications. Obstet Gynecol Surv 2012; 66:431-42. [PMID: 21944155 DOI: 10.1097/ogx.0b013e31822dfbe2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
UNLABELLED Cell-free fetal nucleic acids circulating in the blood of pregnant women afford the opportunity for early, noninvasive prenatal genetic testing. The predominance of admixed maternal genetic material in circulation demands innovative means for identification and analysis of cell-free fetal DNA and RNA. Techniques using polymerase chain reaction, mass spectrometry, and sequencing have been developed for the purposes of detecting fetal-specific sequences, such as paternally inherited or de novo mutations, or determining allelic balance or chromosome dosage. Clinical applications of these methods include fetal sex determination and blood group typing, which are currently available commercially although not offered routinely in the United States. Other uses of cell-free fetal DNA and RNA being explored are the detection of single-gene disorders, chromosomal abnormalities, and inheritance of parental polymorphisms across the whole fetal genome. The concentration of cell-free fetal DNA may also provide predictive capabilities for pregnancy-associated complications. The roles that cell-free fetal nucleic acid testing assume in the existing framework of prenatal screening and invasive diagnostic testing will depend on factors such as costs, clinical validity and utility, and perceived benefit-risk ratios for different applications. As cell-free fetal DNA and RNA testing continues to be developed and translated, significant ethical, legal, and social questions will arise that will need to be addressed by those with a stake in the use of this technology. TARGET AUDIENCE Obstetricians & Gynecologists and Family Physicians Learning Objectives: After participating in this activity, physicians should be better able to evaluate techniques and tools for analyzing cell-free fetal nucleic acids, assess clinical applications of prenatal testing, using cell-free fetal nucleic acids and barriers to implementation, and distinguish between relevant clinical features of cell-free fetal nucleic acid testing and existing prenatal genetic screening and diagnostic procedures.
Collapse
|
31
|
Paiva P, Whitehead C, Saglam B, Palmer K, Tong S. Measurement of mRNA transcripts of very high placental expression in maternal blood as biomarkers of preeclampsia. J Clin Endocrinol Metab 2011; 96:E1807-15. [PMID: 21865357 DOI: 10.1210/jc.2011-1233] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT mRNA of placental origin in maternal blood shows potential as a clinical biomarker of obstetric diseases such as preeclampsia (PE). We hypothesized that mRNA transcripts very highly expressed in the placenta relative to other tissues will be differentially expressed in PE and be useful as mRNA biomarkers in maternal blood. OBJECTIVE Our objective was to identify a panel of genes highly expressed in the placenta and compare their expression in placenta and maternal whole blood from PE vs. control pregnancies. SETTING Placental tissue and maternal whole blood specimens were obtained from normotensive controls (n = 15) and pregnancies complicated by severe preterm PE (n = 21). INTERVENTION mRNA expression was evaluated by quantitative real-time RT-PCR. RESULTS We identified 20 genes exhibiting highest to fourth highest expression in the placenta relative to all other tissues. All genes were detectable in placenta. Nine of the 20 genes were detectable in maternal whole blood. Four of the nine genes detectable in blood (i.e. PLAC3, PLAC4, CRH, and ERVWE1) were significantly increased in both maternal blood and placenta from PE pregnancies. The remaining five genes detectable in maternal blood were unchanged in both blood and placenta from PE pregnancies. Thus, there was complete correlation of gene expression between maternal blood and placenta. CONCLUSIONS Circulating mRNA coding genes of high placental expression show strong correlation with transcript levels in preeclamptic placenta. Such transcripts may be promising candidates to screen as mRNA biomarkers of PE in maternal whole blood.
Collapse
Affiliation(s)
- Premila Paiva
- The Ritchie Centre, Monash Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
32
|
Hahn S, Rusterholz C, Hösli I, Lapaire O. Cell-free nucleic acids as potential markers for preeclampsia. Placenta 2011; 32 Suppl:S17-20. [PMID: 21257079 DOI: 10.1016/j.placenta.2010.06.018] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Revised: 06/28/2010] [Accepted: 06/28/2010] [Indexed: 01/10/2023]
Abstract
Preeclampsia is one of the leading causes of maternal and fetal/neonatal mortality and morbidity worldwide. Therefore, widely applicable and affordable tests are needed to make an early diagnosis before the occurrence of the clinical symptoms. Circulating cell-free nucleic acids in plasma and serum are novel biomarkers with promising clinical applications in different medical fields, including prenatal diagnosis. Quantitative changes of cell-free fetal (cff)DNA in maternal plasma as an indicator for impending preeclampsia have been reported in different studies, using real-time quantitative PCR for the male-specific SRY or DYS 14 loci. In case of early onset preeclampsia, elevated levels may be already seen in the first trimester. The increased levels of cffDNA before the onset of symptoms may be due to hypoxia/reoxygenation within the intervillous space leading to tissue oxidative stress and increased placental apoptosis and necrosis. In addition to the evidence for increased shedding of cffDNA into the maternal circulation, there is also evidence for reduced renal clearance of cffDNA in preeclampsia. As the amount of fetal DNA is currently determined by quantifying Y-chromosome specific sequences, alternative approaches such as the measurement of total cell-free DNA or the use of gender-independent fetal epigenetic markers, such as DNA methylation, offer a promising alternative. Cell-free RNA of placental origin might be another potentially useful biomarker for screening and diagnosis of preeclampsia in clinical practice. Fetal RNA is associated with subcellular placental particles that protect it from degradation. Its levels are ten-fold higher in pregnant women with preeclampsia compared to controls. In conclusion, through the use of gender-independent sequences, the universal incorporation of fetal nucleic acids into routine obstetric care and into screening or diagnostic settings using combined markers may soon become a reality. Effort has now to be put into the establishment of standardized and simplified protocols for the analysis of these biomarkers in a clinical setting.
Collapse
Affiliation(s)
- S Hahn
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital, Spitalstrasse 21, CH-4031 Basel, Switzerland
| | | | | | | |
Collapse
|
33
|
Simonazzi G, Farina A, Curti A, Pilu G, Santini D, Zucchini C, Sekizawa A, Rizzo N. Higher circulating mRNA levels of placental specific genes in a patient with placenta accreta. Prenat Diagn 2011; 31:827-9. [DOI: 10.1002/pd.2761] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 03/13/2011] [Accepted: 03/13/2011] [Indexed: 11/11/2022]
|
34
|
Kodama M, Miyoshi H, Fujito N, Samura O, Kudo Y. Plasma mRNA concentrations of placenta-specific 1 (PLAC1) and pregnancy associated plasma protein A (PAPP-A) are higher in early-onset than late-onset pre-eclampsia. J Obstet Gynaecol Res 2011; 37:313-8. [DOI: 10.1111/j.1447-0756.2010.01349.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
35
|
Cetin I, Huppertz B, Burton G, Cuckle H, Gonen R, Lapaire O, Mandia L, Nicolaides K, Redman C, Soothill P, Spencer K, Thilaganathan B, Williams D, Meiri H. Pregenesys pre-eclampsia markers consensus meeting: What do we require from markers, risk assessment and model systems to tailor preventive strategies? Placenta 2011; 32 Suppl:S4-16. [DOI: 10.1016/j.placenta.2010.11.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 11/26/2010] [Accepted: 11/29/2010] [Indexed: 10/18/2022]
|
36
|
Rusterholz C, Messerli M, Hoesli I, Hahn S. Placental Microparticles, DNA, and RNA in Preeclampsia. Hypertens Pregnancy 2010; 30:364-75. [DOI: 10.3109/10641951003599571] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Miura K, Miura S, Yamasaki K, Higashijima A, Kinoshita A, Yoshiura KI, Masuzaki H. Identification of pregnancy-associated microRNAs in maternal plasma. Clin Chem 2010; 56:1767-71. [PMID: 20729298 DOI: 10.1373/clinchem.2010.147660] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Several placental microRNAs (miRNAs) have been identified as pregnancy-associated molecules with the potential for use in estimating the condition of the placenta. Our understanding of these novel molecules is still limited, however. The aim of this study was to isolate and characterize pregnancy-associated miRNAs in maternal plasma. METHODS By microarray-based screening of 723 human miRNAs, we selected miRNAs that exhibited signal intensities >100 times higher in placental tissues than in the corresponding whole blood samples. Subsequent quantitative real-time reverse-transcription PCR revealed miRNAs produced predominantly in the placenta that showed significantly decreased concentrations in maternal plasma after delivery. These miRNAs were identified as pregnancy-associated miRNAs. RESULTS We selected 82 miRNAs produced predominantly in the placenta and identified 24 as pregnancy-associated miRNAs. The genes encoding these miRNAs included 16 that are clustered on 19q13.42 and 5 clustered on 14q32. As the pregnancy progressed into the third trimester, the plasma concentrations of cell-free chromosome 19-derived miRNAs (has-miR-515-3p, has-miR-517a, has-miR-517c, has-miR-518b, and has-miR-526b) increased significantly (P = 0.0284, 0.0069, 0.0125, 0.0284, and 0.0093, respectively, Wilcoxon signed rank test), whereas that of cell-free has-miR-323-3p on chromosome 14q32.31 showed no change (P = 0.2026). CONCLUSIONS In addition to the known pregnancy-associated miRNAs, we identified new pregnancy-associated miRNAs with our microarray-based approach. Most of the genes encoding these miRNAs were clustered on 19q13.42 or 14q32, which are critical regions for placental and embryonic development. These new pregnancy-associated miRNAs may be useful molecular markers for monitoring pregnancy-associated diseases.
Collapse
Affiliation(s)
- Kiyonori Miura
- Department of Obstetrics and Gynecology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Miura K, Miura S, Yamasaki K, Shimada T, Kinoshita A, Niikawa N, Yoshiura KI, Masuzaki H. The possibility of microarray-based analysis using cell-free placental mRNA in maternal plasma. Prenat Diagn 2010; 30:849-61. [DOI: 10.1002/pd.2570] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
39
|
Chen Y, Holzman C, Chung H, Senagore P, Talge NM, Siler-Khodr T. Levels of maternal serum corticotropin-releasing hormone (CRH) at midpregnancy in relation to maternal characteristics. Psychoneuroendocrinology 2010; 35:820-32. [PMID: 20006448 PMCID: PMC2875356 DOI: 10.1016/j.psyneuen.2009.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 11/11/2009] [Accepted: 11/16/2009] [Indexed: 12/26/2022]
Abstract
BACKGROUND Corticotropin-releasing hormone (CRH) in maternal blood originates primarily from gestational tissues and elevated levels in midpregnancy have been linked to adverse pregnancy outcomes. Investigators have hypothesized that high levels of maternal stress might lead to elevated CRH levels in pregnancy. Yet a few studies have measured maternal CRH levels among subgroups of women who experience disproportionate socioeconomic disadvantage, such as African-American and Hispanic women, and found that these groups have lower CRH levels in pregnancy. Our goal was to identify maternal characteristics related to CRH levels in midpregnancy and examine which if any of these factors help to explain race differences in CRH levels. METHODS The Pregnancy Outcomes and Community Health (POUCH) Study prospectively enrolled women at 15-27 weeks' gestation from 52 clinics in five Michigan communities (1998-2004). Data from the POUCH Study were used to examine maternal demographics, anthropometrics, health behaviors, and psychosocial factors (independent variables) in relation to midpregnancy blood CRH levels modeled as logCRHpg/ml (dependent variable). Analyses were conducted within a sub-cohort from the POUCH Study (671 non-Hispanic Whites, 545 African-Americans) and repeated in the sub-cohort subset with uncomplicated pregnancies (n=746). Blood levels of CRH and independent variables were ascertained at the time of enrollment. All regression models included week of enrollment as a covariate. In addition, final multivariate regression models alternately incorporated different psychosocial measures along with maternal demographics and weight. Psychosocial variables included measures of current depressive symptoms, perceived stress, coping style, hostility, mastery, anomie, and a chronic stressor (history of abuse as a child and adult). RESULTS In sub-cohort models, the adjusted mean log CRH level was significantly lower in African-Americans vs. non-Hispanic Whites; the difference was -0.48pg/ml (P<0.01). This difference was reduced by 21% (-0.38pg/ml, P<0.01) after inclusion of other relevant covariates. Adjusted mean log CRH levels were also lower among women with <12 years vs. >or=12 years of education (minimal difference=-0.19pg/ml, P<0.05), and among women with high levels of depressive symptoms who did not use antidepressants vs. women with lower levels of depressive symptoms and no antidepressant use (minimal difference=-0.13pg/ml, P<0.01). Log CRH levels were inversely associated with maternal weight (-0.03pg/ml per 10 pound increase, P<.05) but unrelated to smoking and all other psychosocial measures. Results were similar in the subset of women with uncomplicated pregnancies, except that lower CRH levels were also linked to higher perceived stress. CONCLUSION African-American women have lower blood CRH levels at midpregnancy and the race difference in CRH levels is reduced modestly after adjustment for other maternal characteristics. CRH levels were not elevated among women with high levels of perceived stress or more chronic stressors. The inverse association between CRH levels and maternal weight is likely due to a hemodilution effect. Relations among maternal CRH levels and maternal race, educational level, and depressive symptoms are difficult to explain and invite further investigation. Our results highlight a group of covariates that merit consideration in studies that address CRH in the context of pregnancy and/or post-partum complications.
Collapse
Affiliation(s)
- Yumin Chen
- Department of Epidemiology, Michigan State University, East Lansing USA 48824
| | - Claudia Holzman
- Department of Epidemiology, Michigan State University, East Lansing USA 48824
| | - Hwan Chung
- Department of Epidemiology, Michigan State University, East Lansing USA 48824
| | - Patricia Senagore
- Department of Pathology, Michigan State University, East Lansing USA 48824
| | - Nicole M Talge
- Department of Epidemiology, Michigan State University, East Lansing USA 48824
| | - Theresa Siler-Khodr
- The Center for Investigation of Cell Regulation & Replication, San Antonio USA 78229
| |
Collapse
|
40
|
Fant M, Farina A, Nagaraja R, Schlessinger D. PLAC1 (Placenta-specific 1): a novel, X-linked gene with roles in reproductive and cancer biology. Prenat Diagn 2010; 30:497-502. [PMID: 20509147 PMCID: PMC4627609 DOI: 10.1002/pd.2506] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Placenta-specific 1 (PLAC1) is a recently described X-linked gene with expression restricted primarily to cells derived from trophoblast lineage during embryonic development. PLAC1 localizes to a region of the X chromosome thought to be important in placental development although its role in this process has not been defined. This review summarizes our current understanding of its expression, regulation, and function. PLAC1 is expressed throughout human pregnancy by the differentiated trophoblast and localizes to membranous structures in the syncytiotrophoblast, including the microvillous plasma membrane surface. Recent studies have demonstrated that PLAC1 is also expressed by a wide variety of human cancers. Studies of the PLAC1 promoter regions indicate that its expression in both normal placenta and cancer cells is driven by specific interactions involving a combination of transcription factors. Although functional insight into PLAC1 in the normal trophoblast is lacking, preliminary studies suggest that cancer-derived PLAC1 has the potential to promote tumor growth and function. In addition, it also appears to elicit a specific immunologic response that may influence survival in some cancer patients, suggesting that it may provide a therapeutic target for the treatment of some cancers. We also discuss a potential role for PLAC1 as a biomarker predictive of specific pregnancy complications, such as preeclampsia.
Collapse
Affiliation(s)
- Michael Fant
- Department of Pediatrics, University of South Florida College of Medicine, Tampa, FL 33606-350, USA.
| | | | | | | |
Collapse
|
41
|
Shimizu H, Sekizawa A, Purwosunu Y, Nakamura M, Farina A, Rizzo N, Okai T. PP13 mRNA expression in the cellular component of maternal blood as a marker for preeclampsia. Prenat Diagn 2010; 29:1231-6. [PMID: 19813218 DOI: 10.1002/pd.2380] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To assess the PP13 expression in the cellular component of blood in both preeclamptic patients and asymptomatic pregnant women during the early second trimester. METHODS In the case-control study, peripheral blood samples were obtained from pregnant women with preeclampsia (n = 24) and controls (n = 22). PP13 RNA expression was quantified in the cellular component of the blood by reverse transcription PCR assay. Next, as a cohort study of asymptomatic pregnant women at early gestation, cellular RNA from 41 cases who developed preeclampsia at later gestation and 123 cases of control were analysed, and the possibility of prediction of preeclampsia was assessed. RESULTS In symptomatic patients, PP13 level in preeclampsia was significantly lower than that in controls (p < 0.001). In the asymptomatic pregnant women, the PP13 level in preeclampsia was significantly lower than that in the controls (p = 0.008). A receiver-operating characteristics (ROC) curve yielded a detection rate of 24 and 31% at a false positive rate of 5 and 10% respectively. CONCLUSION An alteration in the cellular PP13 RNA expression would affect the placental pathophysiological change. The cellular PP13 expression level could therefore potentially be one of the key markers to predict the clinical onset of preeclampsia.
Collapse
Affiliation(s)
- Hanako Shimizu
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Sekizawa A, Purwosunu Y, Farina A, Shimizu H, Nakamura M, Wibowo N, Rizzo N, Okai T. Prediction of pre-eclampsia by an analysis of placenta-derived cellular mRNA in the blood of pregnant women at 15-20 weeks of gestation. BJOG 2010; 117:557-64. [DOI: 10.1111/j.1471-0528.2010.02491.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
43
|
Liu LP, Huang W, Lu YC, Liao AH. ORIGINAL ARTICLE: Enhanced Maternal Anti-Fetal Immunity Contributes to the Severity of Hypertensive Disorder Complicating Pregnancy. Am J Reprod Immunol 2010; 63:379-86. [DOI: 10.1111/j.1600-0897.2009.00802.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
44
|
Nakamura M, Sekizawa A, Purwosunu Y, Okazaki S, Farina A, Wibowo N, Shimizu H, Okai T. Cellular mRNA expressions of anti-oxidant factors in the blood of preeclamptic women. Prenat Diagn 2009; 29:691-6. [PMID: 19399816 DOI: 10.1002/pd.2278] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To assess the alterations of mRNA expressions associated with oxidative stress in the cellular component of blood from pregnant women with pre-eclampsia. METHODS Peripheral blood samples were obtained from pregnant women with and without pre-eclampsia. Cellular RNA was subjected to a reverse transcription polymerase chain reaction (PCR) assay in order to examine the mRNA distribution among women with pre-eclampsia (n = 24) and control subjects (n = 24) during 35-41 weeks of gestation. The data were analyzed by non-parametric statistics. RESULTS Significant differences between the pre-eclampsia subjects and the controls were observed in the gene expressions associated with oxidative stress. Lower values in the pre-eclampsia group were found for heme oxygenase (HO)-1, HO-2, catalase and superoxide dismutase (SOD). The HO-1, HO-2 and the catalase levels significantly correlated with proteinuria, and the HO-2 level with systolic blood pressure. CONCLUSION Significantly lower concentrations of HO-1, HO-2, SOD and catalase are found in the cellular component of blood from pre-eclamptic patients. The values correlate with the severity of pre-eclampsia. These findings indicate that enhanced oxidative stress and a decrease in the number of anti-oxidant enzymes may be associated with pre-eclampsia.
Collapse
Affiliation(s)
- Masamitsu Nakamura
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Pang WWI, Tsui MHY, Sahota D, Leung TY, Lau TK, Lo YMD, Chiu RWK. A strategy for identifying circulating placental RNA markers for fetal growth assessment. Prenat Diagn 2009; 29:495-504. [PMID: 19226523 DOI: 10.1002/pd.2230] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To evaluate whether circulating placental mRNAs in maternal plasma could serve as markers for the assessment of fetal growth or intrauterine growth restriction (IUGR). METHODS From a panel of placental transcripts detectable in maternal plasma identified by microarray previously, we chose growth-related transcripts, namely CSH1, GH2, KISS1, and ADAM12, as potential growth markers. Relationships between the maternal plasma mRNA concentrations with several fetal growth indicators were studied. Maternal plasma mRNA concentrations from IUGR pregnancies with or without pre-eclampsia (PET) were compared with gestational age matched controls cross-sectionally and longitudinally. The four transcripts were quantified by one-step real-time RT-PCR. RESULTS Maternal plasma GH2 mRNA significantly correlated with birth weight and fetal biometric measurements. Maternal plasma ADAM12 mRNA concentration was significantly higher in IUGR with PET than normal pregnancies in the cross-sectional comparison. No significant difference was observed for all markers between IUGR without PET and controls in both the cross-sectional and longitudinal comparisons. CONCLUSION This study presents a potential strategy in identifying surrogate markers for the study of fetal growth. Circulating GH2 mRNA in maternal plasma appeared to be associated with fetal growth. The utility of this strategy and the currently assessed markers could be explored in further studies.
Collapse
Affiliation(s)
- Winnie W I Pang
- Centre for Research into Circulating Fetal Nucleic Acids, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Liao AH, Liu LP, Ding WP, Zhang L. Functional changes of human peripheral B-lymphocytes in pre-eclampsia. Am J Reprod Immunol 2009; 61:313-21. [PMID: 19341382 DOI: 10.1111/j.1600-0897.2009.00697.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PROBLEM The aim of our study was to investigate the functional changes of human peripheral B-lymphocytes in healthy and pre-eclamptic pregnancies. METHOD OF STUDY Twenty patients with pre-eclampsia and 15 healthy third-trimester pregnant women were recruited in this study. Peripheral blood mononuclear cells (PBMCs) were isolated and directly stained with fluorescein isothiocyanate (FITC)-labeled anti-CD27 monoclonal antibody (mAb) and phycoerythrin (PE)-labeled anti-CD38 mAb. The percentages of the individual B-cell subsets were estimated out of total lymphocytes by flow cytometric analysis. Additionally, the enriched PBMCs were cultured with or without the stimulation of pokeweed mitogen (PWM) for 5 days. Then morphologic observation of plasma cells was analysed by Wright-Giemsa stain, and antibody-producing cells were detected by enzyme-linked immunospot assay. RESULTS The percentage of CD27(-)CD38(-) naïve B-cells and CD27(-)CD38(+) plasma cells did not differ between study groups (P > 0.05). The percentage of CD27(+)CD38(-) memory B-cells and CD27(+)CD38(+) plasma cell precursors increased in pre-eclamptic women compared with the controls (P < 0.05). Irrespective of whether the PBMCs were stimulated with or w/o PWM in vitro, the mean percentages of generated plasma cells were significantly higher in pre-eclamptic group than in the controls (P < 0.05). There were more antibody-producing cells in pre-eclamptic women following the activation of PWM than those in the controls (P < 0.01). CONCLUSION Our findings implicate that the functional changes of human circulating B-cells might contribute to the etiology of pre-eclampsia.
Collapse
Affiliation(s)
- Ai-Hua Liao
- Family Planning Research Institute, Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | |
Collapse
|
47
|
Purwosunu Y, Sekizawa A, Yoshimura S, Farina A, Wibowo N, Nakamura M, Shimizu H, Okai T. Expression of Angiogenesis-Related Genes in the Cellular Component of the Blood of Preeclamptic Women. Reprod Sci 2009; 16:857-64. [DOI: 10.1177/1933719109336622] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Yuditiya Purwosunu
- Department of Obstetrics and Gynecology, University of Indonesia, Cipto Mangunkusumo National Hospital, Jakarta, Indonesia, Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Akihiko Sekizawa
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan,
| | - Shiho Yoshimura
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Antonio Farina
- Department of Histology and Embryology Division of Prenatal Medicine, University of Bologna, Bologna, Italy
| | - Noroyono Wibowo
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Masamitsu Nakamura
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Hanako Shimizu
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Okai
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
48
|
Purwosunu Y, Sekizawa A, Okazaki S, Farina A, Wibowo N, Nakamura M, Rizzo N, Saito H, Okai T. Prediction of preeclampsia by analysis of cell-free messenger RNA in maternal plasma. Am J Obstet Gynecol 2009; 200:386.e1-7. [PMID: 19217595 DOI: 10.1016/j.ajog.2008.11.035] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 09/16/2008] [Accepted: 11/18/2008] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The purpose of this study was to predict the occurrence of preeclampsia in a series of patients at gestational week 15-20 weeks, with the use of a panel of messenger RNA markers. STUDY DESIGN Data from 62 patients with preeclampsia who were asymptomatic at the time of blood testing and 310 control subjects were analyzed. Multivariable analysis was performed with discriminant analysis. RESULTS Univariable analysis identified vascular endothelial growth factor receptor 1 as the marker with the highest detection rate; placenta-specific 1 with the lowest. Mean estimated score for preeclampsia was 9.4 for control subjects and 72.5 for subjects who experienced preeclampsia. A receiver operating characteristic curve that was obtained with the estimated score for preeclampsia as a test variable yielded a detection rate of 84% (95% CI, 71.8-91.5) at a 5% false-positive rate with an area under the curve of 0.927 (P < .001). Again, detection rate and score for each patient for classification as preeclamptic correlated with severity. CONCLUSION A panel of messenger RNA is able to detect subjects who will experience preeclampsia.
Collapse
|
49
|
Zhang L, Wang Y, Liao AH. Quantitative abnormalities of fetal trophoblast cells in maternal circulation in preeclampsia. Prenat Diagn 2008; 28:1160-6. [DOI: 10.1002/pd.2135] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
50
|
Dong XY, Peng JR, Ye YJ, Chen HS, Zhang LJ, Pang XW, Li Y, Zhang Y, Wang S, Fant ME, Yin YH, Chen WF. Plac1 is a tumor-specific antigen capable of eliciting spontaneous antibody responses in human cancer patients. Int J Cancer 2008; 122:2038-43. [PMID: 18183594 DOI: 10.1002/ijc.23341] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunoselection and tumor evasion constitutes one of the major obstacles in cancer immunotherapy. A potential solution to this problem is the development of polyvalent vaccines, and the identification of more tumor-specific antigens is a prerequisite for the development of cancer vaccines. To identify novel tumor-specific antigens, suppression subtractive hybridization (SSH) was performed to isolate genes differentially expressed in human hepatocellular cancer (HCC) tissues. PLAC1 (PLACenta-specific 1) was one of the genes identified highly expressed in HCC tissues but not in paired noncancerous tissues. Further analyses revealed its expression in several other types of cancer tissues as well as tumor cell lines, but not in normal tissues except for placenta. Among HCC samples tested, 32% (22/69) showed PLAC1 mRNA expression while the protein was detected in 23.3% (7/30). A serological survey revealed that 3.8% (4/101) of HCC patients had anti-PLAC1 antibody response, suggesting the immunogenicity of PLAC1 in HCC patients. PLAC1 represents a new class of tumor associated antigen with restricted expression in placenta and cancer tissues, that may serve as a target for cancer vaccination.
Collapse
Affiliation(s)
- Xue-Yuan Dong
- Department of Immunology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|