1
|
Tindal K, Cousins FL, Ellery SJ, Palmer KR, Gordon A, Filby CE, Gargett CE, Vollenhoven B, Davies-Tuck ML. Investigating Menstruation and Adverse Pregnancy Outcomes: Oxymoron or New Frontier? A Narrative Review. J Clin Med 2024; 13:4430. [PMID: 39124698 PMCID: PMC11312851 DOI: 10.3390/jcm13154430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Not discounting the important foetal or placental contribution, the endometrium is a key determinant of pregnancy outcomes. Given the inherently linked processes of menstruation, pregnancy and parturition with the endometrium, further understanding of menstruation will help to elucidate the maternal contribution to pregnancy. Endometrial health can be assessed via menstrual history and menstrual fluid, a cyclically shed, easily and non-invasively accessible biological sample that represents the distinct, heterogeneous composition of the endometrial environment. Menstrual fluid has been applied to the study of endometriosis, unexplained infertility and early pregnancy loss; however, it is yet to be examined regarding adverse pregnancy outcomes. These adverse outcomes, including preeclampsia, foetal growth restriction (FGR), spontaneous preterm birth and perinatal death (stillbirth and neonatal death), lay on a spectrum of severity and are often attributed to placental dysfunction. The source of this placental dysfunction is largely unknown and may be due to underlying endometrial abnormalities or endometrial interactions during placentation. We present existing evidence for the endometrial contribution to adverse pregnancy outcomes and propose that a more comprehensive understanding of menstruation can provide insight into the endometrial environment, offering great potential value as a diagnostic tool to assess pregnancy risk. As yet, this concept has hardly been explored.
Collapse
Affiliation(s)
- Kirstin Tindal
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia (C.E.G.); (M.L.D.-T.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
- NHMRC Centre for Research Excellence (CRE) in Stillbirth, Brisbane, QLD 4101, Australia;
| | - Fiona L. Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia (C.E.G.); (M.L.D.-T.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
| | - Stacey J. Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia (C.E.G.); (M.L.D.-T.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
| | - Kirsten R. Palmer
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
- Women’s and Newborn Program, Monash Health, Clayton, VIC 3168, Australia
| | - Adrienne Gordon
- NHMRC Centre for Research Excellence (CRE) in Stillbirth, Brisbane, QLD 4101, Australia;
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Caitlin E. Filby
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia (C.E.G.); (M.L.D.-T.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
| | - Beverley Vollenhoven
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
- Women’s and Newborn Program, Monash Health, Clayton, VIC 3168, Australia
| | - Miranda L. Davies-Tuck
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia (C.E.G.); (M.L.D.-T.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia; (K.R.P.); (B.V.)
- NHMRC Centre for Research Excellence (CRE) in Stillbirth, Brisbane, QLD 4101, Australia;
| |
Collapse
|
2
|
Zakeri S, Rahimi Z, Rezvani N, Vaisi-Raygani A, Alibakhshi R, Zakeri S, Yari K. The influence of Nrf2 gene promoter methylation on gene expression and oxidative stress parameters in preeclampsia. BMC Med Genomics 2024; 17:64. [PMID: 38419047 PMCID: PMC10903067 DOI: 10.1186/s12920-023-01791-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 12/26/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND AND AIMS Preeclampsia (PE) is a serious medical condition that usually causes high blood pressure and affects multiple organs. Considering the adverse effect of oxidative stress on the process of PE in pregnant women and regarding the role of the Nrf2 gene in placental oxidative pathways, this study was conducted to investigate the DNA methylation status of Nrf2 in PE and healthy pregnant women. MATERIALS AND METHODS The present case-control study consisted of 70 PE and 70 healthy pregnant women. Blood and placenta samples were taken from all subjects, and the percentage of the Nrf2 gene methylation in the samples was assessed by the Methyl Light PCR method. Also, the Nrf2 gene expression was evaluated by real-time PCR. The total antioxidant capacity (TAC) and total oxidative status (TOS) were measured by the colorimetric method. RESULTS In PE women, there was a significant increase in blood pressure, term of pregnancy, and BMI. In addition, there were enhanced Nrf2 DNA methylation percentage in placenta tissue and increased TOS levels in placenta tissue and blood compared to healthy pregnant women (P < 0.05). Also, in the PE group, there was a significant decrease in Nrf2 gene expression and TAC level in placenta tissue compared to the control group (P < 0.05). CONCLUSION The Nrf2 gene undergoes epigenetic modifications of DNA hypermethylation in the PE placenta. Decreased expression of this gene and the changes in the level of oxidative parameters (TAC, TOS) confirm it.
Collapse
Affiliation(s)
- Saba Zakeri
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, P.O.Box: 67148-69914, Iran
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, P.O.Box: 67148-69914, Iran.
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Nayebali Rezvani
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, P.O.Box: 67148-69914, Iran
| | - Asad Vaisi-Raygani
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, P.O.Box: 67148-69914, Iran
| | - Reza Alibakhshi
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, P.O.Box: 67148-69914, Iran
| | - Sahel Zakeri
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Sanandaj, Iran
| | - Kheirolah Yari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
3
|
Varghese B, Babu S, Jala A, Das P, Raju R, Borkar RM, Adela R. Integrative Placental Multi-Omics Analysis Reveals Perturbed Pathways and Potential Prognostic Biomarkers in Gestational Hypertension. Arch Med Res 2024; 55:102909. [PMID: 37984232 DOI: 10.1016/j.arcmed.2023.102909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/26/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Gestational hypertension (GH) is a severe complication that occurs after 20 weeks of pregnancy; however, its molecular mechanisms are not yet fully understood. OBJECTIVE Through this case-control discovery phase study, we aimed to find disease-specific candidate placental microRNAs (miRNAs) and metabolite markers for differentiating GH by integrating next-generation sequencing and metabolomics multi-omics analysis of placenta. Using small RNA sequencing and metabolomics of placental tissues of healthy pregnant (HP, n = 24) and GH subjects (n = 20), the transcriptome and metabolome were characterized in both groups. RESULTS The study identified a total of 44 downregulated placental miRNAs which includes three novel, three mature and 38 precursor miRNAs. Six miRNAs including three mature (hsa-miR-181a-5p, hsa-miR-498-5p, and hsa-miR-26b-5p) and three novel (NC_000016.10_1061, NC_000005.10_475, and NC_000001.11_53) were considered for final target prediction and functional annotation. Integrative analysis of differentially expressed miRNAs and metabolites yielded five pathways such as purine, glutathione, glycerophospholipid, inositol phosphate and β-alanine to be significantly perturbed in GH. We present fourteen genes (LPCAT1, LPCAT2, DGKH, PISD, GPAT2, PTEN, SACM1L, PGM2, AMPD3, AK7, AK3, CNDP1, IDH2, and ODC1) and eight metabolites (xanthosine, xanthine, spermine, glycine, CDP-Choline, glyceraldehyde 3-phosphate, β-alanine, and histidine) with potential to distinguish GH and HP. CONCLUSION The differential expression of miRNAs, their target genes, altered metabolites and metabolic pathways in GH patients were identified for the first time in our study. Further, the altered miRNAs and metabolites were integrated to build their inter-connectivity network. The findings obtained from our study may be used as a valuable source to further unravel the molecular pathways associated with GH and also for the evaluation of prognostic markers.
Collapse
Affiliation(s)
- Bincy Varghese
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Sreeranjini Babu
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India; Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Aishwarya Jala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Panchanan Das
- Department of Obstetrics and Gynecology, Gauhati Medical College, Guwahati, Assam, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore, India
| | - Roshan M Borkar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India.
| |
Collapse
|
4
|
Kaur L, Sundrani D, Dave K, Randhir K, Mehendale S, Bayyana S, Kalyanaraman K, Chandak GR, Joshi S. Hypoxia Inducible Factors (HIF1α and HIF3α) are differentially methylated in preeclampsia placentae and are associated with birth outcomes. Mol Cell Biochem 2023; 478:2309-2318. [PMID: 36708442 DOI: 10.1007/s11010-023-04661-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/09/2023] [Indexed: 01/29/2023]
Abstract
Preeclampsia is a placental vascular pathology and hypoxia is known to influence placental angiogenesis. Hypoxia Inducible Factors (HIF1α and HIF3α) mediate the response to cellular oxygen concentration and bind to hypoxia response element of target genes. However the mechanism regulating above activity is not well-understood. We investigated if placental DNA methylation (DNAm) and expression of HIF1α and 3α genes are altered and associated with pre-eclampsia, placental weight and birth outcomes. Using a cohort comprising women with preeclampsia [N = 100, delivering at term (N = 43) and preterm (N = 57)] and normotensive controls (N = 100), we analysed DNAm in HIF1α and 3α, and their mRNA expression in placentae, employing pyrosequencing and quantitative real-time PCR, respectively. We observed significant hypermethylation at cg22891070 of HIF3α in preeclampsia placentae compared to controls (β = 1.5%, p = 0.04). CpG8 in the promoter region of HIF1α, showed marginally significant hypomethylation in preterm preeclampsia compared to controls (β = - 0.15%, p = 0.055). HIF1α expression was significantly lower in preterm preeclampsia compared to controls (mean ± SE = 10.16 ± 2.00 vs 4.25 ± 0.90, p = 0.04). Further, DNAm in HIF1α promoter region was negatively associated with its expression levels (β = - 0.165, p = 0.024). Several CpGs in HIF1α were negatively associated with placental weight and birth outcomes including birth weight (β range = - 0.224-0.300) and birth length [β range = - 0.248 to - 0.301 (p < 0.05 for all)]. Overall, we demonstrate altered DNAm in HIF1α and HIF3α in preeclampsia placentae, also associated with various birth outcomes. Correlation of DNAm in HIF1α and its expression suggests a possible role in the pathogenesis of pre-eclampsia. Further investigations on interactions between HIF1α and HIF3α in preeclampsia would be interesting.
Collapse
Affiliation(s)
- Lovejeet Kaur
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, 500007, India
- Maternal and Child Health (MCH), Translational Health Science and Technology Institute (THSTI), Faridabad, 121001, India
| | - Deepali Sundrani
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Kinjal Dave
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Karuna Randhir
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India
| | - Savita Mehendale
- Department of Gynecology and Obstetrics, Bharati Vidyapeeth Medical College and Hospital, Pune, 411043, India
| | - Swati Bayyana
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, 500007, India
| | - Kumaran Kalyanaraman
- CSI Epidemiology Research Unit, Holdsworth Memorial Hospital, Mysore, India
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Giriraj R Chandak
- Genomic Research on Complex Diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, 500007, India.
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be) University, Pune Satara Road, Pune, 411043, India.
| |
Collapse
|
5
|
Tossetta G, Fantone S, Piani F, Crescimanno C, Ciavattini A, Giannubilo SR, Marzioni D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023; 12:1545. [PMID: 37296665 PMCID: PMC10252212 DOI: 10.3390/cells12111545] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Placentation is a key and tightly regulated process that ensures the normal development of the placenta and fetal growth. Preeclampsia (PE) is a hypertensive pregnancy-related disorder involving about 5-8% of all pregnancies and clinically characterized by de novo maternal hypertension and proteinuria. In addition, PE pregnancies are also characterized by increased oxidative stress and inflammation. The NRF2/KEAP1 signaling pathway plays an important role in protecting cells against oxidative damage due to increased reactive oxygen species (ROS) levels. ROS activate NRF2, allowing its binding to the antioxidant response element (ARE) region present in the promoter of several antioxidant genes such as heme oxygenase, catalase, glutathione peroxidase and superoxide dismutase that neutralize ROS, protecting cells against oxidative stress damages. In this review, we analyze the current literature regarding the role of the NRF2/KEAP1 pathway in preeclamptic pregnancies, discussing the main cellular modulators of this pathway. Moreover, we also discuss the main natural and synthetic compounds that can regulate this pathway in in vivo and in vitro models.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| | - Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40128 Bologna, Italy;
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Caterina Crescimanno
- School of Human and Social Science, University “Kore” of Enna, 94100 Enna, Italy;
| | - Andrea Ciavattini
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, 60123 Ancona, Italy; (A.C.); (S.R.G.)
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, 60123 Ancona, Italy; (A.C.); (S.R.G.)
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy; (S.F.); (D.M.)
| |
Collapse
|
6
|
Liu S, Fu H, Ray M, Heinsberg LW, Conley YP, Anderson CM, Hubel CA, Roberts JM, Jeyabalan A, Weeks DE, Schmella MJ. A longitudinal epigenome-wide association study of preeclamptic and normotensive pregnancy. EPIGENETICS COMMUNICATIONS 2023; 3:1. [PMID: 37063698 PMCID: PMC10101051 DOI: 10.1186/s43682-022-00014-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/31/2022] [Indexed: 01/28/2023]
Abstract
Background While preeclampsia (PE) is a leading cause of pregnancy-related morbidity/mortality, its underlying mechanisms are not fully understood. DNA methylation (DNAm) is a dynamic regulator of gene expression that may offer insight into PE pathophysiology and/or serve as a biomarker (e.g., risk, subtype, a therapeutic response). This study's purpose was to evaluate for differences in blood-based DNAm across all trimesters between individuals eventually diagnosed with PE (cases) and individuals who remained normotensive throughout pregnancy, did not develop proteinuria, and birthed a normally grown infant (controls). Results In the discovery phase, longitudinal, genome-wide DNAm data were generated across three trimesters of pregnancy in 56 participants (n=28 cases, n=28 controls) individually matched on self-identified race, pre-pregnancy body mass index, smoking, and gestational age at sample collection. An epigenome-wide association study (EWAS) was conducted, using surrogate variable analysis to account for unwanted sources of variation. No CpGs met the genome-wide significance p value threshold of 9×10-8, but 16 CpGs (trimester 1: 5; trimester 2: 1; trimester 3: 10) met the suggestive significance threshold of 1×10-5. DNAm data were also evaluated for differentially methylated regions (DMRs) by PE status. Three DMRs in each trimester were significant after Bonferonni-adjustment. Since only third-trimester samples were available from an independent replication sample (n=64 cases, n=50 controls), the top suggestive hits from trimester 3 (cg16155413 and cg21882990 associated with TRAF3IP2-AS1/TRAF3IP2 genes, which also made up the top DMR) were carried forward for replication. During replication, DNAm data were also generated for validation purposes from discovery phase third trimester samples. While significant associations between DNAm and PE status were observed at both sites in the validation sample, no associations between DNAm and PE status were observed in the independent replication sample. Conclusions The discovery phase findings for cg16155413/cg21882990 (TRAF3IP2-AS1/TRAF3IP2) were validated with a new platform but were not replicated in an independent sample. Given the differences in participant characteristics between the discovery and replication samples, we cannot rule out important signals for these CpGs. Additional research is warranted for cg16155413/cg21882990, as well as top hits in trimesters 1-2 and significant DMRs that were not examined in the replication phase.
Collapse
Affiliation(s)
- Shuwei Liu
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haoyi Fu
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mitali Ray
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, 440 Victoria Building, 3500 Victoria Street, Pittsburgh, PA 15261, USA
| | - Lacey W. Heinsberg
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yvette P. Conley
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, 440 Victoria Building, 3500 Victoria Street, Pittsburgh, PA 15261, USA
| | - Cindy M. Anderson
- Martha S. Pitzer Center for Women, Children and Youth, College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Carl A. Hubel
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - James M. Roberts
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Arun Jeyabalan
- Magee-Womens Research Institute and Foundation, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel E. Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mandy J. Schmella
- Department of Health Promotion and Development, School of Nursing, University of Pittsburgh, 440 Victoria Building, 3500 Victoria Street, Pittsburgh, PA 15261, USA
| |
Collapse
|
7
|
Marić I, Contrepois K, Moufarrej MN, Stelzer IA, Feyaerts D, Han X, Tang A, Stanley N, Wong RJ, Traber GM, Ellenberger M, Chang AL, Fallahzadeh R, Nassar H, Becker M, Xenochristou M, Espinosa C, De Francesco D, Ghaemi MS, Costello EK, Culos A, Ling XB, Sylvester KG, Darmstadt GL, Winn VD, Shaw GM, Relman DA, Quake SR, Angst MS, Snyder MP, Stevenson DK, Gaudilliere B, Aghaeepour N. Early prediction and longitudinal modeling of preeclampsia from multiomics. PATTERNS (NEW YORK, N.Y.) 2022; 3:100655. [PMID: 36569558 PMCID: PMC9768681 DOI: 10.1016/j.patter.2022.100655] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 09/28/2022] [Accepted: 11/11/2022] [Indexed: 12/13/2022]
Abstract
Preeclampsia is a complex disease of pregnancy whose physiopathology remains unclear. We developed machine-learning models for early prediction of preeclampsia (first 16 weeks of pregnancy) and over gestation by analyzing six omics datasets from a longitudinal cohort of pregnant women. For early pregnancy, a prediction model using nine urine metabolites had the highest accuracy and was validated on an independent cohort (area under the receiver-operating characteristic curve [AUC] = 0.88, 95% confidence interval [CI] [0.76, 0.99] cross-validated; AUC = 0.83, 95% CI [0.62,1] validated). Univariate analysis demonstrated statistical significance of identified metabolites. An integrated multiomics model further improved accuracy (AUC = 0.94). Several biological pathways were identified including tryptophan, caffeine, and arachidonic acid metabolisms. Integration with immune cytometry data suggested novel associations between immune and proteomic dynamics. While further validation in a larger population is necessary, these encouraging results can serve as a basis for a simple, early diagnostic test for preeclampsia.
Collapse
Affiliation(s)
- Ivana Marić
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mira N. Moufarrej
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiaoyuan Han
- University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA 94103, USA
| | - Andy Tang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ronald J. Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gavin M. Traber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Huda Nassar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Camilo Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad S. Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Canada
| | - Elizabeth K. Costello
- Departments of Medicine, and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuefeng B. Ling
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gary L. Darmstadt
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gary M. Shaw
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A. Relman
- Departments of Medicine, and of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Infectious Diseases Section, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Stephen R. Quake
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David K. Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brice Gaudilliere
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nima Aghaeepour
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
8
|
Hu B, Li D, Tang D, Shangguan Y, Cao Y, Guo R, Luan S, Yun C, Morgera S, Hocher B, Krämer BK, Wang Y, Yin L, Dai Y. Integrated proteome and acetylome analyses unveil protein features of gestational diabetes mellitus and preeclampsia. Proteomics 2022; 22:e2200124. [PMID: 36097143 DOI: 10.1002/pmic.202200124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 12/29/2022]
Abstract
Gestational diabetes mellitus (GDM) and preeclampsia (PE) are associated with maternal and infant health. Although the pathogenesis of PE and GDM remains controversial, oxidative stress is involved in the underlying pathology of GDM and PE. Protein lysine acetylation (Kac) plays an important regulatory role in biological processes. There is little data regarding the association of the maternal acetylome with GDM and PE. This study aimed to assess the potential value of the proteome and acetylome for GDM and PE. In our study, we included placental tissues from healthy individuals (n = 6), GDM patients (n = 6), and PE patients (n = 6) to perform 4D-label free quantification proteomics analysis and PRM analysis. We identified 22 significantly regulated proteins and 192 significantly regulated acetylated proteins between the GDM and PE groups. Furthermore, 192 significantly regulated acetylated proteins were mainly enriched in endoplasmic reticulum stress (ERS) and ferroptosis pathways. Seventeen acetylated sites in these two pathways were verified by PRM analysis. Our comprehensive analysis revealed key features of GDM/PE-significantly regulated acetylated proteins in the placentas from GDM and PE. The results of signaling pathway analysis focused on ERS and ferroptosis. These findings may help explore the underlying pathology, new biomarkers, and therapeutic targets of GDM and PE.
Collapse
Affiliation(s)
- Biying Hu
- Clinical Medical Research Center, Guangdong Clinical Medical Research Center of Birth Defects, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China.,Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Dandan Li
- Clinical Medical Research Center, Guangdong Clinical Medical Research Center of Birth Defects, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Clinical Medical Research Center of Birth Defects, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Yu Shangguan
- Clinical Medical Research Center, Guangdong Clinical Medical Research Center of Birth Defects, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China
| | - Yuzhi Cao
- Department of Obstetrics and Gynecology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Ruonan Guo
- Department of Obstetrics and Gynecology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Shaodong Luan
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Guangdong, Guangdong, China
| | - Chen Yun
- Charité -Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.,Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | | | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Yinglan Wang
- Department of Obstetrics and Gynecology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Clinical Medical Research Center of Birth Defects, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Mundal SB, Rakner JJ, Silva GB, Gierman LM, Austdal M, Basnet P, Elschot M, Bakke SS, Ostrop J, Thomsen LCV, Moses EK, Acharya G, Bjørge L, Iversen AC. Divergent Regulation of Decidual Oxidative-Stress Response by NRF2 and KEAP1 in Preeclampsia with and without Fetal Growth Restriction. Int J Mol Sci 2022; 23:ijms23041966. [PMID: 35216082 PMCID: PMC8875334 DOI: 10.3390/ijms23041966] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/01/2023] Open
Abstract
Utero-placental development in pregnancy depends on direct maternal–fetal interaction in the uterine wall decidua. Abnormal uterine vascular remodeling preceding placental oxidative stress and placental dysfunction are associated with preeclampsia and fetal growth restriction (FGR). Oxidative stress is counteracted by antioxidants and oxidative repair mechanisms regulated by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2). We aimed to determine the decidual regulation of the oxidative-stress response by NRF2 and its negative regulator Kelch-like ECH-associated protein 1 (KEAP1) in normal pregnancies and preeclamptic pregnancies with and without FGR. Decidual tissue from 145 pregnancies at delivery was assessed for oxidative stress, non-enzymatic antioxidant capacity, cellular NRF2- and KEAP1-protein expression, and NRF2-regulated transcriptional activation. Preeclampsia combined with FGR was associated with an increased oxidative-stress level and NRF2-regulated gene expression in the decidua, while decidual NRF2- and KEAP1-protein expression was unaffected. Although preeclampsia with normal fetal growth also showed increased decidual oxidative stress, NRF2-regulated gene expression was reduced, and KEAP1-protein expression was increased in areas of high trophoblast density. The trophoblast-dependent KEAP1-protein expression in preeclampsia with normal fetal growth indicates control of decidual oxidative stress by maternal–fetal interaction and underscores the importance of discriminating between preeclampsia with and without FGR.
Collapse
Affiliation(s)
- Siv Boon Mundal
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
- Women’s Health and Perinatology Research Group, Department of Clinical Medicine, UiT—The Arctic University of Norway, 9037 Tromsø, Norway; (P.B.); (G.A.)
| | - Johanne Johnsen Rakner
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
| | - Gabriela Brettas Silva
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
- Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Lobke Marijn Gierman
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
- Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Marie Austdal
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
- Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Department of Research, Stavanger University Hospital, 4068 Stavanger, Norway
| | - Purusotam Basnet
- Women’s Health and Perinatology Research Group, Department of Clinical Medicine, UiT—The Arctic University of Norway, 9037 Tromsø, Norway; (P.B.); (G.A.)
- Department of Obstetrics and Gynecology, University Hospital of Northern Norway, 9037 Tromsø, Norway
| | - Mattijs Elschot
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway;
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Siril Skaret Bakke
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
| | - Jenny Ostrop
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
| | - Liv Cecilie Vestrheim Thomsen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, 5058 Bergen, Norway; (L.C.V.T.); (L.B.)
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Eric Keith Moses
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia;
| | - Ganesh Acharya
- Women’s Health and Perinatology Research Group, Department of Clinical Medicine, UiT—The Arctic University of Norway, 9037 Tromsø, Norway; (P.B.); (G.A.)
- Department of Clinical Science, Division of Obstetrics and Gynecology, Intervention and Technology, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Line Bjørge
- Department of Gynecology and Obstetrics, Haukeland University Hospital, 5058 Bergen, Norway; (L.C.V.T.); (L.B.)
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway; (S.B.M.); (J.J.R.); (G.B.S.); (L.M.G.); (M.A.); (S.S.B.); (J.O.)
- Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
- Correspondence: ; Tel.: +47-93283877
| |
Collapse
|
10
|
Garrido-Gómez T, Castillo-Marco N, Cordero T, Simón C. Decidualization resistance in the origin of preeclampsia. Am J Obstet Gynecol 2022; 226:S886-S894. [PMID: 33007270 DOI: 10.1016/j.ajog.2020.09.039] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022]
Abstract
Preeclampsia is a major obstetrical complication with short- and long-term life-threatening consequences for both mother and child. Shallow cytotrophoblast invasion through the uterine decidua into the spiral arteries is implicated in the pathogenesis of preeclampsia, although the cause of deficient arterial invasion remains unknown. Research that is focused on the "soil"-the maternal decidua-highlights the importance of this poorly understood but influential uterine layer. Decidualization of endometrial cells regulates embryo invasion, which is essential for spiral artery remodeling and establishing the maternal-fetal interface. Exploration of the association between impaired decidualization and preeclampsia revealed suboptimal endometrial maturation and uterine natural killer cells present in the decidua before preeclampsia development. Furthermore, decidualization defects in the endometrium of women with severe preeclampsia, characterized by impaired cytotrophoblast invasion, were detected at the time of delivery and persisted 5 years after the affected pregnancy. Recently, a maternal deficiency of annexin A2 expression was found to influence aberrant decidualization and shallow cytotrophoblast invasion, suggesting that decidualization resistance, which is a defective endometrial cell differentiation during the menstrual cycle, could underlie shallow trophoblast invasion and the poor establishment of the maternal-fetal interface. Based on these findings, the transcriptional signature in the endometrium that promotes decidualization deficiency could be detected before (or after) conception. This would serve to identify women at risk of developing severe preeclampsia and aid the development of therapies focused on improving decidualization, perhaps also preventing severe preeclampsia. Here, we discuss decidualization deficiency as a contributor to the pathogenesis of pregnancy disorders with particular attention to severe preeclampsia. We also review current diagnostic strategies and discuss future directions in diagnostic methods based on decidualization.
Collapse
|
11
|
Li N, Hou R, Liu C, Yang T, Qiao C, Wei J. Integration of transcriptome and proteome profiles in placenta accreta reveals trophoblast over-migration as the underlying pathogenesis. Clin Proteomics 2021; 18:31. [PMID: 34963445 PMCID: PMC8903580 DOI: 10.1186/s12014-021-09336-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 12/09/2021] [Indexed: 12/03/2022] Open
Abstract
Background Placenta accreta (PA) is a major cause of maternal morbidity and mortality in modern obstetrics, few studies have explored the underlying molecular mechanisms. Methods In our study, transcriptome and proteome profiling were performed in placental tissues from ten participants including five cases each in the PA and control groups to clarify the pathogenesis of PA. Results We identified differential expression of 37,743 transcripts and 160 proteins between the PA and control groups with an overlap rate of 0.09%. The 33 most-significant transcripts and proteins were found and further screened and analyzed. Adhesion-related signature, chemotaxis related signatures and immune related signature were found in the PA group and played a certain role. Sum up two points, three significant indicators, methyl-CpG-binding domain protein 2 (MeCP2), podocin (PODN), and apolipoprotein D (ApoD), which participate in “negative regulation of cell migration”, were downregulated at the mRNA and protein levels in PA group. Furthermore, transwell migration and invasion assay of HTR-8/SVneo cell indicated the all of them impaired the migration and invasion of trophoblast. Conclusion A poor correlation was observed between the transcriptome and proteome data and MeCP2, PODN, and ApoD decreased in transcriptome and proteome profiling, resulting in increased migration of trophoblasts in the PA group, which clarify the mechanism of PA and might be the biomarkers or therapy targets in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09336-8.
Collapse
Affiliation(s)
- Na Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Rui Hou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Caixia Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China
| | - Tian Yang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chong Qiao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China. .,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China.
| | - Jun Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China. .,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province; Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi, China.
| |
Collapse
|
12
|
Amin-Beidokhti M, Sadeghi H, Pirjani R, Gachkar L, Gholami M, Mirfakhraie R. Differential expression of Hsa-miR-517a/b in placental tissue may contribute to the pathogenesis of preeclampsia. J Turk Ger Gynecol Assoc 2021; 22:273-278. [PMID: 34866368 PMCID: PMC8666996 DOI: 10.4274/jtgga.galenos.2021.2021.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: Preeclampsia (PE) is a pregnancy hypertensive disorder that affects both maternal and fetal health. Many studies have investigated possible mechanisms in the pathogenesis of PE although the role of the placenta is undeniable. Evaluation of placental-specific microRNAs may provide additional data about the pathogenic mechanism of PE. This study compared the expression levels of Hsa-miR-517a/b in placental tissues obtained from PE patients and healthy controls. Material and Methods: One hundred tissues were obtained from fetal and maternal sides of the placenta of PE patients and healthy controls. Expression analysis was performed using quantitative real-time polymerase chain reaction. Results: Hsa-miR-517a/b level was significantly decreased in PE compared to controls (expression ratio: 0.40; p=0.007). Down-regulation of Hsa-miR-517a/b was also detected in fetal-side placental samples when compared to maternal-side in PE (expression ratio: 0.33; p=0.04). Furthermore, decreased expression of Hsa-miR-517a/b was detected in fetal-side tissue from PE cases compared to fetal-side samples from healthy pregnancies (expression ratio: 0.36; p=0.03). In maternal-side placental samples the expression level did not differ between PE and healthy pregnancies (p=0.1). Conclusion: These results demonstrate a differential expression of Hsa-miR-517a/b within placentas in pregnancies affected by PE and between placentas from PE and healthy pregnancies. Further studies are required to investigate a possible role for Hsa-miR-517a/b in the pathogenesis of PE.
Collapse
Affiliation(s)
- Mona Amin-Beidokhti
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Sadeghi
- Genomic Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reihaneh Pirjani
- Obstetrics and Gynecology Department, Arash Women Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Latif Gachkar
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Gholami
- Department of Biochemistry and Genetics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Reza Mirfakhraie
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Genomic Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Garrido-Gomez T, Castillo-Marco N, Clemente-Ciscar M, Cordero T, Muñoz-Blat I, Amadoz A, Jimenez-Almazan J, Monfort-Ortiz R, Climent R, Perales-Marin A, Simon C. Disrupted PGR-B and ESR1 signaling underlies defective decidualization linked to severe preeclampsia. eLife 2021; 10:70753. [PMID: 34709177 PMCID: PMC8553341 DOI: 10.7554/elife.70753] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Decidualization of the uterine mucosa drives the maternal adaptation to invasion by the placenta. Appropriate depth of placental invasion is needed to support a healthy pregnancy; shallow invasion is associated with the development of severe preeclampsia (sPE). Maternal contribution to sPE through failed decidualization is an important determinant of placental phenotype. However, the molecular mechanism underlying the in vivo defect linking decidualization to sPE is unknown. Methods: Global RNA sequencing was applied to obtain the transcriptomic profile of endometrial biopsies collected from nonpregnant women who suffer sPE in a previous pregnancy and women who did not develop this condition. Samples were randomized in two cohorts, the training and the test set, to identify the fingerprinting encoding defective decidualization in sPE and its subsequent validation. Gene Ontology enrichment and an interaction network were performed to deepen in pathways impaired by genetic dysregulation in sPE. Finally, the main modulators of decidualization, estrogen receptor 1 (ESR1) and progesterone receptor B (PGR-B), were assessed at the level of gene expression and protein abundance. Results: Here, we discover the footprint encoding this decidualization defect comprising 120 genes—using global gene expression profiling in decidua from women who developed sPE in a previous pregnancy. This signature allowed us to effectively segregate samples into sPE and control groups. ESR1 and PGR were highly interconnected with the dynamic network of the defective decidualization fingerprint. ESR1 and PGR-B gene expression and protein abundance were remarkably disrupted in sPE. Conclusions: Thus, the transcriptomic signature of impaired decidualization implicates dysregulated hormonal signaling in the decidual endometria in women who developed sPE. These findings reveal a potential footprint that could be leveraged for a preconception or early prenatal screening of sPE risk, thus improving prevention and early treatments. Funding: This work has been supported by the grant PI19/01659 (MCIU/AEI/FEDER, UE) from the Spanish Carlos III Institute awarded to TGG. NCM was supported by the PhD program FDGENT/2019/008 from the Spanish Generalitat Valenciana. IMB was supported by the PhD program PRE2019-090770 and funding was provided by the grant RTI2018-094946-B-100 (MCIU/AEI/FEDER, UE) from the Spanish Ministry of Science and Innovation with CS as principal investigator. This research was funded partially by Igenomix S.L.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rogelio Monfort-Ortiz
- Department of Obstetrics and Gynecology, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Reyes Climent
- Department of Obstetrics and Gynecology, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Alfredo Perales-Marin
- Department of Obstetrics and Gynecology, University and Polytechnic La Fe Hospital, Valencia, Spain.,Department of Obstetrics and Gynecology, School of Medicine, Valencia University, Valencia, Spain
| | - Carlos Simon
- Igenomix Foundation, INCLIVA, Valencia, Spain.,Department of Obstetrics and Gynecology, School of Medicine, Valencia University, Valencia, Spain.,Obstetrics & Gynecology, BIDMC Harvard University, Boston, United States
| |
Collapse
|
14
|
Almada M, Costa L, Fonseca B, Alves P, Braga J, Gonçalves D, Teixeira N, Correia-da-Silva G. The endocannabinoid 2-arachidonoylglycerol promotes endoplasmic reticulum stress in placental cells. Reproduction 2021; 160:171-180. [PMID: 32357311 PMCID: PMC7354702 DOI: 10.1530/rep-19-0539] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/01/2020] [Indexed: 12/19/2022]
Abstract
Proliferation, differentiation and apoptosis of trophoblast cells are required for normal placental development. Impairment of those processes may lead to pregnancy-related diseases. Disruption of endoplasmic reticulum (ER) homeostasis has been associated with several reproductive pathologies including recurrent pregnancy loss and preeclampsia. In the unfolded protein response (UPR), specific ER-stress signalling pathways are activated to restore ER homeostasis, but if the adaptive response fails, apoptosis is triggered. Protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1) and Activating transcription factor 6 (ATF6) are central players in UPR and in ER-stress-induced apoptosis, as well as downstream transcription factors, as C/EBP homologous protein (CHOP). Our previous studies have shown that the endocannabinoid 2-arachidonoylglycerol (2-AG) modulates trophoblast cell turnover. Nevertheless, the role of ER-stress on 2-AG induced apoptosis and cannabinoid signalling in trophoblast has never been addressed. In this work, we used BeWo cells and human primary cytotrophoblasts isolated from term-placenta. The expression of ER-stress markers was analysed by qRT-PCR and Western blotting. ROS generation was assessed by fluorometric methods, while apoptosis was detected by the evaluation of caspase -3/-7 activities and Poly (ADP-ribose) polymerase (PARP) cleavage. Our findings indicate that 2-AG is able to induce ER-stress and apoptosis. Moreover, the eukaryotic initiation factor 2 (eIF2α)/CHOP pathway involved in ER-stress-induced apoptosis is triggered through a mechanism dependent on cannabinoid receptor CB2 activation. The results bring novel insights on the importance of ER-stress and cannabinoid signalling on 2-AG mechanisms of action in placenta.
Collapse
Affiliation(s)
- Marta Almada
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Lia Costa
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.,Departamento de Biologia, Universidade de Aveiro, Aveiro, Portugal
| | - Bruno Fonseca
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Patrícia Alves
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Jorge Braga
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte Dr Albino Aroso, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Daniela Gonçalves
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte Dr Albino Aroso, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Natércia Teixeira
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO, REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
15
|
Chen S, Yin Q, Hu H, Chen Q, Huang Q, Zhong M. AOPPs induce HTR-8/SVneo cell apoptosis by downregulating the Nrf-2/ARE/HO-1 anti-oxidative pathway: Potential implications for preeclampsia. Placenta 2021; 112:1-8. [PMID: 34237527 DOI: 10.1016/j.placenta.2021.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/29/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Advanced oxidation protein products (AOPPs), which are novel markers of oxidant-mediated protein damage, are prevalent in numerous diseases. We previously demonstrated that AOPPs act as a new class of pathogenic mediators in preeclampsia by causing trophoblast damage and dysfunction. Herein, we explored whether AOPPs could regulate the Nrf-2/ARE/HO-1 anti-oxidative pathway to facilitate the progression of preeclampsia. METHODS To investigate the pathophysiology of preeclampsia, we evaluated the effects of AOPPs on trophoblast damage, apoptotic proteins, and Nrf-2/ARE/HO-1 anti-oxidative pathway expression, as well as their underlying mechanisms. RESULTS AOPPs directly increased the expression of apoptotic proteins and significantly inhibited the expression of Nrf-2/ARE/HO-1 pathway in trophoblasts. Nrf-2 silencing aggravated the AOPPs-induced cell apoptosis in vitro by activating p53 and caspase cascade, whereas Nrf-2 overexpression had the opposite effect. Moreover, Nrf-2 exerted cytoprotective effects by increasing HO-1. DISCUSSION These findings suggest that AOPPs induce trophoblast apoptosis by triggering p53 and caspase activation via inhibition of the Nrf-2/ARE/HO-1 anti-oxidative pathway. Hence, Nrf-2/ARE/HO-1 pathway activation plays a protective role in AOPPs-induced cell apoptosis; thus, holding potential as a therapeutic target against preeclampsia.
Collapse
Affiliation(s)
- Shuying Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Yin
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoyue Hu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qitao Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
Yong HEJ, Chan SY. Current approaches and developments in transcript profiling of the human placenta. Hum Reprod Update 2021; 26:799-840. [PMID: 33043357 PMCID: PMC7600289 DOI: 10.1093/humupd/dmaa028] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The placenta is the active interface between mother and foetus, bearing the molecular marks of rapid development and exposures in utero. The placenta is routinely discarded at delivery, providing a valuable resource to explore maternal-offspring health and disease in pregnancy. Genome-wide profiling of the human placental transcriptome provides an unbiased approach to study normal maternal–placental–foetal physiology and pathologies. OBJECTIVE AND RATIONALE To date, many studies have examined the human placental transcriptome, but often within a narrow focus. This review aims to provide a comprehensive overview of human placental transcriptome studies, encompassing those from the cellular to tissue levels and contextualize current findings from a broader perspective. We have consolidated studies into overarching themes, summarized key research findings and addressed important considerations in study design, as a means to promote wider data sharing and support larger meta-analysis of already available data and greater collaboration between researchers in order to fully capitalize on the potential of transcript profiling in future studies. SEARCH METHODS The PubMed database, National Center for Biotechnology Information and European Bioinformatics Institute dataset repositories were searched, to identify all relevant human studies using ‘placenta’, ‘decidua’, ‘trophoblast’, ‘transcriptome’, ‘microarray’ and ‘RNA sequencing’ as search terms until May 2019. Additional studies were found from bibliographies of identified studies. OUTCOMES The 179 identified studies were classifiable into four broad themes: healthy placental development, pregnancy complications, exposures during pregnancy and in vitro placental cultures. The median sample size was 13 (interquartile range 8–29). Transcriptome studies prior to 2015 were predominantly performed using microarrays, while RNA sequencing became the preferred choice in more recent studies. Development of fluidics technology, combined with RNA sequencing, has enabled transcript profiles to be generated of single cells throughout pregnancy, in contrast to previous studies relying on isolated cells. There are several key study aspects, such as sample selection criteria, sample processing and data analysis methods that may represent pitfalls and limitations, which need to be carefully considered as they influence interpretation of findings and conclusions. Furthermore, several areas of growing importance, such as maternal mental health and maternal obesity are understudied and the profiling of placentas from these conditions should be prioritized. WIDER IMPLICATIONS Integrative analysis of placental transcriptomics with other ‘omics’ (methylome, proteome and metabolome) and linkage with future outcomes from longitudinal studies is crucial in enhancing knowledge of healthy placental development and function, and in enabling the underlying causal mechanisms of pregnancy complications to be identified. Such understanding could help in predicting risk of future adversity and in designing interventions that can improve the health outcomes of both mothers and their offspring. Wider collaboration and sharing of placental transcriptome data, overcoming the challenges in obtaining sufficient numbers of quality samples with well-defined clinical characteristics, and dedication of resources to understudied areas of pregnancy will undoubtedly help drive the field forward.
Collapse
Affiliation(s)
- Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Shiao-Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Serebrova VN, Trifonova EA, Stepanov VA. Pregnancy as a Factor of Adaptive Human Evolution. The Role of Natural Selection in the Origin of Preeclampsia. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421010142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Rakner JJ, Silva GB, Mundal SB, Thaning AJ, Elschot M, Ostrop J, Thomsen LCV, Bjørge L, Gierman LM, Iversen AC. Decidual and placental NOD1 is associated with inflammation in normal and preeclamptic pregnancies. Placenta 2021; 105:23-31. [PMID: 33529885 DOI: 10.1016/j.placenta.2021.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 01/13/2021] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Inflammation is a normal physiological process that increases to harmful levels in preeclampsia. It affects the interaction between maternal immune cells and fetal trophoblasts at both sites of the maternal-fetal interface; decidua and placenta. The pattern recognition receptor nucleotide-binding oligomerization domain-containing protein (NOD)1 is expressed at both sites. This study aimed to characterize the cellular expression and functionality of NOD1 at the maternal-fetal interface of normal and preeclamptic pregnancies. METHODS Women with normal or preeclamptic pregnancies delivered by caesarean section were included. Decidual (n = 90) and placental (n = 91) samples were analyzed for NOD1 expression by immunohistochemistry and an automated image-based quantification method. Decidual and placental explants were incubated with or without the NOD1-agonist iE-DAP and cytokine responses measured by ELISA. RESULTS NOD1 was markedly expressed by maternal cells in the decidua and by fetal trophoblasts in both decidua and placenta, with trophoblasts showing the highest NOD1 expression. Preeclampsia with normal fetal growth was associated with a trophoblast-dependent increase in decidual NOD1 expression density. Compared to normal pregnancies, preeclampsia demonstrated stronger correlation between decidual and placental NOD1 expression levels. Increased production of interleukin (IL)-6 or IL-8 after in vitro explant stimulation confirmed NOD1 functionality. DISCUSSION These findings suggest that NOD1 contributes to inflammation at the maternal-fetal interface in normal pregnancies and preeclampsia and indicate a role in direct maternal-fetal communication. The strong expression of NOD1 by all trophoblast types highlights the importance of combined assessment of decidua and placenta for overall understanding of pathophysiological processes at the maternal-fetal interface.
Collapse
Affiliation(s)
- Johanne Johnsen Rakner
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Gabriela Brettas Silva
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Siv Boon Mundal
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Astrid Josefin Thaning
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Mattijs Elschot
- Department of Circulation and Medical Imaging, NTNU, Trondheim and Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jenny Ostrop
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Liv Cecilie Vestrheim Thomsen
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen and Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Line Bjørge
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen and Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lobke Marijn Gierman
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research (CEMIR) and Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Gynecology and Obstetrics, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
19
|
Vishnyakova P, Poltavets A, Nikitina M, Muminova K, Potapova A, Vtorushina V, Loginova N, Midiber K, Mikhaleva L, Lokhonina A, Khodzhaeva Z, Pyregov A, Elchaninov A, Fatkhudinov T, Sukhikh G. Preeclampsia: inflammatory signature of decidual cells in early manifestation of disease. Placenta 2021; 104:277-283. [PMID: 33472135 DOI: 10.1016/j.placenta.2021.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/28/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Preeclampsia is a pregnancy-specific complication characterized by hypertension in combination with proteinuria and/or various manifestations of multiple organ failure. It is believed that etiology of preeclampsia lies in dysfunction of the placenta and disorder of the maternal-fetal interactions. In preeclampsia decidual membrane, the maternal part of the placenta which normally supports immunological tolerance of the maternal organism to the semi-allogeneic fetus, becomes a site of inflammation. METHODS The aim of our study was to characterize the phenotype of decidual macrophages and plasma profiles in patients with late- and early-onset preeclampsia as compared with controls (n = 43). Decidual cells were obtained by enzymatic digestion method and characterized by flow cytometry analysis, real-time PCR, bioinformatics analysis, immunohistochemistry, and Western blot. Plasma samples were analyzed by multiplex assay. RESULTS The number of inflammation-associated CD86+ and CX3CR1+ cells was significantly higher in the early-onset preeclampsia while the portion of CD163+ cells was significantly higher among studied groups. We observed significant increase of endothelin-1 gene expression and a significant decrease in eNOS and GNB3 expression and TGFβ relative protein level in decidual cells of the early-onset preeclampsia samples. We also revealed elevation of pro- and anti-inflammatory cytokines in plasma of preeclampsia groups. DISCUSSION Our findings reflect profound early-onset preeclampsia-associated alterations in the decidua and emphasize the importance of the decidua as a link in the development of preeclampsia.
Collapse
Affiliation(s)
- P Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia; Peoples' Friendship University of Russia (RUDN University), 117198, Moscow, Russia.
| | - A Poltavets
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - M Nikitina
- Scientific Research Institute of Human Morphology, 117418, Moscow, Russia
| | - K Muminova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - A Potapova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - V Vtorushina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - N Loginova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - K Midiber
- Scientific Research Institute of Human Morphology, 117418, Moscow, Russia
| | - L Mikhaleva
- Scientific Research Institute of Human Morphology, 117418, Moscow, Russia
| | - A Lokhonina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia; Peoples' Friendship University of Russia (RUDN University), 117198, Moscow, Russia
| | - Z Khodzhaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - A Pyregov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - A Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| | - T Fatkhudinov
- Peoples' Friendship University of Russia (RUDN University), 117198, Moscow, Russia; Scientific Research Institute of Human Morphology, 117418, Moscow, Russia
| | - G Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997, Moscow, Russia
| |
Collapse
|
20
|
Santos HP, Bhattacharya A, Joseph RM, Smeester L, Kuban KCK, Marsit CJ, O'Shea TM, Fry RC. Evidence for the placenta-brain axis: multi-omic kernel aggregation predicts intellectual and social impairment in children born extremely preterm. Mol Autism 2020; 11:97. [PMID: 33308293 PMCID: PMC7730750 DOI: 10.1186/s13229-020-00402-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Children born extremely preterm are at heightened risk for intellectual and social impairment, including Autism Spectrum Disorder (ASD). There is increasing evidence for a key role of the placenta in prenatal developmental programming, suggesting that the placenta may, in part, contribute to origins of neurodevelopmental outcomes. METHODS We examined associations between placental transcriptomic and epigenomic profiles and assessed their ability to predict intellectual and social impairment at age 10 years in 379 children from the Extremely Low Gestational Age Newborn (ELGAN) cohort. Assessment of intellectual ability (IQ) and social function was completed with the Differential Ability Scales-II and Social Responsiveness Scale (SRS), respectively. Examining IQ and SRS allows for studying ASD risk beyond the diagnostic criteria, as IQ and SRS are continuous measures strongly correlated with ASD. Genome-wide mRNA, CpG methylation and miRNA were assayeds with the Illumina Hiseq 2500, HTG EdgeSeq miRNA Whole Transcriptome Assay, and Illumina EPIC/850 K array, respectively. We conducted genome-wide differential analyses of placental mRNA, miRNA, and CpG methylation data. These molecular features were then integrated for a predictive analysis of IQ and SRS outcomes using kernel aggregation regression. We lastly examined associations between ASD and the multi-omic-predicted component of IQ and SRS. RESULTS Genes with important roles in neurodevelopment and placental tissue organization were associated with intellectual and social impairment. Kernel aggregations of placental multi-omics strongly predicted intellectual and social function, explaining approximately 8% and 12% of variance in SRS and IQ scores via cross-validation, respectively. Predicted in-sample SRS and IQ showed significant positive and negative associations with ASD case-control status. LIMITATIONS The ELGAN cohort comprises children born pre-term, and generalization may be affected by unmeasured confounders associated with low gestational age. We conducted external validation of predictive models, though the sample size (N = 49) and the scope of the available out-sample placental dataset are limited. Further validation of the models is merited. CONCLUSIONS Aggregating information from biomarkers within and among molecular data types improves prediction of complex traits like social and intellectual ability in children born extremely preterm, suggesting that traits within the placenta-brain axis may be omnigenic.
Collapse
Affiliation(s)
- Hudson P Santos
- Biobehavioral Laboratory, School of Nursing, University of North Carolina, 544 Carrington Hall, Campus Box 7460, Chapel Hill, NC, 27599-7460, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Arjun Bhattacharya
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Robert M Joseph
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Lisa Smeester
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Karl C K Kuban
- Department of Pediatrics, Division of Pediatric Neurology, Boston University Medical Center, Boston, MA, USA
| | - Carmen J Marsit
- Department of Environmental Health, Emory University, Atlanta, GA, 30322, USA
| | - T Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
21
|
Guo H, Wang Y, Liu D. Silibinin ameliorats H 2O 2-induced cell apoptosis and oxidative stress response by activating Nrf2 signaling in trophoblast cells. Acta Histochem 2020; 122:151620. [PMID: 33068964 DOI: 10.1016/j.acthis.2020.151620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia is a pregnancy-specific syndrome and is one of the major causes of maternal mortality around the world. Cell apoptosis and oxidative stress are involved in development of preeclampsia. Silibinin has been known with anti-inflammatory, anti-oxidative and anti-tumor roles. In this study, hydrogen peroxide (H2O2) administration induced apoptosis in HTR-8/SVneo trophoblast cells, evidenced by decreased level of Bcl-2 and increased levels of Bax and cleaved caspase-3. Western blot and JC-1 staining revealed that H2O2 led to decline of mitochondrial membrane potential (Δψm) and release of cytochrome C from mitochondria to cytoplasm. H2O2 also resulted in reactive oxygen species production and oxidative stress response, evidenced by elevated levels of malondialdehyde, and reduced activity of superoxide dismutase and glutathione peroxidase. Silibinin suppressed H2O2-induced apoptosis, decrease of Δψm and oxidative stress response. In addition, immunofluorescent staining and electrophoretic mobility shift assay demonstrated that H2O2 enhanced expression and nuclear translocation of nuclear factor-erythroid 2-like 2 (Nrf2), and the expression levels of heme oxygenases-1 and quinone oxidoreductase 1 were increased, suggesting the activation of Nrf2 signaling. The activity of Nrf2 signaling was further promoted by silibinin administration. Interestingly, the effect of silibinin on apoptosis and oxidative stress was abolished by interference RNA of Nrf2. In conclusion, we demonstrated that silibinin ameliorated H2O2-induced apoptosis and oxidative stress response by activating Nrf2 signaling in trophoblast cells. These findings may provide novel insights for treatment of preeclampsia.
Collapse
|
22
|
Leseva MN, Binder AM, Ponsonby AL, Vuillermin P, Saffery R, Michels KB. Differential gene expression and limited epigenetic dysregulation at the materno-fetal interface in preeclampsia. Hum Mol Genet 2020; 29:335-350. [PMID: 31868881 DOI: 10.1093/hmg/ddz287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Despite the many advances made in the diagnosis and management of preeclampsia, this syndrome remains a leading cause of maternal mortality and life-long morbidity, as well as adverse fetal outcomes. Successful prediction and therapeutic intervention require an improved understanding of the molecular mechanisms, which underlie preeclampsia pathophysiology. We have used an integrated approach to discover placental genetic and epigenetic markers of preeclampsia and validated our findings in an independent cohort of women. We observed the microRNA, MIR138, to be upregulated in singleton preeclamptic placentas; however, this appears to be a female infant sex-specific effect. We did not identify any significant differentially methylated positions (DMPs) in singleton pregnancies, indicating that DNA methylation changes in mild forms of the disease are likely limited. However, we identified infant sex-specific preeclampsia-associated differentially methylated regions among singletons. Disease-associated DMPs were more obvious in a limited sampling of twin pregnancies. Interestingly, 2 out of the 10 most significant changes in methylation over larger regions overlap between singletons and twins and correspond to NAPRT1 and ZNF417.
Collapse
Affiliation(s)
- Milena N Leseva
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79110, Germany
| | - Alexandra M Binder
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Anne-Louise Ponsonby
- Discovery Theme, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.,Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Peter Vuillermin
- Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia.,School of Medicine, Deakin University, Geelong, Victoria 3220, Australia.,Child Health Research Unit, Barwon Health, Geelong, Victoria 3220, Australia
| | - Richard Saffery
- Cell Biology Theme, The Murdoch Children's Research Institute, Royal Children's Hospital, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Karin B Michels
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg 79110, Germany.,Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
23
|
Chen B, Hong W, Tang Y, Zhao Y, Aguilar ZP, Xu H. Protective effect of the NAC and Sal on zinc oxide nanoparticles-induced reproductive and development toxicity in pregnant mice. Food Chem Toxicol 2020; 143:111552. [PMID: 32640348 DOI: 10.1016/j.fct.2020.111552] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/22/2020] [Accepted: 06/20/2020] [Indexed: 12/12/2022]
Abstract
The growing use of zinc oxide nanoparticles (ZnO NPs) in various applications has raised many concerns about the potential risks to human health. In this research, the protective effects of cellular oxidative stress inhibitor N-Acetyl-cysteine (NAC) and endoplasmic reticulum (ER) stress inhibitor Salubrinal (Sal) on reproductive toxicity induced by ZnO NPs were investigated. The results showed that application of these two kinds of cell stress inhibitors after oral ingestion of ZnO NPs could prevent the weight loss of pregnant mice; reduce zinc content in the uterus, placenta and fetus; reduce abnormal development of the offspring; and decrease fetal abortion. Furthermore, RT-qPCR, Western blot and immunofluorescence assay results indicated that NAC restored the expression of Gclc, reduced the expression of ATF4, JNK and Caspase-12, and decreased the expression of eNOS and IGF-1, in the placenta. Sal decreased the expression of ATF4, JNK and Caspase-12, and increased the expression of eNOS and IGF-1caused by the oral ingestion of ZnO NPs. These results indicated that treatment with NAC and Sal after oral exposure could reduce reproductive and development toxicity caused by ZnO NPs which induced reproductive and development toxicity that was probably caused by the activation of oxide stress and ER stress.
Collapse
Affiliation(s)
- Bolu Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Wuding Hong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yizhou Tang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yu Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | | | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China.
| |
Collapse
|
24
|
Hao S, You J, Chen L, Zhao H, Huang Y, Zheng L, Tian L, Maric I, Liu X, Li T, Bianco YK, Winn VD, Aghaeepour N, Gaudilliere B, Angst MS, Zhou X, Li YM, Mo L, Wong RJ, Shaw GM, Stevenson DK, Cohen HJ, Mcelhinney DB, Sylvester KG, Ling XB. Changes in pregnancy-related serum biomarkers early in gestation are associated with later development of preeclampsia. PLoS One 2020; 15:e0230000. [PMID: 32126118 PMCID: PMC7053753 DOI: 10.1371/journal.pone.0230000] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/19/2020] [Indexed: 12/19/2022] Open
Abstract
Background Placental protein expression plays a crucial role during pregnancy. We hypothesized that: (1) circulating levels of pregnancy-associated, placenta-related proteins throughout gestation reflect the temporal progression of the uncomplicated, full-term pregnancy, and can effectively estimate gestational ages (GAs); and (2) preeclampsia (PE) is associated with disruptions in these protein levels early in gestation; and can identify impending PE. We also compared gestational profiles of proteins in the human and mouse, using pregnant heme oxygenase-1 (HO-1) heterozygote (Het) mice, a mouse model reflecting PE-like symptoms. Methods Serum levels of placenta-related proteins–leptin (LEP), chorionic somatomammotropin hormone like 1 (CSHL1), elabela (ELA), activin A, soluble fms-like tyrosine kinase 1 (sFlt-1), and placental growth factor (PlGF)–were quantified by ELISA in blood serially collected throughout human pregnancies (20 normal subjects with 66 samples, and 20 subjects who developed PE with 61 samples). Multivariate analysis was performed to estimate the GA in normal pregnancy. Mean-squared errors of GA estimations were used to identify impending PE. The human protein profiles were then compared with those in the pregnant HO-1 Het mice. Results An elastic net-based gestational dating model was developed (R2 = 0.76) and validated (R2 = 0.61) using serum levels of the 6 proteins measured at various GAs from women with normal uncomplicated pregnancies. In women who developed PE, the model was not (R2 = -0.17) associated with GA. Deviations from the model estimations were observed in women who developed PE (P = 0.01). The model developed with 5 proteins (ELA excluded) performed similarly from sera from normal human (R2 = 0.68) and WT mouse (R2 = 0.85) pregnancies. Disruptions of this model were observed in both human PE-associated (R2 = 0.27) and mouse HO-1 Het (R2 = 0.30) pregnancies. LEP outperformed sFlt-1 and PlGF in differentiating impending PE at early human and late mouse GAs. Conclusions Serum placenta-related protein profiles are temporally regulated throughout normal pregnancies and significantly disrupted in women who develop PE. LEP changes earlier than the well-established biomarkers (sFlt-1 and PlGF). There may be evidence of a causative action of HO-1 deficiency in LEP upregulation in a PE-like murine model.
Collapse
Affiliation(s)
- Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Jin You
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Lin Chen
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Hui Zhao
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Yujuan Huang
- Department of Emergency, Shanghai Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Le Zheng
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Lu Tian
- Department of Health Research and Policy, Stanford University, Stanford, CA, United States of America
| | - Ivana Maric
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xin Liu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Tian Li
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ylayaly K. Bianco
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco-Fresno, Fresno, CA, United States of America
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Harvey J. Cohen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Doff B. Mcelhinney
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Xuefeng B. Ling
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children’s Hospital, Palo Alto, CA, United States of America
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Massimiani M, Lacconi V, La Civita F, Ticconi C, Rago R, Campagnolo L. Molecular Signaling Regulating Endometrium-Blastocyst Crosstalk. Int J Mol Sci 2019; 21:E23. [PMID: 31861484 PMCID: PMC6981505 DOI: 10.3390/ijms21010023] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/29/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Implantation of the embryo into the uterine endometrium is one of the most finely-regulated processes that leads to the establishment of a successful pregnancy. A plethora of factors are released in a time-specific fashion to synchronize the differentiation program of both the embryo and the endometrium. Indeed, blastocyst implantation in the uterus occurs in a limited time frame called the "window of implantation" (WOI), during which the maternal endometrium undergoes dramatic changes, collectively called "decidualization". Decidualization is guided not just by maternal factors (e.g., estrogen, progesterone, thyroid hormone), but also by molecules secreted by the embryo, such as chorionic gonadotropin (CG) and interleukin-1β (IL-1 β), just to cite few. Once reached the uterine cavity, the embryo orients correctly toward the uterine epithelium, interacts with specialized structures, called pinopodes, and begins the process of adhesion and invasion. All these events are guided by factors secreted by both the endometrium and the embryo, such as leukemia inhibitory factor (LIF), integrins and their ligands, adhesion molecules, Notch family members, and metalloproteinases and their inhibitors. The aim of this review is to give an overview of the factors and mechanisms regulating implantation, with a focus on those involved in the complex crosstalk between the blastocyst and the endometrium.
Collapse
Affiliation(s)
- Micol Massimiani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
- Saint Camillus International University of Health Sciences, Via di Sant’Alessandro, 8, 00131 Rome, Italy
| | - Valentina Lacconi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| | - Fabio La Civita
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| | - Carlo Ticconi
- Department of Surgical Sciences, Section of Gynecology and Obstetrics, University Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy;
| | - Rocco Rago
- Physiopathology of Reproduction and Andrology Unit, Sandro Pertini Hospital, Via dei Monti Tiburtini 385/389, 00157 Rome, Italy;
| | - Luisa Campagnolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (M.M.); (V.L.); (F.L.C.)
| |
Collapse
|
26
|
RITA Is Expressed in Trophoblastic Cells and Is Involved in Differentiation Processes of the Placenta. Cells 2019; 8:cells8121484. [PMID: 31766533 PMCID: PMC6953008 DOI: 10.3390/cells8121484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Preeclampsia (PE) remains a leading cause of maternal and perinatal mortality and morbidity worldwide. Its pathogenesis has not been fully elucidated and no causal therapy is currently available. It is of clinical relevance to decipher novel molecular biomarkers. RITA (RBP-J (recombination signal binding protein J)-interacting and tubulin-associated protein) has been identified as a negative modulator of the Notch pathway and as a microtubule-associated protein important for cell migration and invasion. In the present work, we have systematically studied RITA’s expression in primary placental tissues from patients with early- and late-onset PE as well as in various trophoblastic cell lines. RITA is expressed in primary placental tissues throughout gestation, especially in proliferative villous cytotrophoblasts, in the terminally differentiated syncytiotrophoblast, and in migrating extravillous trophoblasts. RITA’s messenger RNA (mRNA) level is decreased in primary tissue samples from early-onset PE patients. The deficiency of RITA impairs the motility and invasion capacity of trophoblastic cell lines, and compromises the fusion ability of trophoblast-derived choriocarcinoma cells. These data suggest that RITA may play important roles in the development of the placenta and possibly in the pathogenesis of PE.
Collapse
|
27
|
Snyder-Talkington BN, Dong C, Castranova V, Qian Y, Guo NL. Differential gene regulation in human small airway epithelial cells grown in monoculture versus coculture with human microvascular endothelial cells following multiwalled carbon nanotube exposure. Toxicol Rep 2019; 6:482-488. [PMID: 31194188 PMCID: PMC6554470 DOI: 10.1016/j.toxrep.2019.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 05/08/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022] Open
Abstract
Coculture gene expression may have opposite direction of changes than monoculture. Cells grow and treated in monoculture may exaggerate toxicological responses. Coculture of cells may provide a more in-depth assessment of toxicological responses.
Concurrent with rising production of carbon-based engineered nanomaterials is a potential increase in respiratory and cardiovascular diseases due to exposure to nanomaterials in the workplace atmosphere. While single-cell models of pulmonary exposure are often used to determine the potential toxicity of nanomaterials in vitro, previous studies have shown that coculture cell models better represent the cellular response and crosstalk that occurs in vivo. This study identified differential gene regulation in human small airway epithelial cells (SAECs) grown either in monoculture or in coculture with human microvascular endothelial cells following exposure of the SAECs to multiwalled carbon nanotubes (MWCNTs). SAEC genes that either changed their regulation direction from upregulated in monoculture to downregulated in coculture (or vice versa) or had a more than a two-fold changed in the same regulation direction were identified. Genes that changed regulation direction were most often involved in the processes of cellular growth and proliferation and cellular immune response and inflammation. Genes that had a more than a two-fold change in regulation in the same direction were most often involved in the inflammatory response. The direction and fold-change of this differential gene regulation suggests that toxicity testing in monoculture may exaggerate cellular responses to MWCNTs, and coculture of cells may provide a more in-depth assessment of toxicological responses.
Collapse
Affiliation(s)
- Brandi N Snyder-Talkington
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States
| | - Chunlin Dong
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States
| | - Vincent Castranova
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, United States
| | - Yong Qian
- National Institute for Occupational and Environmental Safety and Health, 1095 Willowdale Rd., Morgantown, WV, 26505, United States
| | - Nancy L Guo
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States.,Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV, 26506, United States
| |
Collapse
|
28
|
Reply to Liu et al.: Decidualization defect in severe preeclampsia. Proc Natl Acad Sci U S A 2018; 115:E7656-E7657. [PMID: 30068606 DOI: 10.1073/pnas.1802916115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Xu K, Liu G, Fu C. The Tryptophan Pathway Targeting Antioxidant Capacity in the Placenta. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1054797. [PMID: 30140360 PMCID: PMC6081554 DOI: 10.1155/2018/1054797] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Abstract
The placenta plays a vital role in fetal development during pregnancy. Dysfunction of the placenta can be caused by oxidative stress and can lead to abnormal fetal development. Preventing oxidative stress of the placenta is thus an important measure to ensure positive birth outcomes. Research shows that tryptophan and its metabolites can efficiently clean free radicals (including the reactive oxygen species and activated chlorine). Consequently, tryptophan and its metabolites are suggested to act as potent antioxidants in the placenta. However, the mechanism of these antioxidant properties in the placenta is still unknown. In this review, we summarize research on the antioxidant properties of tryptophan, tryptophan metabolites, and metabolic enzymes. Two predicted mechanisms of tryptophan's antioxidant properties are discussed. (1) Tryptophan could activate the phosphorylation of p62 after the activation of mTORC1; phosphorylated p62 then uncouples the interaction between Nrf2 and Keap1, and activated Nrf2 enters the nucleus to induce expressions of antioxidant proteins, thus improving cellular antioxidation. (2) 3-Hydroxyanthranilic acid, a tryptophan kynurenine pathway metabolite, changes conformation of Keap1, inducing the dissociation of Nrf2 and Keap1, activating Nrf2 to enter the nucleus and induce expressions of antioxidant proteins (such as HO-1), thereby enhancing cellular antioxidant capacity. These mechanisms may enrich the theory of how to apply tryptophan as an antioxidant during pregnancy, providing technical support for its use in regulating the pregnancy's redox status and enriching our understanding of amino acids' nutritional value.
Collapse
Affiliation(s)
- Kang Xu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Gang Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, Hunan 410125, China
| | - Chenxing Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan 410128, China
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients and Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, Hunan 410128, China
| |
Collapse
|
30
|
Transcriptional signature of the decidua in preeclampsia. Proc Natl Acad Sci U S A 2018; 115:E5434-E5436. [PMID: 29848633 DOI: 10.1073/pnas.1802234115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
31
|
LNX1/LNX2 proteins: functions in neuronal signalling and beyond. Neuronal Signal 2018; 2:NS20170191. [PMID: 32714586 PMCID: PMC7373230 DOI: 10.1042/ns20170191] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/02/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022] Open
Abstract
Ligand of NUMB Protein X1 and X2 (LNX1 and LNX2) are E3 ubiquitin ligases, named for their ability to interact with and promote the degradation of the cell fate determinant protein NUMB. On this basis they are thought to play a role in modulating NUMB/NOTCH signalling during processes such as cortical neurogenesis. However, LNX1/2 proteins can bind, via their four PDZ (PSD95, DLGA, ZO-1) domains, to an extraordinarily large number of other proteins besides NUMB. Many of these interactions suggest additional roles for LNX1/2 proteins in the nervous system in areas such as synapse formation, neurotransmission and regulating neuroglial function. Twenty years on from their initial discovery, I discuss here the putative neuronal functions of LNX1/2 proteins in light of the anxiety-related phenotype of double knockout mice lacking LNX1 and LNX2 in the central nervous system (CNS). I also review what is known about non-neuronal roles of LNX1/2 proteins, including their roles in embryonic patterning and pancreas development in zebrafish and their possible involvement in colorectal cancer (CRC), osteoclast differentiation and immune function in mammals. The emerging picture places LNX1/2 proteins as potential regulators of multiple cellular signalling processes, but in many cases the physiological significance of such roles remains only partly validated and needs to be considered in the context of the tight control of LNX1/2 protein levels in vivo.
Collapse
|
32
|
The genetic component of preeclampsia: A whole-exome sequencing study. PLoS One 2018; 13:e0197217. [PMID: 29758065 PMCID: PMC5951572 DOI: 10.1371/journal.pone.0197217] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/27/2018] [Indexed: 01/08/2023] Open
Abstract
Preeclampsia is a major cause of maternal and perinatal deaths. The aetiology of preeclampsia is largely unknown but a polygenetic component is assumed. To explore this hypothesis, we performed an in-depth whole-exome sequencing study in women with (cases, N = 50) and without (controls, N = 50) preeclampsia. The women were identified in an unselected cohort of 2,545 pregnant women based on data from the Danish National Patient Registry and the Medical Birth Registry. Matching DNA was obtained from a biobank containing excess blood from routine antenatal care visits. Novogene performed the whole-exome sequencing blinded to preeclampsia status. Variants for comparison between cases and controls were filtered in the Ingenuity Variant Analysis software. We applied two different strategies; a disease association panel approach, which included variants in single genes associated with established clinical risk factors for preeclampsia, and a gene panel approach, which included biological pathways harbouring genes previously reported to be associated with preeclampsia. Variant variability was compared in cases and controls at the level of biological processes, signalling pathways, and in single genes. Regardless of the applied strategy and the level of variability examined, we consistently found positive correlations between variant numbers in cases and controls (all R2s>0.88). Contrary to what was expected, cases carried fewer variants in biological processes and signalling pathways than controls (all p-values ≤0.02). In conclusion, our findings challenge the hypothesis of a polygenetic aetiology for preeclampsia with a common network of susceptibility genes. The greater genetic diversity among controls may suggest a protective role of genetic diversity against the development of preeclampsia.
Collapse
|
33
|
Zhao Y, Dong D, Reece EA, Wang AR, Yang P. Oxidative stress-induced miR-27a targets the redox gene nuclear factor erythroid 2-related factor 2 in diabetic embryopathy. Am J Obstet Gynecol 2018; 218:136.e1-136.e10. [PMID: 29100869 DOI: 10.1016/j.ajog.2017.10.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Maternal diabetes induces neural tube defects, and oxidative stress is a causal factor for maternal diabetes-induced neural tube defects. The redox gene nuclear factor erythroid 2-related factor 2 is the master regulator of the cellular antioxidant system. OBJECTIVE In this study, we aimed to determine whether maternal diabetes inhibits nuclear factor erythroid 2-related factor 2 expression and nuclear factor erythroid 2-related factor 2-controlled antioxidant genes through the redox-sensitive miR-27a. STUDY DESIGN We used a well-established type 1 diabetic embryopathy mouse model induced by streptozotocin for our in vivo studies. Embryos at embryonic day 8.5 were harvested for analysis of nuclear factor erythroid 2-related factor 2, nuclear factor erythroid 2-related factor 2-controlled antioxidant genes, and miR-27a expression. To determine if mitigating oxidative stress inhibits the increase of miR-27a and the decrease of nuclear factor erythroid 2-related factor 2 expression, we induced diabetic embryopathy in superoxide dismutase 2 (mitochondrial-associated antioxidant gene)-overexpressing mice. This model exhibits reduced mitochondria reactive oxygen species even in the presence of hyperglycemia. To investigate the causal relationship between miR-27a and nuclear factor erythroid 2-related factor 2 in vitro, we examined C17.2 neural stem cells under normal and high-glucose conditions. RESULTS We observed that the messenger RNA and protein levels of nuclear factor erythroid 2-related factor 2 were significantly decreased in embryos on embryonic day 8.5 from diabetic dams compared to those from nondiabetic dams. High-glucose also significantly decreased nuclear factor erythroid 2-related factor 2 expression in a dose- and time-dependent manner in cultured neural stem cells. Our data revealed that miR-27a was up-regulated in embryos on embryonic day 8.5 exposed to diabetes, and that high glucose increased miR-27a levels in a dose- and time-dependent manner in cultured neural stem cells. In addition, we found that a miR-27a inhibitor abrogated the inhibitory effect of high glucose on nuclear factor erythroid 2-related factor 2 expression, and a miR-27a mimic suppressed nuclear factor erythroid 2-related factor 2 expression in cultured neural stem cells. Furthermore, our data indicated that the nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1 were down-regulated by maternal diabetes in embryos on embryonic day 8.5 and high glucose in cultured neural stem cells. Inhibiting miR-27a restored expression of glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione S-transferase A1. Overexpressing superoxide dismutase 2 reversed the maternal diabetes-induced increase of miR-27a and suppression of nuclear factor erythroid 2-related factor 2 and nuclear factor erythroid 2-related factor 2-controlled antioxidant enzymes. CONCLUSION Our study demonstrates that maternal diabetes-induced oxidative stress increases miR-27a, which, in turn, suppresses nuclear factor erythroid 2-related factor 2 and its responsive antioxidant enzymes, resulting in diabetic embryopathy.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Daoyin Dong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD
| | - Ashley R Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD
| | - Peixin Yang
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Chashan University-town, Wenzhou, Zhejiang, China; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD.
| |
Collapse
|
34
|
Swegen A, Grupen CG, Gibb Z, Baker MA, Ruijter‐Villani M, Smith ND, Stout TAE, Aitken RJ. From Peptide Masses to Pregnancy Maintenance: A Comprehensive Proteomic Analysis of The Early Equine Embryo Secretome, Blastocoel Fluid, and Capsule. Proteomics 2017; 17. [DOI: 10.1002/pmic.201600433] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 07/19/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Aleona Swegen
- Priority Research Centre in Reproductive Science University of Newcastle Callaghan NSW Australia
| | - Christopher G. Grupen
- Faculty of Veterinary Science School of Life and Environmental Sciences University of Sydney Camden NSW Australia
| | - Zamira Gibb
- Priority Research Centre in Reproductive Science University of Newcastle Callaghan NSW Australia
| | - Mark A. Baker
- Priority Research Centre in Reproductive Science University of Newcastle Callaghan NSW Australia
| | - Marta Ruijter‐Villani
- Department of Equine Sciences Faculty of Veterinary Medicine Utrecht University Utrecht the Netherlands
| | - Nathan D. Smith
- Analytical and Biomolecular Research Facility University of Newcastle Callaghan NSW Australia
| | - Tom A. E. Stout
- Department of Equine Sciences Faculty of Veterinary Medicine Utrecht University Utrecht the Netherlands
| | - R. John Aitken
- Priority Research Centre in Reproductive Science University of Newcastle Callaghan NSW Australia
| |
Collapse
|
35
|
Transcriptomic Profiling in Human Decidua of Severe Preeclampsia Detected by RNA Sequencing. J Cell Biochem 2017; 119:607-615. [DOI: 10.1002/jcb.26221] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/14/2017] [Indexed: 12/27/2022]
|
36
|
Emerging role for dysregulated decidualization in the genesis of preeclampsia. Placenta 2017; 60:119-129. [PMID: 28693893 DOI: 10.1016/j.placenta.2017.06.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/10/2017] [Accepted: 06/07/2017] [Indexed: 12/31/2022]
Abstract
In normal human placentation, uterine invasion by trophoblast cells and subsequent spiral artery remodeling depend on cooperation among fetal trophoblasts and maternal decidual, myometrial, immune and vascular cells in the uterine wall. Therefore, aberrant function of anyone or several of these cell-types could theoretically impair placentation leading to the development of preeclampsia. Because trophoblast invasion and spiral artery remodeling occur during the first half of pregnancy, the molecular pathology of fetal placental and maternal decidual tissues following delivery may not be informative about the genesis of impaired placentation, which transpired months earlier. Therefore, in this review, we focus on the emerging prospective evidence supporting the concept that deficient or defective endometrial maturation in the late secretory phase and during early pregnancy, i.e., pre-decidualization and decidualization, respectively, may contribute to the genesis of preeclampsia. The first prospectively-acquired data directly supporting this concept were unexpectedly revealed in transcriptomic analyses of chorionic villous samples (CVS) obtained during the first trimester of women who developed preeclampsia 5 months later. Additional supportive evidence arose from investigations of Natural Killer cells in first trimester decidua from elective terminations of women with high resistance uterine artery indices, a surrogate for deficient trophoblast invasion. Last, circulating insulin growth factor binding protein-1, which is secreted by decidual stromal cells was decreased during early pregnancy in women who developed preeclampsia. We conclude this review by making recommendations for further prospectively-designed studies to corroborate the concept of endometrial antecedents of preeclampsia. These studies could also enable identification of women at increased risk for developing preeclampsia, unveil the molecular mechanisms of deficient or defective (pre)decidualization, and lead to preventative strategies designed to improve (pre)decidualization, thereby reducing risk for preeclampsia development.
Collapse
|
37
|
Nezu M, Souma T, Yu L, Sekine H, Takahashi N, Wei AZS, Ito S, Fukamizu A, Zsengeller ZK, Nakamura T, Hozawa A, Karumanchi SA, Suzuki N, Yamamoto M. Nrf2 inactivation enhances placental angiogenesis in a preeclampsia mouse model and improves maternal and fetal outcomes. Sci Signal 2017; 10:10/479/eaam5711. [DOI: 10.1126/scisignal.aam5711] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Hayashi KG, Hosoe M, Kizaki K, Fujii S, Kanahara H, Takahashi T, Sakumoto R. Differential gene expression profiling of endometrium during the mid-luteal phase of the estrous cycle between a repeat breeder (RB) and non-RB cows. Reprod Biol Endocrinol 2017; 15:20. [PMID: 28335821 PMCID: PMC5364712 DOI: 10.1186/s12958-017-0237-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 03/03/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Repeat breeding directly affects reproductive efficiency in cattle due to an increase in services per conception and calving interval. This study aimed to investigate whether changes in endometrial gene expression profile are involved in repeat breeding in cows. Differential gene expression profiles of the endometrium were investigated during the mid-luteal phase of the estrous cycle between repeat breeder (RB) and non-RB cows using microarray analysis. METHODS The caruncular (CAR) and intercaruncular (ICAR) endometrium of both ipsilateral and contralateral uterine horns to the corpus luteum were collected from RB (inseminated at least three times but not pregnant) and non-RB cows on Day 15 of the estrous cycle (4 cows/group). Global gene expression profiles of these endometrial samples were analyzed with a 15 K custom-made oligo-microarray for cattle. Immunohistochemistry was performed to investigate the cellular localization of proteins of three identified transcripts in the endometrium. RESULTS Microarray analysis revealed that 405 and 397 genes were differentially expressed in the CAR and ICAR of the ipsilateral uterine horn of RB, respectively when compared with non-RB cows. In the contralateral uterine horn, 443 and 257 differentially expressed genes were identified in the CAR and ICAR of RB, respectively when compared with non-RB cows. Gene ontology analysis revealed that genes involved in development and morphogenesis were mainly up-regulated in the CAR of RB cows. In the ICAR of both the ipsilateral and contralateral uterine horns, genes related to the metabolic process were predominantly enriched in the RB cows when compared with non-RB cows. In the analysis of the whole uterus (combining the data above four endometrial compartments), RB cows showed up-regulation of 37 genes including PRSS2, GSTA3 and PIPOX and down-regulation of 39 genes including CHGA, KRT35 and THBS4 when compared with non-RB cows. Immunohistochemistry revealed that CHGA, GSTA3 and PRSS2 proteins were localized in luminal and glandular epithelial cells and stroma of the endometrium. CONCLUSION The present study showed that endometrial gene expression profiles are different between RB and non-RB cows. The identified candidate endometrial genes and functions in each endometrial compartment may contribute to bovine reproductive performance.
Collapse
Affiliation(s)
- Ken-Go Hayashi
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Misa Hosoe
- 0000 0001 2222 0432grid.416835.dDivision of Animal Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, 305-8602 Japan
| | - Keiichiro Kizaki
- 0000 0001 0018 0409grid.411792.8Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, 020-8550 Japan
| | - Shiori Fujii
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Hiroko Kanahara
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| | - Toru Takahashi
- 0000 0001 0018 0409grid.411792.8Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, 020-8550 Japan
| | - Ryosuke Sakumoto
- 0000 0000 9191 6962grid.419600.aDivision of Animal Breeding and Reproduction Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901 Japan
| |
Collapse
|
39
|
Buckberry S, Bianco-Miotto T, Bent SJ, Clifton V, Shoubridge C, Shankar K, Roberts CT. Placental transcriptome co-expression analysis reveals conserved regulatory programs across gestation. BMC Genomics 2017; 18:10. [PMID: 28049421 PMCID: PMC5209944 DOI: 10.1186/s12864-016-3384-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 12/07/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Mammalian development in utero is absolutely dependent on proper placental development, which is ultimately regulated by the placental genome. The regulation of the placental genome can be directly studied by exploring the underlying organisation of the placental transcriptome through a systematic analysis of gene-wise co-expression relationships. RESULTS In this study, we performed a comprehensive analysis of human placental co-expression using RNA sequencing and intergrated multiple transcriptome datasets spanning human gestation. We identified modules of co-expressed genes that are preserved across human gestation, and also identifed modules conserved in the mouse indicating conserved molecular networks involved in placental development and gene expression patterns more specific to late gestation. Analysis of co-expressed gene flanking sequences indicated that conserved co-expression modules in the placenta are regulated by a core set of transcription factors, including ZNF423 and EBF1. Additionally, we identified a gene co-expression module enriched for genes implicated in the pregnancy pathology preeclampsia. By using an independnet transcriptome dataset, we show that these co-expressed genes are differentially expressed in preeclampsia. CONCLUSIONS This study represents a comprehensive characterisation of placental co-expression and provides insight into potential transcriptional regulators that govern conserved molecular programs fundamental to placental development.
Collapse
Affiliation(s)
- Sam Buckberry
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia.,University of Western Australia, Harry Perkins Institute of Medical Research, Perth, 6009, Australia.,University of Western Australia, Australian Research Council Centre of Excellence in Plant Energy Biology, Perth, 6009, Australia
| | - Tina Bianco-Miotto
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia.,The University of Adelaide, School of agriculture, food and wine, Adelaide, 5005, Australia
| | - Stephen J Bent
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia
| | - Vicki Clifton
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia
| | - Cheryl Shoubridge
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia
| | - Kartik Shankar
- University of Arkansas for Medical Sciences, The Department of Pediatrics, Little Rock, 72202, USA
| | - Claire T Roberts
- The Robinson Research Institute, The University of Adelaide, School of Paediatrics and Reproductive Health, Adelaide, 5005, Australia.
| |
Collapse
|
40
|
Li J, Zhou J, Ye Y, Liu Q, Wang X, Zhang N, Wang X. Increased Heme Oxygenase-1 and Nuclear Factor Erythroid 2-Related Factor-2 in the Placenta Have a Cooperative Action on Preeclampsia. Gynecol Obstet Invest 2016; 81:543-551. [PMID: 27764834 DOI: 10.1159/000451025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/26/2016] [Indexed: 04/13/2024]
Abstract
BACKGROUND Previous studies have shown that oxidative stress is an important factor in preeclampsia (PE). Heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor-2 (Nrf2) are protective proteins that are involved in combating oxidative stress in the body. Nrf2 is also an essential upstream transcription factor regulating HO-1. This study was aimed at exploring the physiological roles of HO-1 and Nrf2 in PE. METHODS Serum and placenta were collected from 30 patients who presented with severe PE and 30 healthy pregnant females. HO-1 and Nrf2 levels in placenta were measured. Following stimulation of the HTR-8/SVneo cell line with tert-butylhydroquinone (tBHQ), an Nrf2 activator, nuclear Nrf2 protein and HO-1 mRNA levels were determined. RESULTS Compared with the healthy pregnancy group, HO-1 protein and mRNA levels were increased in placental samples obtained from the severe PE group (p < 0.01, p < 0.05). Similar increases were also observed for Nrf2 protein levels (p < 0.01). Nuclear Nrf2 protein and HO-1 mRNA levels were both increased in the HTR-8/SVneo cell line following stimulation with tBHQ (p < 0.05). CONCLUSION Patients with severe PE may be protected against oxidative injury following an elevation in HO-1 and Nrf2 levels. Nrf2 is likely to have a synergistic effect on HO-1 in PE.
Collapse
Affiliation(s)
- Jing Li
- Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | | | | | | | | | | |
Collapse
|
41
|
Eidem HR, Rinker DC, Ackerman WE, Buhimschi IA, Buhimschi CS, Dunn-Fletcher C, Kallapur SG, Pavličev M, Muglia LJ, Abbot P, Rokas A. Comparing human and macaque placental transcriptomes to disentangle preterm birth pathology from gestational age effects. Placenta 2016; 41:74-82. [PMID: 27208410 DOI: 10.1016/j.placenta.2016.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/05/2016] [Accepted: 03/10/2016] [Indexed: 11/26/2022]
Abstract
INTRODUCTION A major issue in the transcriptomic study of spontaneous preterm birth (sPTB) in humans is the inability to collect healthy control tissue at the same gestational age (GA) to compare with pathologic preterm tissue. Thus, gene expression differences identified after the standard comparison of sPTB and term tissues necessarily reflect differences in both sPTB pathology and GA. One potential solution is to use GA-matched controls from a closely related species to tease apart genes that are dysregulated during sPTB from genes that are expressed differently as a result of GA effects. METHODS To disentangle genes whose expression levels are associated with sPTB pathology from those linked to GA, we compared RNA sequencing data from human preterm placentas, human term placentas, and rhesus macaque placentas at 80% completed gestation (serving as healthy non-human primate GA-matched controls). We first compared sPTB and term human placental transcriptomes to identify significantly differentially expressed genes. We then overlaid the results of the comparison between human sPTB and macaque placental transcriptomes to identify sPTB-specific candidates. Finally, we overlaid the results of the comparison between human term and macaque placental transcriptomes to identify GA-specific candidates. RESULTS Examination of relative expression for all human genes with macaque orthologs identified 267 candidate genes that were significantly differentially expressed between preterm and term human placentas. 29 genes were identified as sPTB-specific candidates and 37 as GA-specific candidates. Altogether, the 267 differentially expressed genes were significantly enriched for a variety of developmental, metabolic, reproductive, immune, and inflammatory functions. Although there were no notable differences between the functions of the 29 sPTB-specific and 37 GA-specific candidate genes, many of these candidates have been previously shown to be dysregulated in diverse pregnancy-associated pathologies. DISCUSSION By comparing human sPTB and term transcriptomes with GA-matched control transcriptomes from a closely related species, this study disentangled the confounding effects of sPTB pathology and GA, leading to the identification of 29 promising sPTB-specific candidate genes and 37 genes potentially related to GA effects. The apparent similarity in functions of the sPTB and GA candidates may suggest that the effects of sPTB and GA do not correspond to biologically distinct processes. Alternatively, it may reflect the poor state of knowledge of the transcriptional landscape underlying placental development and disease.
Collapse
Affiliation(s)
- Haley R Eidem
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA.
| | - David C Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Program in Human Genetics, Vanderbilt University, Nashville, TN 37235, USA.
| | - William E Ackerman
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH 43210, USA.
| | - Irina A Buhimschi
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH 43210, USA.
| | - Catalin S Buhimschi
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH 43210, USA.
| | - Caitlin Dunn-Fletcher
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA.
| | - Suhas G Kallapur
- Center for Perinatal Research, Nationwide Children's Hospital, Columbus, OH 43210, USA.
| | - Mihaela Pavličev
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA.
| | - Louis J Muglia
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA.
| | - Patrick Abbot
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA.
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Program in Human Genetics, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
42
|
LIU KS, PENG ZH, CHENG WJ, DAI CF, TONG H. Endoplasmic reticulum stress-induced apoptosis in the development of reproduction. REPRODUCTION AND CONTRACEPTION 2016; 27:51-59. [PMID: 32288402 PMCID: PMC7129369 DOI: 10.7669/j.issn.1001-7844.2016.01.0051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Indexed: 12/21/2022]
Abstract
Proteins synthesized in the endoplasmic reticulum (ER) are properly folded with the assistance of ER chaperones. Accumulation of misfolded protein in the ER triggers an adaptive ER stress (ERS) response termed the unfolded protein response. Recent interest has focused on the possibility that the accumulation of misfolded proteins can also contribute to reproductive response, including preimplantation embryos, testicular germ cell, placenta, and unexplained intrauterine growth restriction (IUGR). The major ERS pathway constituents are present at all stages of preimplantation development and that the activation of ERS pathways can be induced at the 8-cell, morula and blastocyst stage. This review mainly introduced the research progress of ERS induced apoptosis of reproductive cells, providing a new direction for the research of reproductive disease therapy.
Collapse
Affiliation(s)
- Kang-sheng LIU
- Department of Clinical Laboratory, State Key Laboratory of Reproductive Medicine Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Zheng-hang PENG
- School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Weng-jun CHENG
- Department of Maternity and Child Health Care, School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Chun-fan DAI
- Department of Obstetrical, Nanjing Maternity and Child Health Care Hospital, Nanjing 210009, China
| | - Hua TONG
- Department of Scientific Research and Education, Nanjing Maternity and Child Health Care Hospital, Nanjing 210009, China
| |
Collapse
|
43
|
Abstract
The focus on disease mechanisms underlying the hypertension and proteinuria defining preeclampsia has increased knowledge of the pathophysiology yet we lack both therapy and predictors. We propose this is in part due to the fact that diagnostic findings identify a "preeclampsia syndrome" but do not necessarily indicate the most important pathophysiology nor if organs are involved as cause or consequence. The increased risk for later life cardiovascular disease in women who develop preeclampsia suggests the stress test of pregnancy exposes pre-existing subclinical vascular disease. The dogma that inadequate trophoblast invasion and ischemia/reperfusion injury to the placenta is "the" cause of preeclampsia is more relevant to early onset preeclampsia (<34 weeks). There is much less evidence for defective placentation in late onset preeclampsia where maternal constitutive factors or susceptibility to vascular damage is more relevant. The contribution of differing disease phenotypes to the syndrome may explain the inability of biomarker studies to identify all preeclampsia. Identification of phenotypes will require large amounts of prospective clinical data and biospecimens, collected in a harmonized manner with analysis in an unbiased discovery approach.
Collapse
|
44
|
Alam SMK, Konno T, Soares MJ. Identification of target genes for a prolactin family paralog in mouse decidua. Reproduction 2016; 149:625-32. [PMID: 25926690 DOI: 10.1530/rep-15-0107] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Prolactin family 8, subfamily a, member 2 (PRL8A2; also called decidual prolactin-related protein; dPRP) is a member of the expanded prolactin family. PRL8A2 is expressed in the uterine decidua and contributes to pregnancy-dependent adaptations to hypoxia. The purpose of this study was to identify gene targets for PRL8A2 action within the uteroplacental compartment. Affymetrix DNA microarray analysis was performed for RNA samples from WT and Prl8a2 null tissues. Validation of the DNA microarray was performed using quantitative RT-PCR. Nine genes were confirmed with decreased expression in Prl8a2 null tissues (e.g., Klk7, Rimklb, Arhgef6, Calm4, Sprr2h, Prl4a1, Ccl27, Lipg, and Htra3). These include potential decidual, endothelial and trophoblast cell targets positively regulated by PRL8A2. A significant upregulation of Derl3, Herpud1, Creld2, Hsp90b1, Ddit3 and Hspa5 was identified in Prl8a2 null tissues, reflecting an increased endoplasmic reticulum (ER) stress response. ER stress genes were prominently expressed in the uterine decidua. We propose that PRL8A2 is a mediator of progesterone-dependent modulation of intrauterine responses to physiological stressors.
Collapse
Affiliation(s)
- S M Khorshed Alam
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Toshihiro Konno
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Michael J Soares
- Department of Pathology and Laboratory MedicineInstitute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
45
|
Martin E, Ray PD, Smeester L, Grace MR, Boggess K, Fry RC. Epigenetics and Preeclampsia: Defining Functional Epimutations in the Preeclamptic Placenta Related to the TGF-β Pathway. PLoS One 2015; 10:e0141294. [PMID: 26510177 PMCID: PMC4624949 DOI: 10.1371/journal.pone.0141294] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022] Open
Abstract
Preeclampsia is a potentially fatal pregnancy disorder affecting millions of women around the globe. Dysregulation in gene and protein expression within key biological pathways controlling angiogenesis has been implicated in the development of preeclampsia. Altered CpG methylation, a type of epimutation, may underlie this pathway dysregulation. In the present study, placental tissue from preeclamptic cases and normotensive controls was analyzed for genome-wide differential CpG methylation and concomitant changes in gene expression. A set of 123 genes, representing 19.9% of all genes with altered CpG methylation, was associated with functional changes in transcript levels. Underscoring the complex relationships between CpG methylation and gene expression, here hypermethylation was never associated with gene silencing, nor was hypomethylation always associated with gene activation. Moreover, the genomic region of the CpG mark was important in predicting the relationship between CpG methylation and gene expression. The 123 genes were enriched for their involvement in the transforming growth factor beta (TGF-β) signaling pathway, a known regulator of placental trophoblast invasion and migration. This is the first study to identify CpG hypomethylation as an activator of TGF-β-associated gene expression in the preeclamptic placenta. The results suggest functional epimutations are associated with preeclampsia disease status and the identified genes may represent novel biomarkers of disease.
Collapse
Affiliation(s)
- Elizabeth Martin
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Paul D. Ray
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Matthew R. Grace
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kim Boggess
- Department of Obstetrics & Gynecology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Rebecca C. Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, 135 Dauer Drive, CB 7431, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
46
|
Sanders AP, Burris HH, Just AC, Motta V, Amarasiriwardena C, Svensson K, Oken E, Solano-Gonzalez M, Mercado-Garcia A, Pantic I, Schwartz J, Tellez-Rojo MM, Baccarelli AA, Wright RO. Altered miRNA expression in the cervix during pregnancy associated with lead and mercury exposure. Epigenomics 2015; 7:885-96. [PMID: 26418635 DOI: 10.2217/epi.15.54] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM Toxic metals including lead and mercury are associated with adverse pregnancy outcomes. This study aimed to assess the association between miRNA expression in the cervix during pregnancy with lead and mercury levels. MATERIALS & METHODS We obtained cervical swabs from pregnant women (n = 60) and quantified cervical miRNA expression. Women's blood lead, bone lead and toenail mercury levels were analyzed. We performed linear regression to examine the association between metal levels and expression of 74 miRNAs adjusting for covariates. RESULTS Seventeen miRNAs were negatively associated with toenail mercury levels, and tibial bone lead levels were associated with decreased expression of miR-575 and miR-4286. CONCLUSION The findings highlight miRNAs in the human cervix as novel responders to maternal chemical exposure during pregnancy.
Collapse
Affiliation(s)
- Alison P Sanders
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Heather H Burris
- Department of Neonatology, Beth Israel Deaconess Medical Center & Division of Newborn Medicine, Boston Children's Hospital & Harvard Medical School, 330 Brookline Ave, RO 318, Boston, MA 02215, USA
| | - Allan C Just
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Valeria Motta
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Department of Clinical Sciences & Community Health University of Milan - Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chitra Amarasiriwardena
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Katherine Svensson
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emily Oken
- Department of Population Medicine, Harvard Medical School & Harvard Pilgrim Health Care Institute, Boston, MA 02215, USA
| | - Maritsa Solano-Gonzalez
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Adriana Mercado-Garcia
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Ivan Pantic
- Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Division of Research in Public Health, National Institute of Perinatology, Mexico City, Mexico
| | - Joel Schwartz
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Martha M Tellez-Rojo
- Center for Nutrition & Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Andrea A Baccarelli
- Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA 02115, USA.,Laboratory of Environmental Epigenetics, Exposure Epidemiology & Risk Program, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Robert O Wright
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
47
|
Badawy AAB. Tryptophan metabolism, disposition and utilization in pregnancy. Biosci Rep 2015; 35:e00261. [PMID: 26381576 PMCID: PMC4626867 DOI: 10.1042/bsr20150197] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 08/27/2015] [Accepted: 09/16/2015] [Indexed: 12/26/2022] Open
Abstract
Tryptophan (Trp) requirements in pregnancy are several-fold: (1) the need for increased protein synthesis by mother and for fetal growth and development; (2) serotonin (5-HT) for signalling pathways; (3) kynurenic acid (KA) for neuronal protection; (4) quinolinic acid (QA) for NAD(+) synthesis (5) other kynurenines (Ks) for suppressing fetal rejection. These goals could not be achieved if maternal plasma [Trp] is depleted. Although plasma total (free + albumin-bound) Trp is decreased in pregnancy, free Trp is elevated. The above requirements are best expressed in terms of a Trp utilization concept. Briefly, Trp is utilized as follows: (1) In early and mid-pregnancy, emphasis is on increased maternal Trp availability to meet the demand for protein synthesis and fetal development, most probably mediated by maternal liver Trp 2,3-dioxygenase (TDO) inhibition by progesterone and oestrogens. (2) In mid- and late pregnancy, Trp availability is maintained and enhanced by the release of albumin-bound Trp by albumin depletion and non-esterified fatty acid (NEFA) elevation, leading to increased flux of Trp down the K pathway to elevate immunosuppressive Ks. An excessive release of free Trp could undermine pregnancy by abolishing T-cell suppression by Ks. Detailed assessment of parameters of Trp metabolism and disposition and related measures (free and total Trp, albumin, NEFA, K and its metabolites and pro- and anti-inflammatory cytokines in maternal blood and, where appropriate, placental and fetal material) in normal and abnormal pregnancies may establish missing gaps in our knowledge of the Trp status in pregnancy and help identify appropriate intervention strategies.
Collapse
Affiliation(s)
- Abdulla A-B Badawy
- School of Health Sciences, Cardiff Metropolitan University, Western Avenue, Cardiff CF5 2YB, Wales, U.K.
| |
Collapse
|
48
|
Watanabe Y, Yoshida M, Yamanishi K, Yamamoto H, Okuzaki D, Nojima H, Yasunaga T, Okamura H, Matsunaga H, Yamanishi H. Genetic analysis of genes causing hypertension and stroke in spontaneously hypertensive rats: Gene expression profiles in the kidneys. Int J Mol Med 2015; 36:712-24. [PMID: 26165378 PMCID: PMC4533772 DOI: 10.3892/ijmm.2015.2281] [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: 03/07/2015] [Accepted: 06/25/2015] [Indexed: 12/15/2022] Open
Abstract
Spontaneously hypertensive rats (SHRs) and stroke-prone SHRs (SHRSP) are frequently used as models not only of essential hypertension and stroke, but also of attention-deficit hyperactivity disorder (ADHD). Normotensive Wistar-Kyoto (WKY) rats are normally used as controls in these studies. In the present study, we aimed to identify the genes causing hypertension and stroke, as well as the genes involved in ADHD using these rats. We previously analyzed gene expression profiles in the adrenal glands and brain. Since the kidneys can directly influence the functions of the cardiovascular, endocrine and sympathetic nervous systems, gene expression profiles in the kidneys of the 3 rat strains were examined using genome-wide microarray technology when the rats were 3 and 6 weeks old, a period in which rats are considered to be in a pre-hypertensive state. Gene expression profiles were compared between the SHRs and WKY rats and also between the SHRSP and SHRs. A total of 232 unique genes showing more than a 4-fold increase or less than a 4-fold decrease in expression were isolated as SHR- and SHRSP-specific genes. Candidate genes were then selected using two different web tools: the 1st tool was the Database for Annotation, Visualization and Integrated Discovery (DAVID), which was used to search for significantly enriched genes and categorized them using Gene Ontology (GO) terms, and the 2nd was Ingenuity Pathway Analysis (IPA), which was used to search for interactions among SHR- and also SHRSP‑specific genes. The analyses of SHR-specific genes using IPA revealed that B-cell CLL/lymphoma 6 (Bcl6) and SRY (sex determining region Y)-box 2 (Sox2) were possible candidate genes responsible for causing hypertension in SHRs. Similar analyses of SHRSP-specific genes revealed that angiotensinogen (Agt), angiotensin II receptor-associated protein (Agtrap) and apolipoprotein H (Apoh) were possible candidate genes responsible for triggering strokes. Since our results revealed that SHRSP-specific genes isolated from the kidneys of rats at 6 weeks of age, included 6 genes related to Huntington's disease, we discussed the genetic association between ADHD and Huntington's disease.
Collapse
Affiliation(s)
- Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Momoko Yoshida
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| | - Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hideyuki Yamamoto
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Daisuke Okuzaki
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hiroshi Nojima
- DNA-Chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Teruo Yasunaga
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Haruki Okamura
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiromichi Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata, Osaka 573-0122, Japan
| |
Collapse
|
49
|
The Impact of Maternal-Fetal Genetic Conflict Situations on the Pathogenesis of Preeclampsia. Biochem Genet 2015; 53:223-34. [DOI: 10.1007/s10528-015-9684-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 06/18/2015] [Indexed: 10/23/2022]
|
50
|
Eidem HR, Ackerman WE, McGary KL, Abbot P, Rokas A. Gestational tissue transcriptomics in term and preterm human pregnancies: a systematic review and meta-analysis. BMC Med Genomics 2015; 8:27. [PMID: 26044726 PMCID: PMC4456776 DOI: 10.1186/s12920-015-0099-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/12/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preterm birth (PTB), or birth before 37 weeks of gestation, is the leading cause of newborn death worldwide. PTB is a critical area of scientific study not only due to its worldwide toll on human lives and economies, but also due to our limited understanding of its pathogenesis and, therefore, its prevention. This systematic review and meta-analysis synthesizes the landscape of PTB transcriptomics research to further our understanding of the genes and pathways involved in PTB subtypes. METHODS We evaluated published genome-wide pregnancy studies across gestational tissues and pathologies, including those that focus on PTB, by performing a targeted PubMed MeSH search and systematically reviewing all relevant studies. RESULTS Our search yielded 2,361 studies on gestational tissues including placenta, decidua, myometrium, maternal blood, cervix, fetal membranes (chorion and amnion), umbilical cord, fetal blood, and basal plate. Selecting only those original research studies that measured transcription on a genome-wide scale and reported lists of expressed genetic elements identified 93 gene expression, 21 microRNA, and 20 methylation studies. Although 30 % of all PTB cases are due to medical indications, 76 % of the preterm studies focused on them. In contrast, only 18 % of the preterm studies focused on spontaneous onset of labor, which is responsible for 45 % of all PTB cases. Furthermore, only 23 of the 10,993 unique genetic elements reported to be transcriptionally active were recovered 10 or more times in these 134 studies. Meta-analysis of the 93 gene expression studies across 9 distinct gestational tissues and 29 clinical phenotypes showed limited overlap of genes identified as differentially expressed across studies. CONCLUSIONS Overall, profiles of differentially expressed genes were highly heterogeneous both between as well as within clinical subtypes and tissues as well as between studies of the same clinical subtype and tissue. These results suggest that large gaps still exist in the transcriptomic study of specific clinical subtypes as well in the generation of the transcriptional profile of well-studied clinical subtypes; understanding the complex landscape of prematurity will require large-scale, systematic genome-wide analyses of human gestational tissues on both understudied and well-studied subtypes alike.
Collapse
Affiliation(s)
- Haley R Eidem
- Department of Biological Sciences, Vanderbilt University, VU Station B #35-1634, Nashville, TN, 37235, USA.
| | - William E Ackerman
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH, 43210, USA.
| | - Kriston L McGary
- Department of Biological Sciences, Vanderbilt University, VU Station B #35-1634, Nashville, TN, 37235, USA.
| | - Patrick Abbot
- Department of Biological Sciences, Vanderbilt University, VU Station B #35-1634, Nashville, TN, 37235, USA.
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, VU Station B #35-1634, Nashville, TN, 37235, USA.
| |
Collapse
|