1
|
Elovitz MA, Gee EPS, Delaney-Busch N, Moe AB, Reddy M, Khodursky A, La J, Abbas I, Mekaru K, Collins H, Siddiqui F, Nolan R, Boelig RC, Kiefer DG, Simmons PM, Saade GR, Saad A, Carter EB, McElrath TF, Quake SR, DePristo MA, Haverty C, Lee M, Namsaraev E, Berghella V, Collier ARY, Frolova AI, Park-Hwang E, Pacheco LD, Sutton EF, Jain M, Rood K, Grobman WA, Biggio JR, Gyamfi-Bannerman C, Jeyabalan A, Rasmussen M. Molecular subtyping of hypertensive disorders of pregnancy. Nat Commun 2025; 16:2948. [PMID: 40199872 PMCID: PMC11978969 DOI: 10.1038/s41467-025-58157-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Hypertensive disorders of pregnancy (HDP), including preeclampsia, affect 1 in 6 pregnancies, are major contributors to maternal morbidity and mortality, yet lack precision medicine strategies. Analyzing transcriptomic data from a prospectively-collected diverse cohort (n = 9102), this study reveals distinct RNA subtypes in maternal blood, reclassifying clinical HDP phenotypes like early/late-onset preeclampsia. The placental gene PAPPA2 strongly predicts the most severe forms of preeclampsia in individuals without pre-existing high risk factors, months before symptoms, and its overexpression correlates with earlier delivery in a dose-dependent manner. Further, molecular subtypes characterized by immune genes are upregulated in less severe forms of HDP. These results reclassify HDP clinical phenotypes into two distinct molecular subtypes, placental-associated or immune-associated. Validation performance for placental-associated HDP yields an AUC of 0.88 in the advanced maternal age population without pre-existing high risk factors. Molecular subtypes create new opportunities to apply precision-based medicine in maternal health.
Collapse
Affiliation(s)
- Michal A Elovitz
- Mirvie Inc., South San Francisco, CA, USA
- Nuttall Women's Health, New York, NY, USA
| | | | | | | | | | | | - Johnny La
- Mirvie Inc., South San Francisco, CA, USA
| | - Ilma Abbas
- Mirvie Inc., South San Francisco, CA, USA
| | - Kay Mekaru
- Mirvie Inc., South San Francisco, CA, USA
| | | | | | - Rory Nolan
- Mirvie Inc., South San Francisco, CA, USA
| | - Rupsa C Boelig
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | - Ebony B Carter
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas F McElrath
- Mirvie Inc., South San Francisco, CA, USA
- Brigham Women's Hospital, Boston, MA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | | | | | | | | | - Vincenzo Berghella
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ai-Ris Y Collier
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | - Kara Rood
- The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | |
Collapse
|
2
|
Hirst JE, Boniface JJ, Le DP, Polpitiya AD, Fox AC, Vu TTK, Dang TT, Fleischer TC, Bui NTH, Hickok DE, Kearney PE, Thwaites G, Kennedy SH, Kestelyn E, Le TQ. Validating the ratio of insulin like growth factor binding protein 4 to sex hormone binding globulin as a prognostic predictor of preterm birth in Viet Nam: a case-cohort study. J Matern Fetal Neonatal Med 2024; 37:2333923. [PMID: 38584143 DOI: 10.1080/14767058.2024.2333923] [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: 08/22/2023] [Accepted: 03/13/2024] [Indexed: 04/09/2024]
Abstract
OBJECTIVE To validate a serum biomarker developed in the USA for preterm birth (PTB) risk stratification in Viet Nam. METHODS Women with singleton pregnancies (n = 5000) were recruited between 19+0-23+6 weeks' gestation at Tu Du Hospital, Ho Chi Minh City. Maternal serum was collected from 19+0-22+6 weeks' gestation and participants followed to neonatal discharge. Relative insulin-like growth factor binding protein 4 (IGFBP4) and sex hormone binding globulin (SHBG) abundances were measured by mass spectrometry and their ratio compared between PTB cases and term controls. Discrimination (area under the receiver operating characteristic curve, AUC) and calibration for PTB <37 and <34 weeks' gestation were tested, with model tuning using clinical factors. Measured outcomes included all PTBs (any birth ≤37 weeks' gestation) and spontaneous PTBs (birth ≤37 weeks' gestation with clinical signs of initiation of parturition). RESULTS Complete data were available for 4984 (99.7%) individuals. The cohort PTB rate was 6.7% (n = 335). We observed an inverse association between the IGFBP4/SHBG ratio and gestational age at birth (p = 0.017; AUC 0.60 [95% CI, 0.53-0.68]). Including previous PTB (for multiparous women) or prior miscarriage (for primiparous women) improved performance (AUC 0.65 and 0.70, respectively, for PTB <37 and <34 weeks' gestation). Optimal performance (AUC 0.74) was seen within 19-20 weeks' gestation, for BMI >21 kg/m2 and age 20-35 years. CONCLUSION We have validated a novel serum biomarker for PTB risk stratification in a very different setting to the original study. Further research is required to determine appropriate ratio thresholds based on the prevalence of risk factors and the availability of resources and preventative therapies.
Collapse
Affiliation(s)
- Jane E Hirst
- Department of Global Women's Health, The George Institute for Global Health, Imperial College London, London, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | | | - Dung Puhong Le
- Department of Obstetrics and Gynaecology, Tu Du Hospital, Ho Chi Minh City, Viet Nam
| | | | - Angela C Fox
- Sera Prognostics, Inc, Salt Lake City, Utah, USA
| | - Thi Thai Kim Vu
- Clinical Trials Unit, Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | - Thuan Trong Dang
- Clinical Trials Unit, Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
| | | | - Nhu Thi Hong Bui
- Department of Obstetrics and Gynaecology, Tu Du Hospital, Ho Chi Minh City, Viet Nam
| | | | | | - Guy Thwaites
- Clinical Trials Unit, Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen H Kennedy
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, UK
| | - Evelyne Kestelyn
- Clinical Trials Unit, Oxford University Clinical Research Unit, Ho Chi Minh City, Viet Nam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thanh Quang Le
- Department of Obstetrics and Gynaecology, Tu Du Hospital, Ho Chi Minh City, Viet Nam
| |
Collapse
|
3
|
Deng F, Lei J, Qiu J, Zhao C, Wang X, Li M, Sun M, Zhang M, Gao Q. DNA methylation landscape in pregnancy-induced hypertension: progress and challenges. Reprod Biol Endocrinol 2024; 22:77. [PMID: 38978060 PMCID: PMC11229300 DOI: 10.1186/s12958-024-01248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
Gestational hypertension (PIH), especially pre-eclampsia (PE), is a common complication of pregnancy. This condition poses significant risks to the health of both the mother and the fetus. Emerging evidence suggests that epigenetic modifications, particularly DNA methylation, may play a role in initiating the earliest pathophysiology of PIH. This article describes the relationship between DNA methylation and placental trophoblast function, genes associated with the placental microenvironment, the placental vascular system, and maternal blood and vascular function, abnormalities of umbilical cord blood and vascular function in the onset and progression of PIH, as well as changes in DNA methylation in the progeny of PIH, in terms of maternal, fetal, and offspring. We also explore the latest research on DNA methylation-based early detection, diagnosis and potential therapeutic strategies for PIH. This will enable the field of DNA methylation research to continue to enhance our understanding of the epigenetic regulation of PIH genes and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Fengying Deng
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Jiahui Lei
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Junlan Qiu
- Department of Oncology and Hematology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, 215153, P.R. China
| | - Chenxuan Zhao
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Xietong Wang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Min Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Miao Sun
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China.
| | - Meihua Zhang
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| | - Qinqin Gao
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China.
| |
Collapse
|
4
|
Gyarmati G, Shroff UN, Riquier-Brison A, Desposito D, Ju W, Stocker SD, Izuhara A, Deepak S, Becerra Calderon A, Burford JL, Kadoya H, Moon JY, Chen Y, Rinschen MM, Ahmadi N, Lau L, Biemesderfer D, James AW, Minichiello L, Zlokovic BV, Gill IS, Kretzler M, Peti-Peterdi J. Neuronally differentiated macula densa cells regulate tissue remodeling and regeneration in the kidney. J Clin Invest 2024; 134:e174558. [PMID: 38598837 PMCID: PMC11142747 DOI: 10.1172/jci174558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 04/09/2024] [Indexed: 04/12/2024] Open
Abstract
Tissue regeneration is limited in several organs, including the kidney, contributing to the high prevalence of kidney disease globally. However, evolutionary and physiological adaptive responses and the presence of renal progenitor cells suggest an existing remodeling capacity. This study uncovered endogenous tissue remodeling mechanisms in the kidney that were activated by the loss of body fluid and salt and regulated by a unique niche of a minority renal cell type called the macula densa (MD). Here, we identified neuronal differentiation features of MD cells that sense the local and systemic environment and secrete angiogenic, growth, and extracellular matrix remodeling factors, cytokines and chemokines, and control resident progenitor cells. Serial intravital imaging, MD nerve growth factor receptor and Wnt mouse models, and transcriptome analysis revealed cellular and molecular mechanisms of these MD functions. Human and therapeutic translation studies illustrated the clinical potential of MD factors, including CCN1, as a urinary biomarker and therapeutic target in chronic kidney disease. The concept that a neuronally differentiated key sensory and regulatory cell type responding to organ-specific physiological inputs controls local progenitors to remodel or repair tissues may be applicable to other organs and diverse tissue-regenerative therapeutic strategies.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Urvi Nikhil Shroff
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Anne Riquier-Brison
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Dorinne Desposito
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Audrey Izuhara
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Sachin Deepak
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Alejandra Becerra Calderon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - James L. Burford
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hiroyuki Kadoya
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Ju-Young Moon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California, USA
| | - Markus M. Rinschen
- Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nariman Ahmadi
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Lester Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel Biemesderfer
- Section of Nephrology and Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Inderbir S. Gill
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - János Peti-Peterdi
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
5
|
Camacho de Gutiérrez AR, Calisici O, Wrenzycki C, Gutiérrez-Añez JC, Hoeflich C, Hoeflich A, Bajcsy ÁC, Schmicke M. Effect of IGFBP-4 during In Vitro Maturation on Developmental Competence of Bovine Cumulus Oocyte Complexes. Animals (Basel) 2024; 14:673. [PMID: 38473060 DOI: 10.3390/ani14050673] [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: 01/11/2024] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Insulin-like growth factors (IGFs) are essential for oocyte maturation. Their bioavailability is regulated by their respective binding proteins (IGFBPs) and proteases. IGFBP-4 blocks the biological effects of IGFs. High IGFBP-4 expression has been associated with follicle atresia. We hypothesized that IGFBP-4 affects oocyte developmental competence during maturation. Therefore, the aim of this study was to examine the effect of IGFBP-4 on the developmental rate of bovine cumulus-oocyte complexes (COCs) during in vitro embryo production. Abattoir-derived COCs were matured with rbIGFBP-4 (2000, 540, and 54 ng/mL) compared to a control. Cumulus expansion, oocyte maturation, cleavage, blastocyst, and hatching rates were evaluated. Furthermore, blastocyst gene expression of SOCS2, STAT3, SLC2A1, SLCA3, BAX, and POU5F1 transcripts were quantified using RT-qPCR. No statistical differences were detected among the groups for cumulus expansion, maturation, cleavage, blastocyst rates, or all gene transcripts analyzed. However, at day 8 and 9, the number of total hatching and successfully hatched blastocysts was lower in 2000 ng/mL rbIGFBP-4 compared to the control (day 8: total hatching: 17.1 ± 0.21 vs. 31.2 ± 0.11%, p = 0.02 and hatched blastocyst 6.7 ± 0.31 vs. 21.5 ± 0.14%, p = 0.004; day 9 total hatching 36.4 ± 0.18 vs. 57.7 ± 0.10%, p = 0.009 and hatched blastocyst 18.2 ± 0.21 vs. 38.1 ± 0.11%, p = 0.004). We concluded that high concentrations of rbIGFBP-4 might negatively affect the subsequent ability of the embryo to hatch and possibly compromise further elongation.
Collapse
Affiliation(s)
| | - Oguz Calisici
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany
| | - Christine Wrenzycki
- Clinic for Veterinary Obstetrics, Gynecology and Andrology of Large and Small Animals, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Juan Carlos Gutiérrez-Añez
- Medical-Surgical Department, College of Veterinary Medicine, University of Zulia, Maracaibo 4001, Venezuela
| | | | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Árpád Csaba Bajcsy
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany
| | - Marion Schmicke
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, 30173 Hannover, Germany
| |
Collapse
|
6
|
Lapehn S, Houghtaling S, Ahuna K, Kadam L, MacDonald JW, Bammler TK, LeWinn KZ, Myatt L, Sathyanarayana S, Paquette AG. Mono(2-ethylhexyl) phthalate induces transcriptomic changes in placental cells based on concentration, fetal sex, and trophoblast cell type. Arch Toxicol 2023; 97:831-847. [PMID: 36695872 PMCID: PMC9968694 DOI: 10.1007/s00204-023-03444-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
Phthalates are ubiquitous plasticizer chemicals found in consumer products. Exposure to phthalates during pregnancy has been associated with adverse pregnancy and birth outcomes and differences in placental gene expression in human studies. The objective of this research was to evaluate global changes in placental gene expression via RNA sequencing in two placental cell models following exposure to the phthalate metabolite mono(2-ethylhexyl) phthalate (MEHP). HTR-8/SVneo and primary syncytiotrophoblast cells were exposed to three concentrations (1, 90, 180 µM) of MEHP for 24 h with DMSO (0.1%) as a vehicle control. mRNA and lncRNAs were quantified using paired-end RNA sequencing, followed by identification of differentially expressed genes (DEGs), significant KEGG pathways, and enriched transcription factors (TFs). MEHP caused gene expression changes across all concentrations for HTR-8/SVneo and primary syncytiotrophoblast cells. Sex-stratified analysis of primary cells identified different patterns of sensitivity in response to MEHP dose by sex, with male placentas being more responsive to MEHP exposure. Pathway analysis identified 11 KEGG pathways significantly associated with at least one concentration in both cell types. Four ligand-inducible nuclear hormone TFs (PPARG, PPARD, ESR1, AR) were enriched in at least three treatment groups. Overall, we demonstrated that MEHP differentially affects placental gene expression based on concentration, fetal sex, and trophoblast cell type. This study confirms prior studies, as enrichment of nuclear hormone receptor TFs were concordant with previously published mechanisms of phthalate disruption, and generates new hypotheses, as we identified many pathways and genes not previously linked to phthalate exposure.
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, 1900 9th Ave, Jack R. MacDonald Building, Seattle, WA 98101 USA
| | - Scott Houghtaling
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, 1900 9th Ave, Jack R. MacDonald Building, Seattle, WA 98101 USA
| | - Kylia Ahuna
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239 USA
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239 USA
| | - James W. MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195 USA
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195 USA
| | - Kaja Z. LeWinn
- Department of Psychiatry, University of California-San Francisco, San Francisco, CA 94143 USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239 USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington, Seattle, WA 98195 USA
- Center for Child Health, Behavior and Development, Seattle Children’s Research Institute, Seattle, WA 98101 USA
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, 1900 9th Ave, Jack R. MacDonald Building, Seattle, WA 98101 USA
- Department of Pediatrics, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
7
|
Li A, Zhang L, Liu Q, Fang Z, Sun Y, Li S, Peng Y, Zhang M, Wang X. Proteomic analysis of amniotic fluid to identify potential targets predicting preterm delivery. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2023; 1871:140879. [PMID: 36396099 DOI: 10.1016/j.bbapap.2022.140879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Preterm delivery is a common complication of pregnancy which leads to significant neonatal mortality and morbidity. Identifying predictive markers linked to spontaneous preterm delivery (SPTD) is important for effective treatment and prevention of PTD. To explore potential biomarkers related to SPTD, we performed proteomics analysis in amniotic fluid (AF). In total, we enrolled 30 pregnant women with singleton gestation who underwent clinically indicated amniocentesis at 15-24 weeks of gestation. LC-MS analysis was used to analyze the AF samples of 10 women with SPTD < 34 weeks after cervix cerclage (Preterm group), 10 women with term delivery (TD) ≥ 34 weeks after cervix cerclage (Term group), and 10 women who delivered at term (Normal group). ELISA validation was performed for candidate proteins in a second independent cohort. As a result, we identified 44 differentially expressed proteins (DEPs, P < 0.05) via proteomic analysis, and based on that, 9 primary pathways were also determined in SPTD. Results of the ELISA assay confirmed that the increased concentration of Serpin A1, decreased concentrations of Renin and IGFBP4 were significantly associated with SPTD at ≤34 weeks.
Collapse
Affiliation(s)
- Anna Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Lin Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Qunying Liu
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Zhenya Fang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Yaqiong Sun
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Shuxian Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Yanjie Peng
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China.
| | - Xietong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and child health care hospital of Shandong province, Jinan 250014, Shandong, China; Department of Obstetrics and Gynecology, Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China.
| |
Collapse
|
8
|
Burchard J, Polpitiya AD, Fox AC, Randolph TL, Fleischer TC, Dufford MT, Garite TJ, Critchfield GC, Boniface JJ, Saade GR, Kearney PE. Clinical Validation of a Proteomic Biomarker Threshold for Increased Risk of Spontaneous Preterm Birth and Associated Clinical Outcomes: A Replication Study. J Clin Med 2021; 10:5088. [PMID: 34768605 PMCID: PMC8584743 DOI: 10.3390/jcm10215088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/22/2022] Open
Abstract
Preterm births are the leading cause of neonatal death in the United States. Previously, a spontaneous preterm birth (sPTB) predictor based on the ratio of two proteins, IBP4/SHBG, was validated as a predictor of sPTB in the Proteomic Assessment of Preterm Risk (PAPR) study. In particular, a proteomic biomarker threshold of -1.37, corresponding to a ~two-fold increase or ~15% risk of sPTB, significantly stratified earlier deliveries. Guidelines for molecular tests advise replication in a second independent study. Here we tested whether the significant association between proteomic biomarker scores above the threshold and sPTB, and associated adverse outcomes, was replicated in a second independent study, the Multicenter Assessment of a Spontaneous Preterm Birth Risk Predictor (TREETOP). The threshold significantly stratified subjects in PAPR and TREETOP for sPTB (p = 0.041, p = 0.041, respectively). Application of the threshold in a Kaplan-Meier analysis demonstrated significant stratification in each study, respectively, for gestational age at birth (p < 001, p = 0.0016) and rate of hospital discharge for both neonate (p < 0.001, p = 0.005) and mother (p < 0.001, p < 0.001). Above the threshold, severe neonatal morbidity/mortality and mortality alone were 2.2 (p = 0.0083,) and 7.4-fold higher (p = 0.018), respectively, in both studies combined. Thus, higher predictor scores were associated with multiple adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Julja Burchard
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Ashoka D. Polpitiya
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Angela C. Fox
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Todd L. Randolph
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Tracey C. Fleischer
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Max T. Dufford
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Thomas J. Garite
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - Gregory C. Critchfield
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - J. Jay Boniface
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| | - George R. Saade
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Paul E. Kearney
- Sera Prognostics, Incorporated, Salt Lake City, UT 84109, USA; (J.B.); (A.D.P.); (A.C.F.); (T.L.R.); (M.T.D.); (T.J.G.); (G.C.C.); (J.J.B.); (P.E.K.)
| |
Collapse
|
9
|
Grabner M, Burchard J, Nguyen C, Chung H, Gangan N, Boniface JJ, Zupancic JAF, Stanek E. Cost-Effectiveness of a Proteomic Test for Preterm Birth Prediction. CLINICOECONOMICS AND OUTCOMES RESEARCH 2021; 13:809-820. [PMID: 34548799 PMCID: PMC8449551 DOI: 10.2147/ceor.s325094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Preterm birth (PTB) carries increased risk of short- and long-term health problems as well as higher healthcare costs. Current strategies using clinically accepted maternal risk factors (prior PTB, short cervix) can only identify a minority of singleton PTBs. Objective We modeled the cost-effectiveness of a risk-screening-and-treat strategy versus usual care for commercially insured pregnant US women without clinically accepted PTB risk factors. The risk-screening-and-treat strategy included use of a novel PTB prognostic blood test (PreTRM®) in the 19th–20th week of pregnancy, followed by treatment with a combined regimen of multi-component high-intensity-case-management and pharmacologic interventions for the remainder of the pregnancy for women assessed as higher-risk by the test, and usual care in women without higher risk. Methods We built a cost-effectiveness model using a combined decision-tree/Markov approach and a US payer perspective. We modeled 1-week cycles of pregnancy from week 19 to birth (preterm or term) and assessed costs throughout the pregnancy, and further to 12-months post-delivery in mothers and 30-months in infants. PTB rates and costs were based on >40,000 mothers and infants from the HealthCore Integrated Research Database® with birth events in 2016. Estimates of test performance, treatment effectiveness, and other model inputs were derived from published literature. Results In the base case, the risk-screening-and-treat strategy dominated usual care with an estimated 870 fewer PTBs (20% reduction) and $54 million less in total cost ($863 net savings per pregnant woman). Reductions were projected for neonatal intensive care admissions (10%), overall length-of-stay (7%), and births <32 weeks (33%). Treatment effectiveness had the strongest influence on cost-effectiveness estimates. The risk-screening-and-treat strategy remained dominant in the majority of probabilistic sensitivity analysis simulations and model scenarios. Conclusion Use of a novel prognostic test during pregnancy to identify women at risk of PTB combined with evidence-based treatment is estimated to reduce total costs while preventing PTBs and their consequences.
Collapse
Affiliation(s)
| | - Julja Burchard
- Research and Development, Sera Prognostics, Salt Lake City, UT, USA
| | - Chi Nguyen
- Health Economics and Outcomes Research, HealthCore, Inc., Wilmington, DE, USA
| | - Haechung Chung
- Research Operations, HealthCore, Inc., Wilmington, DE, USA
| | - Nilesh Gangan
- Health Economics and Outcomes Research, HealthCore, Inc., Wilmington, DE, USA
| | - J Jay Boniface
- Research and Development, Sera Prognostics, Salt Lake City, UT, USA
| | - John A F Zupancic
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Eric Stanek
- Scientific Affairs, HealthCore, Inc., Wilmington, DE, USA
| |
Collapse
|
10
|
Hong S, Lee JE, Kim YM, Park Y, Choi JW, Park KH. Identifying potential biomarkers related to pre-term delivery by proteomic analysis of amniotic fluid. Sci Rep 2020; 10:19648. [PMID: 33184413 PMCID: PMC7665029 DOI: 10.1038/s41598-020-76748-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 11/02/2020] [Indexed: 12/31/2022] Open
Abstract
We sought to identify biomarkers in the amniotic fluid (AF) and specific signaling pathways related to spontaneous preterm delivery (SPTD, < 34 weeks) in women with preterm labor (PTL) without intra-uterine infection/inflammation (IUI). This was a retrospective cohort study of a total of 139 PTL women with singleton gestation (24 + 0 to 32 + 6 weeks) who underwent amniocentesis and who displayed no evidence of IUI. A nested case-control was conducted using pooled AF samples (n = 20) analyzed via label-free liquid chromatography-tandem mass spectrometry. In the total cohort, an ELISA validation study was performed for seven candidate proteins of interest. Proteomic analysis identified 77 differentially expressed proteins (DEPs, P < 0.05) in the AF from SPTD cases compared to term delivery controls. ELISA validation confirmed that women who had an SPTD before 34 weeks had significantly independently lower levels of VEGFR-1 and higher levels of lipocalin-2 and the Fc fragment of IgG binding protein in the AF. Five principle pathways associated with the 77 DEPs were identified, including glycolysis, gluconeogenesis, and iron homeostasis. The proteomic analysis data of AFs from women with PTL identified several novel biomarkers and specific protein pathways related to SPTD in the absence of IUI.
Collapse
Affiliation(s)
- Subeen Hong
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Yu Mi Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 463-707, Korea
| | - Yehyon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 463-707, Korea
| | - Ji-Woong Choi
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 463-707, Korea.
| |
Collapse
|
11
|
Ji Y, Zhang W, Yang J, Li C. MiR-193b inhibits autophagy and apoptosis by targeting IGFBP5 in high glucose-induced trophoblasts. Placenta 2020; 101:185-193. [PMID: 33010605 DOI: 10.1016/j.placenta.2020.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Inhibiting apoptosis of trophoblasts in women with gestational diabetes mellitus (GDM) is expected to guarantee adequate nutrition for the fetus and avoid abortion. MiR-193b is one of the most downregulated miRNAs in GDM patients. However, less is known about the role of miR-193b in autophagy and apoptosis in GDM patients. METHODS We detected the expression of miR-193b in GDM patients. Then, we cultured human trophoblasts (HTR8 cells) with high glucose (HG) to simulate a diabetic environment in vitro, and further explored the effects of miR-193b on apoptosis and autophagy of HG-treated HTR8 cells. RESULTS The expression of miR-193b was significantly downregulated in the peripheral blood of GDM patients compared with healthy controls, and decreased miR-193b caused apparent autophagy and a substantially high apoptosis rate in HG-treated HTR8 cells. These effects were reversed by enhancing miR-193b expression or using the autophagy inhibitor 3-MA. Inhibiting miR-193b induced the pro-autophagic, cytostatic, and pro-apoptotic effects reduced by 3-MA in HTR8 cells upon HG treatment. Moreover, the expression of insulin-like growth factor-binding protein 5 (IGFBP5) was upregulated notably in the peripheral blood of GDM patients, and IGFBP5 appears to represent a direct miR-193b target. Note that silencing IGFBP5 blocked autophagy and apoptosis in HG-treated HTR8 cells, an effect that was diminished by inhibiting miR-193b. CONCLUSION Our data indicate that aberrantly low expression of miR-193b in HG-induced trophoblasts results in massive apoptosis events by upregulating IGFBP5-induced autophagy, which may trigger GDM. Therefore, miR-193b may became a potential target for GDM therapy.
Collapse
Affiliation(s)
- Yanting Ji
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Wenfeng Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Jin Yang
- Department of Obstetrics and Gynecology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Marinello WP, Mohseni ZS, Cunningham SJ, Crute C, Huang R, Zhang JJ, Feng L. Perfluorobutane sulfonate exposure disrupted human placental cytotrophoblast cell proliferation and invasion involving in dysregulating preeclampsia related genes. FASEB J 2020; 34:14182-14199. [PMID: 32901980 DOI: 10.1096/fj.202000716rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
We reported that maternal PFBS, an emerging pollutant, exposure is positively associated with preeclampsia which can result from aberrant trophoblasts invasion and subsequent placental ischemia. In this study, we investigated the effects of PFBS on trophoblasts proliferation/invasion and signaling pathways. We exposed a human trophoblast line, HTR8/SVneo, to PFBS. Cell viability, proliferation, and cell cycle were evaluated by the MTS assay, Ki-67 staining, and flow cytometry, respectively. We assessed cell migration and invasion with live-cell imaging-based migration assay and matrigel invasion assay, respectively. Signaling pathways were examined by Western blot, RNA-seq, and qPCR. PFBS exposure interrupted cell proliferation and invasion in a dose-dependent manner. PFBS (100 μM) did not cause cell death but instead significant cell proliferation without cell cycle disruption. PFBS (10 and 100 μM) decreased cell migration and invasion, while PFBS (0.1 μM) significantly increased cell invasion but not migration. Further, RNA-seq analysis identified dysregulated HIF-1α target genes that are relevant to cell proliferation/invasion and preeclampsia, while Western Blot data showed the activation of HIF-1α, but not Notch, ERK1/2, (PI3K)AKT, and P38 pathways. PBFS exposure altered trophoblast cell proliferation/invasion which might be mediated by preeclampsia-related genes, suggesting a possible association between prenatal PFBS exposure and adverse placentation.
Collapse
Affiliation(s)
- William P Marinello
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Zahra S Mohseni
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Sarah J Cunningham
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Christine Crute
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Jun J Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Markenson GR, Saade GR, Laurent LC, Heyborne KD, Coonrod DV, Schoen CN, Baxter JK, Haas DM, Longo S, Grobman WA, Sullivan SA, Major CA, Wheeler SM, Pereira LM, Su EJ, Boggess KA, Hawk AF, Crockett AH, Fox AC, Polpitiya A, Fleischer TC, Critchfield GC, Burchard J, Boniface JJ, Lam GK. Performance of a proteomic preterm delivery predictor in a large independent prospective cohort. Am J Obstet Gynecol MFM 2020; 2:100140. [PMID: 33345877 DOI: 10.1016/j.ajogmf.2020.100140] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Preterm birth remains a common and devastating complication of pregnancy. There remains a need for effective and accurate screening methods for preterm birth. Using a proteomic approach, we previously discovered and validated (Proteomic Assessment of Preterm Risk study, NCT01371019) a preterm birth predictor comprising a ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin. OBJECTIVE To determine the performance of the ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin to predict both spontaneous and medically indicated very preterm births, in an independent cohort distinct from the one in which it was developed. STUDY DESIGN This was a prospective observational study (Multicenter Assessment of a Spontaneous Preterm Birth Risk Predictor, NCT02787213) at 18 sites in the United States. Women had blood drawn at 170/7 to 216/7 weeks' gestation. For confirmation, we planned to analyze a randomly selected subgroup of women having blood drawn between 191/7 and 206/7 weeks' gestation, with the results of the remaining study participants blinded for future validation studies. Serum from participants was analyzed by mass spectrometry. Neonatal morbidity and mortality were analyzed using a composite score by a method from the PREGNANT trial (NCT00615550, Hassan et al). Scores of 0-3 reflect increasing numbers of morbidities or length of neonatal intensive care unit stay, and 4 represents perinatal mortality. RESULTS A total of 5011 women were enrolled, with 847 included in this planned substudy analysis. There were 9 preterm birth cases at <320/7 weeks' gestation and 838 noncases at ≥320/7 weeks' gestation; 21 of 847 infants had neonatal composite morbidity and mortality index scores of ≥3, and 4 of 21 had a score of 4. The ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin ratio was substantially higher in both preterm births at <320/7 weeks' gestation and there were more severe neonatal outcomes. The ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin ratio was significantly predictive of birth at <320/7 weeks' gestation (area under the receiver operating characteristic curve, 0.71; 95% confidence interval, 0.55-0.87; P=.016). Stratification by body mass index, optimized in the previous validation study (22<body mass index≤37 kg/m2), resulted in an area under the receiver operating characteristic curve of 0.76 (95% confidence interval, 0.59-0.93; P=.023). The ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin ratio predicted neonatal outcomes with respective area under the receiver operating characteristic curve of 0.67 (95% confidence interval, 0.57-0.77; P=.005) and 0.78 (95% confidence interval, 0.63-0.93; P=.026) for neonatal composite morbidity and mortality scores of ≥3 or 4. In addition, the ratio of insulin-like growth factor-binding protein 4 to sex hormone binding globulin significantly stratified neonates with increased length of hospital stay (log rank P=.023). CONCLUSION We confirmed in an independent cohort the ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin ratio as a predictor of very preterm birth, with additional prediction of increased length of neonatal hospital stay and increased severity of adverse neonatal outcomes. Potential uses of the ratio of insulin-like growth factor-binding protein 4 to sex hormone-binding globulin predictor may be to risk stratify patients for implementation of preterm birth preventive strategies and direct patients to appropriate levels of care.
Collapse
Affiliation(s)
- Glenn R Markenson
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Boston Medical Center, Boston, MA
| | - George R Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - Louise C Laurent
- Division of Maternal-Fetal Medicine, Department of Reproductive Sciences, University of California, San Diego, CA
| | - Kent D Heyborne
- Department of Obstetrics and Gynecology, Denver Health and Hospital Authority
| | - Dean V Coonrod
- Department of Obstetrics and Gynecology, Maricopa Integrated Health System
| | - Corina N Schoen
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Massachusetts-Baystate
| | - Jason K Baxter
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Thomas Jefferson University Hospital
| | - David M Haas
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Indiana University
| | - Sherri Longo
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Ochsner Baptist Medical Center, New Orleans, LA
| | - William A Grobman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Scott A Sullivan
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, SC
| | - Carol A Major
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California, Irvine, CA
| | - Sarahn M Wheeler
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University, Durham, NC
| | - Leonardo M Pereira
- Division Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR
| | - Emily J Su
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO
| | - Kim A Boggess
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC
| | | | - Amy H Crockett
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Greenville Health System, Greenville, SC
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Lamale-Smith LM, Gumina DL, Kramer AW, Browne VA, Toledo-Jaldin L, Julian CG, Winn VD, Moore LG. Uteroplacental Ischemia Is Associated with Increased PAPP-A2. Reprod Sci 2020; 27:529-536. [PMID: 31994005 DOI: 10.1007/s43032-019-00050-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/22/2019] [Indexed: 11/29/2022]
Abstract
Residence at high altitude (> 2500 m) has been associated with an increased frequency of preeclampsia. Pappalysin-2 (PAPP-A2) is an insulin-like growth factor binding protein-5 (IGFBP-5) protease that is elevated in preeclampsia, and up-regulated by hypoxia in placental explants. The relationships between PAPP-A2, altitude, and indices of uteroplacental ischemia are unknown. We aimed to evaluate the association of altitude, preeclampsia, and uterine artery flow or vascular resistance with PAPP-A2 levels. PAPP-A2, uterine artery diameter, volumetric blood flow, and pulsatility indices were measured longitudinally in normotensive Andean women residing at low or high altitudes in Bolivia and in a separate Andean high-altitude cohort with or without preeclampsia. PAPP-A2 levels increased with advancing gestation, with the rise tending to be greater at high compared to low altitude, and higher in early-onset preeclamptic compared to normotensive women at high altitude. Uterine artery blood flow was markedly lower and pulsatility index higher in early-onset preeclamptic normotensive women compared to normotensive women. PAPP-A2 was unrelated to uterine artery pulsatility index in normotensive women but positively correlated in the early-onset preeclampsia cases. We concluded that PAPP-A2 is elevated at high altitude and especially in cases of early-onset preeclampsia with Doppler indices of uteroplacental ischemia.
Collapse
Affiliation(s)
- Leah M Lamale-Smith
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, San Diego, CA, USA.
| | - Diane L Gumina
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| | - Anita W Kramer
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| | - Vaughn A Browne
- Department of Emergency Medicine, University of Colorado Denver, Aurora, CO, USA
| | | | - Colleen G Julian
- Department of Medicine, University of Colorado, Denver, Aurora, CO, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Lorna G Moore
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
15
|
The potential role of pregnancy-associated plasma protein-A2 in angiogenesis and development of preeclampsia. Hypertens Res 2019; 42:970-980. [DOI: 10.1038/s41440-019-0224-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/06/2019] [Accepted: 01/15/2019] [Indexed: 12/25/2022]
|
16
|
Starks RR, Biswas A, Jain A, Tuteja G. Combined analysis of dissimilar promoter accessibility and gene expression profiles identifies tissue-specific genes and actively repressed networks. Epigenetics Chromatin 2019; 12:16. [PMID: 30795793 PMCID: PMC6385419 DOI: 10.1186/s13072-019-0260-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The assay for transposase-accessible chromatin (ATAC-seq) is a powerful method to examine chromatin accessibility. While many studies have reported a positive correlation between gene expression and promoter accessibility, few have investigated the genes that deviate from this trend. In this study, we aimed to understand the relationship between gene expression and promoter accessibility in multiple cell types while also identifying gene regulatory networks in the placenta, an understudied organ that is critical for a successful pregnancy. RESULTS We started by assaying the open chromatin landscape in the mid-gestation placenta, when the fetal vasculature has started developing. After incorporating transcriptomic data generated in the placenta at the same time point, we grouped genes based on their expression levels and ATAC-seq promoter coverage. We found that the genes with the strongest correlation (high expression and high coverage) are likely involved in housekeeping functions, whereas tissue-specific genes were highly expressed and had only medium-low coverage. We also predicted that genes with medium-low expression and high promoter coverage were actively repressed. Within this group, we extracted a protein-protein interaction network enriched for neuronal functions, likely preventing the cells from adopting a neuronal fate. We further confirmed that a repressive histone mark is bound to the promoters of genes in this network. Finally, we ran our pipeline using ATAC-seq and RNA-seq data generated in ten additional cell types. We again found that genes with the strongest correlation are enriched for housekeeping functions and that genes with medium-low promoter coverage and high expression are more likely to be tissue-specific. These results demonstrate that only two data types, both of which require relatively low starting material to generate and are becoming more commonly available, can be integrated to understand multiple aspects of gene regulation. CONCLUSIONS Within the placenta, we identified an active placenta-specific gene network as well as a repressed neuronal network. Beyond the placenta, we demonstrate that ATAC-seq data and RNA-seq data can be integrated to identify tissue-specific genes and actively repressed gene networks in multiple cell types.
Collapse
Affiliation(s)
- Rebekah R. Starks
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011 USA
- Bioinformatics and Computational Biology, Iowa State University, Ames, IA 50011 USA
| | - Anilisa Biswas
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011 USA
- Molecular, Cellular, and Developmental Biology, Iowa State University, Ames, IA 50011 USA
| | - Ashish Jain
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011 USA
- Bioinformatics and Computational Biology, Iowa State University, Ames, IA 50011 USA
| | - Geetu Tuteja
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011 USA
- Bioinformatics and Computational Biology, Iowa State University, Ames, IA 50011 USA
- Molecular, Cellular, and Developmental Biology, Iowa State University, Ames, IA 50011 USA
| |
Collapse
|
17
|
Ma M, Zhou QJ, Xiong Y, Li B, Li XT. Preeclampsia is associated with hypermethylation of IGF-1 promoter mediated by DNMT1. Am J Transl Res 2018; 10:16-39. [PMID: 29422991 PMCID: PMC5801344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 11/13/2017] [Indexed: 06/08/2023]
Abstract
Previous studies have demonstrated a dynamic epigenetic regulation of genes expression in placenta trophoblasts and a dynamic imbalance of DNA methylation and hydroxymethylation. Reduced IGF-1 has been observed in preeclampsia. This study was to investigate the interactive roles between IGF-1 and the global DNA methylation/hydroxymethylation, and the status of DNA methylation/hydroxymethylation and associated enzymes such as DNMTs and TETs in peeeclamptic placentas and hypoxic trophoblasts. It was found that IGF-1 was decreased in preeclamptic placentas and hypoxic trophoblasts when compared to the control group using immunohistochemisty, western blot, qRT-PCR and ELISA. Pyrophosphate sequencing showed IGF-1 promoter was significantly hypermethylated in preeclamptic placentas, which was responsible for reduced IGF-1 expression. Preeclamptic placentas and hypoxic trophoblasts were hypermethylated and hypohydroxymethylated accompanied by remarkably higher 5mC, DNMT1 and DNMT3b, and lower DNMT3a, 5hmC, TET1, TET2 and TET3 detected by immunohistochemisty, western blot, qRT-PCR and ELISA. Pearson's correlation confirmed a statistically significant negative correlation between IGF-1 and DNMT1. Furthermore, both treatment with 5-Aza-dc and DNMT1-siRNA significantly increased the expression of IGF-1 in HTR8 cells, indicating the potential mechanism of DNMT1-mediated DNA methylation in IGF-1 regulation. However, IGF-1 didn't change DNA methylation or hydroxymethylation. These findings suggest that preeclampsia is associated with hypermethylation of IGF-1 promoter mediated by DNMT1 and provide new insights into the diagnosis and treatment of preeclampsia.
Collapse
Affiliation(s)
- Min Ma
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan UniversityShanghai 200011, China
- Department of Obstetrics and Gynecology, Medical College of Yangzhou UniversityYangzhou 225000, Jiangsu, China
- Department of Obstetrics, Affiliated Hospital of Yangzhou UniversityYangzhou 225000, Jiangsu, China
| | - Qiong-Jie Zhou
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan UniversityShanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
- Women’s Health and Perinatology Research Group, Department of Clinical Medicine, UiT-The Arctic University of NorwayTromso, Norway
| | - Yu Xiong
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan UniversityShanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
| | - Bin Li
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan UniversityShanghai 200011, China
| | - Xiao-Tian Li
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan UniversityShanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, China
- Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College of Fudan UniversityShanghai 200032, China
- Institute of Biomedical Sciences, Shanghai Medical College of Eudan UniversityShanghai 200032, China
- Shanghai Key Laboratory of Birth DefectsShanghai 200032, China
| |
Collapse
|
18
|
Jia Y, Li T, Huang X, Xu X, Zhou X, Jia L, Zhu J, Xie D, Wang K, Zhou Q, Jin L, Zhang J, Duan T. Dysregulated DNA Methyltransferase 3A Upregulates IGFBP5 to Suppress Trophoblast Cell Migration and Invasion in Preeclampsia. Hypertension 2017; 69:356-366. [DOI: 10.1161/hypertensionaha.116.08483] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 10/10/2016] [Accepted: 12/03/2016] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a unique multiple system disorder during human pregnancy, which affects ≈5% to 8% of pregnancies. Its risks and complications have become the major causes of maternal and fetal morbidity and mortality. Although abnormal placentation to which DNA methylation dysregulation is always linked is speculated to be one of the reasons causing preeclampsia, the underlying mechanisms still remain elusive to date. Here we revealed that aberrant DNA methyltransferase 3A (DNMT3A) plays a critical role in preeclampsia. Our results show that the expression and localization of DNMT3A are dysregulated in preeclamptic placenta. Moreover, knockdown of DNMT3A obviously inhibits trophoblast cell migration and invasion. Mechanistically, IGFBP5 (insulin-like growth factor–binding protein 5), known as a suppressor, is upregulated by decreased DNMT3A because of promoter hypomethylation. Importantly, IGFBP5 downregulation can rescue the defects caused by DNMT3A knockdown, thereby, consolidating the significance of IGFBP5 in the downstream of DNMT3A in trophoblast. Furthermore, we detected low promoter methylation and high protein expression of IGFBP5 in the clinical samples of preeclamptic placenta. Collectively, our study suggests that dysregulation of DNMT3A and IGFBP5 is relevant to preeclampsia. Thus, we propose that DNMT3A and IGFBP5 can serve as potential markers and targets for the clinical diagnosis and therapy of preeclampsia.
Collapse
Affiliation(s)
- Yuanhui Jia
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Ting Li
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Xiaojie Huang
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Xianghong Xu
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Xinyao Zhou
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Linyan Jia
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Jingping Zhu
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Dandan Xie
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Kai Wang
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Qian Zhou
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Liping Jin
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Jiqin Zhang
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| | - Tao Duan
- From the Clinical and Translational Research Center (Y.J., X.H., X.X., X.Z., L. Jia, J. Zhu, D.X., K.W., Q.Z., L. Jin) and Department of Obstetrics (T.L., T.D.), Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, China; and Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J. Zhang)
| |
Collapse
|
19
|
Maymon R, Trahtenherts A, Svirsky R, Melcer Y, Madar-Shapiro L, Klog E, Meiri H, Cuckle H. Developing a new algorithm for first and second trimester preeclampsia screening in twin pregnancies. Hypertens Pregnancy 2016; 36:108-115. [DOI: 10.1080/10641955.2016.1242605] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ron Maymon
- Institute of Ultrasound, Department of Obstetrics and Gynecology, Assaf Harofeh Medical Centre, Affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
- Tel-Aviv University, Tel-Aviv, Israel
| | | | - Ran Svirsky
- Institute of Ultrasound, Department of Obstetrics and Gynecology, Assaf Harofeh Medical Centre, Affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
- Tel-Aviv University, Tel-Aviv, Israel
| | - Yaakov Melcer
- Institute of Ultrasound, Department of Obstetrics and Gynecology, Assaf Harofeh Medical Centre, Affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
- Tel-Aviv University, Tel-Aviv, Israel
| | | | - Esther Klog
- Institute of Ultrasound, Department of Obstetrics and Gynecology, Assaf Harofeh Medical Centre, Affiliated with the Sackler School of Medicine, Tel-Aviv, Israel
- Tel-Aviv University, Tel-Aviv, Israel
| | | | | |
Collapse
|
20
|
Development and validation of a spontaneous preterm delivery predictor in asymptomatic women. Am J Obstet Gynecol 2016; 214:633.e1-633.e24. [PMID: 26874297 DOI: 10.1016/j.ajog.2016.02.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/25/2016] [Accepted: 02/04/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Preterm delivery remains the leading cause of perinatal mortality. Risk factors and biomarkers have traditionally failed to identify the majority of preterm deliveries. OBJECTIVE To develop and validate a mass spectrometry-based serum test to predict spontaneous preterm delivery in asymptomatic pregnant women. STUDY DESIGN A total of 5501 pregnant women were enrolled between 17(0/7) and 28(6/7) weeks gestational age in the prospective Proteomic Assessment of Preterm Risk study at 11 sites in the United States between 2011 and 2013. Maternal blood was collected at enrollment and outcomes collected following delivery. Maternal serum was processed by a proteomic workflow, and proteins were quantified by multiple reaction monitoring mass spectrometry. The discovery and verification process identified 2 serum proteins, insulin-like growth factor-binding protein 4 (IBP4) and sex hormone-binding globulin (SHBG), as predictors of spontaneous preterm delivery. We evaluated a predictor using the log ratio of the measures of IBP4 and SHBG (IBP4/SHBG) in a clinical validation study to classify spontaneous preterm delivery cases (<37(0/7) weeks gestational age) in a nested case-control cohort different from subjects used in discovery and verification. Strict blinding and independent statistical analyses were employed. RESULTS The predictor had an area under the receiver operating characteristic curve value of 0.75 and sensitivity and specificity of 0.75 and 0.74, respectively. The IBP4/SHBG predictor at this sensitivity and specificity had an odds ratio of 5.04 for spontaneous preterm delivery. Accuracy of the IBP4/SHBG predictor increased using earlier case-vs-control gestational age cutoffs (eg, <35(0/7) vs ≥35(0/7) weeks gestational age). Importantly, higher-risk subjects defined by the IBP4/SHBG predictor score generally gave birth earlier than lower-risk subjects. CONCLUSION A serum-based molecular predictor identifies asymptomatic pregnant women at risk of spontaneous preterm delivery, which may provide utility in identifying women at risk at an early stage of pregnancy to allow for clinical intervention. This early detection would guide enhanced levels of care and accelerate development of clinical strategies to prevent preterm delivery.
Collapse
|
21
|
Costa MA. The endocrine function of human placenta: an overview. Reprod Biomed Online 2015; 32:14-43. [PMID: 26615903 DOI: 10.1016/j.rbmo.2015.10.005] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/13/2015] [Accepted: 10/14/2015] [Indexed: 12/25/2022]
Abstract
During pregnancy, several tightly coordinated and regulated processes take place to enable proper fetal development and gestational success. The formation and development of the placenta is one of these critical pregnancy events. This organ plays essential roles during gestation, including fetal nourishment, support and protection, gas exchange and production of several hormones and other mediators. Placental hormones are mainly secreted by the syncytiotrophoblast, in a highly and tightly regulated way. These hormones are important for pregnancy establishment and maintenance, exerting autocrine and paracrine effects that regulate decidualization, placental development, angiogenesis, endometrial receptivity, embryo implantation, immunotolerance and fetal development. In addition, because they are released into maternal circulation, the profile of their blood levels throughout pregnancy has been the target of intense research towards finding potential robust and reliable biomarkers to predict and diagnose pregnancy-associated complications. In fact, altered levels of these hormones have been associated with some pathologies, such as chromosomal anomalies or pre-eclampsia. This review proposes to revise and update the main pregnancy-related hormones, addressing their major characteristics, molecular targets, function throughout pregnancy, regulators of their expression and their potential clinical interest.
Collapse
Affiliation(s)
- Mariana A Costa
- Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
22
|
Christians JK, Beristain AG. ADAM12 and PAPP-A: Candidate regulators of trophoblast invasion and first trimester markers of healthy trophoblasts. Cell Adh Migr 2015; 10:147-53. [PMID: 26417939 DOI: 10.1080/19336918.2015.1083668] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Proper placental development and function is crucial for a healthy pregnancy, and there has been substantial research to identify markers of placental dysfunction for the early detection of pregnancy complications. Low first-trimester levels of a disintegrin and metalloproteinase 12 (ADAM12) and pregnancy-associated plasma protein-A (PAPP-A) have been consistently associated with the subsequent development of preeclampsia and fetal growth restriction. These molecules are both metalloproteinases secreted by the placenta that cleave insulin-like growth factor binding proteins (IGFBPs), although ADAM12 also has numerous other substrates. Recent work has identified ADAM12, and particularly its shorter variant, ADAM12S, as a regulator of the migration and invasion of trophoblasts into the lining of the uterus, a critical step in normal placental development. While the mechanisms underlying this regulation are not yet clear, they may involve the liberation of heparin-binding EGF-like growth factor (HB-EGF) and/or IGFs from IGFBPs. In contrast, there has been relatively little functional work examining PAPP-A or the IGFBP substrates of ADAM12 and PAPP-A. Understanding the functions of these markers and the mechanisms underlying their association with disease could improve screening strategies and enable the development of new therapeutic interventions.
Collapse
Affiliation(s)
| | - Alexander G Beristain
- b Department of Obstetrics and Gynecology , The University of British Columbia , Vancouver , Canada.,c The Child and Family Research Institute , Vancouver , Canada
| |
Collapse
|
23
|
Changes in Functional Activity of JEG-3 Trophoblast Cell Line in the Presence of Factors Secreted by Placenta. Arch Med Res 2015; 46:245-56. [PMID: 26003221 DOI: 10.1016/j.arcmed.2015.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 05/12/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIMS Cells in the maternal-fetal interface secrete cytokines that regulate proliferation, migration, and trophoblast invasion during the first trimester of pregnancy and the limitation of these processes during the third trimester. The aim of the study was to evaluate the influence of factors secreted by human placenta during the first and third trimester of pregnancy on cytokine receptor expression and proliferative and migratory activity of JEG-3 trophoblast cells. METHODS The research was conducted using the explant conditioned media of placentas obtained from healthy women with elective termination of pregnancy at 9-11 weeks and placentas of women whose pregnancy progressed without complications at 38-39 weeks. Assessment of surface molecule expression was performed using FACS Canto II flow cytometer (BD, USA). The proliferative activity of JEG-3 trophoblast cells was evaluated by dyeing with crystal violet vital dye. The migration activity of JEG-3 was evaluated using 24-well insert plates with polycarbonate inserts (pore size 8 microns). RESULTS Expression of CD116, CD118, CD119, IFNγ-R2, CD120b, CD183, CD192, CD295, EGFR, and TGFβ-R2 on JEG-3 was higher when the cells were incubated in the presence of the third trimester placental factors in comparison with the first trimester placental factors. Factors secreted by the placenta during the third trimester of pregnancy had more pronounced stimulatory effect on the proliferation and migration of trophoblast in comparison with baseline levels and with the effect of the first trimester placental factors. CONCLUSIONS The findings suggest that the behavior of trophoblasts in vitro might not be representative of in vivo behavior in the absence of additional local factors that influence the trophoblast in vivo.
Collapse
|