1
|
Hu L, Li J, Hu L, Zhang M, Wang Y, Wang M, Xu Y. Relationship between gastrointestinal disturbances, blood lipid levels, inflammatory markers, and preterm birth. J OBSTET GYNAECOL 2025; 45:2475065. [PMID: 40254961 DOI: 10.1080/01443615.2025.2475065] [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: 12/11/2024] [Accepted: 02/27/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The challenging incidence of preterm birth, the underlying causes of preterm birth remain unclear. This study determined the relationship between disturbed gastrointestinal symptoms, inflammatory markers, blood lipid levels, and preterm birth. METHOD One hundred and twenty pregnant women with preterm labour were compared to 120 pregnant women with full-term deliveries. All subjects underwent lactose breath and serologic testing. The correlation between small intestinal bacterial overgrowth (SIBO)-positivity, gastrointestinal symptoms, inflammatory factors, and blood lipid metabolism and preterm birth was analysed using the Spearman method. RESULTS SIBO, hydrogen, and methane levels were significantly higher in the preterm birth (PTB) group than the full-term birth (FTB) group at different time points (P < 0.05); Levels of high-sensitivity C-reactive protein (hs-CRP) (3.95[2.70-5.77] vs. 2.47[1.45-3.83]), Interleukin (IL)-10 (3.05[2.27-4.33] vs. 2.09[1.04-3.47]), IL-6 (5.23[3.95-8.50] vs. 2.98[2.22-4.44]), tumour necrosis factor -alpha (TNF-α) (3.23[1.55-4.90] vs. 1.76[0.98-3.10]), total cholesterol (TC) (5.52[4.97-5.95] vs. 5.24[4.73-5.85]), and triglycerides (TG) (2.58[2.04-3.53] vs. 2.24[1.59-3.05]) were significantly higher in the PTB group than the FTB group (P < 0.05). Abdominal distension (2.67[1.67-3.00] vs. 2.33[1.67-2.67]) and constipation (2.00[1.33-2.00] vs. 1.67[1.33-2.00]) scores were also markedly higher in the PTB group than the FTB group (P < 0.05). Preterm birth was positively correlated with SIBO, TC, and TG levels. Additionally, SIBO was positively correlated with hs-CRP, IL-10, IL-6, and TNF-α levels, abdominal distension, and constipation (P < 0.05). Logistic regression analysis found the close association between positive SIBO, biochemistry indicators and preterm birth. CONCLUSION Gastrointestinal disturbances, hyperlipidaemia and SIBO-positivity are more likely to occur among pregnant women with preterm labour. Further research with a large sample size in multi-centers is needed to validate the results.
Collapse
Affiliation(s)
- Lulu Hu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Li
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Hu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Miao Zhang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yixin Wang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengqi Wang
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yajuan Xu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Guo Z, Wang K, Huang X, Li K, Ouyang G, Yang X, Tan J, Shi H, Luo L, Zhang M, Han B, Zhai X, Deng J, Beatson R, Wu Y, Yang F, Yang X, Tang J. Genome-wide nucleosome footprints of plasma cfDNA predict preterm birth: A case-control study. PLoS Med 2025; 22:e1004571. [PMID: 40233080 PMCID: PMC11999135 DOI: 10.1371/journal.pmed.1004571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 03/03/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Preterm birth (PTB) occurs in approximately 11% of all births worldwide, resulting in significant morbidity and mortality for both mothers and their offspring. Identifying pregnancies at risk of preterm birth during early pregnancy may help improve interventions and reduce its incidence. Plasma cell-free DNA (cfDNA), derived from placenta and other maternal tissues, serves as a dynamic indicator of biological processes and pathological changes in pregnancy. These properties establish cfDNA as a valuable biomarker for investigating pregnancy complications, including PTB. METHODS AND FINDINGS To date, there are few methods available for PTB prediction that have been developed with large sample sizes, high-throughput screening, and validated in independent cohorts. To address this gap, we established a large-scale, multi-center case-control study involving 2,590 pregnancies (2,072 full-term and 518 preterm) from three independent hospitals to develop a spontaneous preterm birth classifier. We performed whole-genome sequencing on cfDNA, focusing on promoter profiling (read depth of promoter regions spanning from -1 to +1 kb around transcriptional start sites). Using four machine learning models and two feature selection algorithms, we developed classifiers for predicting preterm birth. Among these, the classifier based on the support vector machine model, named PTerm (Promoter profiling classifier for preterm prediction), exhibited the highest area under the curve (AUC) value of 0.878 (0.852-0.904) following leave-one-out cross-validation. Additionally, PTerm exhibited strong performance in three independent validation cohorts, achieving an overall AUC of 0.849 (0.831-0.866). CONCLUSIONS In summary, PTerm demonstrated high accuracy in predicting preterm birth. Additionally, it can be utilized with current non-invasive prenatal test data without changing its procedures or increasing detection cost, making it easily adaptable for preclinical tests.
Collapse
Affiliation(s)
- Zhiwei Guo
- Department of Obstetrics and Gynaecology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ke Wang
- Department of Obstetrics and Gynaecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang Huang
- Prenatal Diagnosis Center, Foshan Women and Children Hospital, Foshan, Guangdong, China
| | - Kun Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Guojun Ouyang
- Guangzhou Darui Biotechnology Co, Ltd., Guangzhou, China
| | - Xu Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jiayu Tan
- Emergency Department, Foshan Women and Children Hospital, Foshan, Guangdong, China
| | - Haihong Shi
- Medical Genetics Center, Jiangmen Maternity and Child Health Care Hospital, Jiangmen, Guangdong, China
| | - Liangping Luo
- School of Medicine, Jinan University, Guangzhou, China
| | - Min Zhang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Bowei Han
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiangming Zhai
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer Research, Comprehensive Cancer Centre, Kings College London, London, United Kingdom
| | - Richard Beatson
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, London, United Kingdom
| | - Yingsong Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Fang Yang
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuexi Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jia Tang
- NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
- Medical Genetics Center, Jiangmen Maternity and Child Health Care Hospital, Jiangmen, Guangdong, China
- School of Medicine, Jinan University, Guangzhou, China
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
3
|
Feyaerts D, Diop M, Galaz J, Einhaus JF, Arck PC, Diemert A, Winn VD, Parast M, Gyamfi-Bannerman C, Prins JR, Gomez-Lopez N, Stelzer IA. The single-cell immune profile throughout gestation and its potential value for identifying women at risk for spontaneous preterm birth. Eur J Obstet Gynecol Reprod Biol X 2025; 25:100371. [PMID: 40052005 PMCID: PMC11883378 DOI: 10.1016/j.eurox.2025.100371] [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: 07/30/2024] [Revised: 11/23/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Precisely timed immune adaptations, observed in the maternal circulation, underpin the notion of an immune clock of human pregnancy that supports its successful progression and completion at delivery. This immune clock is divided into three immunological phases, with the first phase starting at the time of conception and implantation, shifting into the second phase that supports homeostasis and tolerance throughout pregnancy, and culminating in the last phase of labor and parturition. Disruptions of this immune clock are reported in pregnancy complications such as spontaneous preterm birth. However, our understanding of the immune clock preceding spontaneous preterm birth remains scattered. In this review, we describe the chronology of maternal immune cell adaptations during healthy pregnancies and highlight its disruption in spontaneous preterm birth. With a focus on single-cell cytometric, proteomic and transcriptomic approaches, we review recent studies of term and spontaneous preterm pregnancies and discuss the need for future prospective studies aimed at tracking pregnancies longitudinally on a multi-omic scale. Such studies will be critical in determining whether spontaneous preterm pregnancies progress at an accelerated pace or follow a preterm-intrinsic pattern when compared to those delivered at term.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Maïgane Diop
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Galaz
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jakob F. Einhaus
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Petra C. Arck
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mana Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Cynthia Gyamfi-Bannerman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jelmer R. Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nardhy Gomez-Lopez
- Departments of Obstetrics and Gynecology & Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Ina A. Stelzer
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
4
|
Chaemsaithong P, Romero R, Pongchaikul P, Warintaksa P, Mongkolsuk P, Bhuwapathanapun M, Kotchompoo K, Nimsamer P, Kruasuwan W, Amnuaykiatlert O, Vivithanaporn P, Meyyazhagan A, Awonuga A, Settacomkul R, Singhsnaeh A, Laolerd W, Santanirand P, Thaipisuttikul I, Wongsurawat T, Jenjaroenpun P. The rapid diagnosis of intraamniotic infection with nanopore sequencing. Am J Obstet Gynecol 2025:S0002-9378(25)00091-2. [PMID: 39952543 DOI: 10.1016/j.ajog.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Intraamniotic infection (defined as intraamniotic inflammation with microorganisms) is an important cause of the preterm labor syndrome. Methods for the detection of microorganisms in amniotic fluid are culture and/or polymerase chain reaction assay. However, both methods take time, and the results are rarely available for clinical decision-making. Nanopore sequencing technology offers real-time, long-read sequencing that can produce rapid results. OBJECTIVE To determine 1) the diagnostic performance of the 16S rDNA nanopore sequencing method for the identification of microorganisms in patients with intraamniotic inflammation and 2) the relationship between microbial burden and the intensity of the amniotic fluid inflammatory response. STUDY DESIGN We performed a prospective cohort study that included singleton pregnancies presenting with symptoms of preterm labor with intact membranes or of preterm prelabor rupture of the membranes. Amniotic fluid samples were obtained for the evaluation of bacteria in the amniotic cavity using cultivation and polymerase chain reaction-based 16S Sanger sequencing methods. Participants were classified into 4 groups according to the results of an amniotic fluid culture, 16S Sanger sequencing, and an amniotic fluid interleukin 6 concentration: 1) no intraamniotic infection and intraamniotic inflammation (interleukin 6 <2.6 ng/mL, and no microorganisms in the amniotic cavity, as determined by culture or 16S Sanger sequencing); 2) microbial invasion of the amniotic cavity without intraamniotic inflammation, defined by the presence of bacteria detected by culture or 16S Sanger sequencing; 3) sterile intraamniotic inflammation (interleukin 6 ≥2.6 ng/mL without microbial invasion of the amniotic cavity); and 4) intraamniotic infection (interkeukin 6 ≥2.6 ng/mL with microbial invasion of the amniotic cavity). Patients who underwent a mid-trimester amniocentesis, had no intraamniotic infection or intraamniotic inflammation, and delivered at term represented the control group. 16S rDNA nanopore sequencing was performed and the diagnostic indices for the identification of intraamniotic infection were determined. Bioinformatic analysis was carried out to identify microorganisms, and a read count of at least 100 or a read count exceeding that of the background species from the control group, along with a relative abundance of no less than 1%, was used. RESULTS 1) The 16S nanopore sequencing had a sensitivity of 88.9% (8/9), specificity of 95.4% (41/43), positive predictive value of 80.0% (8/10), negative predictive value of 97.6% (41/42), positive likelihood ratio of 19.1 (95% confidence interval, 4.8-75.4), negative likelihood ratio of 0.1 (95% confidence interval, 0.02-0.7), and an accuracy of 94.2% (49/52) for the identification of intraamniotic infection (prevalence, 17% [9/52]); 2) the microbial load determined by the 16S nanopore sequencing had a strong positive correlation with the intensity of an intraamniotic inflammatory response (amniotic fluid interleukin 6 concentration; Spearman's correlation 0.9; P=.002); and 3) a subgroup of patients with intraamniotic inflammation did not have bacteria determined by culture, Sanger sequencing, or nanopore 16S, thus confirming the existence of sterile intraamniotic inflammation. CONCLUSION The 16S nanopore sequencing has high diagnostic indices, predictive values, likelihood ratios, and accuracy in the diagnosis of intraamniotic infection.
Collapse
Affiliation(s)
- Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom, Thailand; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Paninee Mongkolsuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand
| | - Maolee Bhuwapathanapun
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kanyaphat Kotchompoo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pattaraporn Nimsamer
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Long-Read Lab (Si-LoL), Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Worarat Kruasuwan
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Long-Read Lab (Si-LoL), Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Orrakanya Amnuaykiatlert
- Mahidol University International Demonstration School, Mahidol University, Nakhon Pathom, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand
| | - Arun Meyyazhagan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Life Sciences, Christ University, Bengaluru, India
| | - Awoniyi Awonuga
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Rapeewan Settacomkul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Samut Prakarn, Thailand
| | - Arunee Singhsnaeh
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Warawut Laolerd
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pitak Santanirand
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Iyarit Thaipisuttikul
- Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thidathip Wongsurawat
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Long-Read Lab (Si-LoL), Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Piroon Jenjaroenpun
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Long-Read Lab (Si-LoL), Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR.
| |
Collapse
|
5
|
Valensin C, Côté EJM, Pereira-Carvalho D, Gardner RA, Nishku G, Giles CL, Gill C, Brockbank A, Story L, Shennan AH, Suff N, Gibbons DL, Tribe RM. INSIGHT-2: mechanistic studies into pregnancy complications and their impact on maternal and child health-study protocol. Reprod Health 2024; 21:177. [PMID: 39609862 PMCID: PMC11605920 DOI: 10.1186/s12978-024-01911-0] [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: 10/09/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Pregnancy and early childhood cohorts provide a framework for investigating the complex interplay between early-life exposures and health outcomes, thereby informing prevention strategies and interventions to improve maternal and child health. In this paper, we outline the objectives, methodologies and expected contributions of INSIGHT-2, a comprehensive cohort study dedicated to advancing our understanding of pregnancy and pregnancy complications towards improving the health and well-being of mothers and their offspring. METHODS Over the course of 5 years, the study aims to establish a diverse cohort of 1700 pregnant women and to follow up their children up to 2 years of age. Recruitment targets participants with healthy pregnancies, preexisting conditions, and/or risk factors for pregnancy complications or later child health problems. Clinical and lifestyle data and a range of biological samples will be collected, providing a comprehensive resource for biomarker investigations and cross-sectional analyses. It is anticipated that the cohort will continue beyond this initial 5-year plan. DISCUSSION By gathering a wide range of biological samples and using diverse analytical techniques, this study supports broad participation, potential replication and collaboration across various sites. The extensive collection of longitudinal data and samples not only facilitates current investigations but also establishes a biobank for future research. The exploration of pre-pregnancy and pregnancy factors that may contribute to disease processes and impact fetal well-being and future health will provide a comprehensive picture of disease mechanisms in both mothers and children, facilitating the identification of biomarkers for the prediction, diagnosis, and management of pregnancy complications. Additionally, our diverse population allows for the capture of various pregnancy complications and outcomes, enhancing external validity and addressing health disparities. This comprehensive design ultimately aims to improve maternal and child health outcomes by providing a valuable longitudinal study of the relationships among the in utero environment, pregnancy management, and long-term maternal and child health, ensuring that findings are relevant and beneficial to a broader population.
Collapse
Affiliation(s)
- Carlotta Valensin
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK.
| | - Emilie J M Côté
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Daniela Pereira-Carvalho
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Peter Gorer Department of Immunobiology, Guy's Hospital, King's College London, London, UK
| | - Rachael A Gardner
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Glen Nishku
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Caitlin L Giles
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Carolyn Gill
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Anna Brockbank
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Lisa Story
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Department of Perinatal Imaging, St Thomas' Hospital, King's College London, London, UK
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Andrew H Shennan
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
- Guy's and St Thomas' National Health Service Foundation Trust, London, UK
| | - Natalie Suff
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| | - Deena L Gibbons
- Peter Gorer Department of Immunobiology, Guy's Hospital, King's College London, London, UK
| | - Rachel M Tribe
- Department of Women and Children's Health, St Thomas' Hospital, King's College London, London, UK
| |
Collapse
|
6
|
Bernier E, Couture C, Borchers A, Brien ME, Graham CH, Girard S. Circulating Immune Cells from Early- and Late-onset Pre-eclampsia Displays Distinct Profiles with Differential Impact on Endothelial Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1292-1304. [PMID: 39302114 PMCID: PMC11491498 DOI: 10.4049/jimmunol.2400196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Pre-eclampsia (PE) affects 5-8% of pregnancies and has detrimental effects on maternal-fetal health. PE is characterized by de novo hypertension after 20 wk of gestation and end-organ damage. Systemic inflammatory imbalance has been associated with PE, but its contribution to the pathology is poorly understood. Our objective was to investigate maternal systemic immune changes in early-onset PE (EOPE) and late-onset PE (LOPE) versus uncomplicated pregnancies (control [CTRL]), and their contribution to endothelial activation, hallmark of hypertension. Blood samples were analyzed by flow cytometry, multiplex assay, intracellular cytokine staining, and single-cell RNA sequencing. We performed cocultures between circulating immune cells and HUVECs to assess endothelial activation. We found that EOPE had decreased regulatory T cells (4.64±0.33, p < 0.05) and monocytes (33.92±3.08, p < 0.01), whereas LOPE had decreased regulatory T cells (4.60±0.30, p < 0.05) and Th2 cells (7.50±0.62, p < 0.01) versus CTRL. Compared to CTRL, elevated cytokines/chemokines, and growth factors were observed in LOPE, whereas EOPE primarily showed decreased levels. Using intracellular cytokine staining, we observed more monocytes producing IL-12, TNF-α, and IL-1β (all p < 0.05) in LOPE versus CTRL. At the transcriptomic level, we found differentially expressed genes between EOPE and CTRL, predominantly related to upregulation of immune activation pathways. Lastly, EOPE PBMCs induced heightened endothelial activation in vitro observed by increased ICAM-1 and ET-1 (p < 0.05), whereas LOPE PBMCs required LPS stimulation. Although significant proteomic changes are observed in the LOPE group, the EOPE displayed changes mostly at the transcriptomic levels and could induce endothelial activation in vitro.
Collapse
Affiliation(s)
- Elsa Bernier
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Camille Couture
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Anna Borchers
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN
- Department of Immunology, Mayo Clinic, Rochester, MN
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Sylvie Girard
- Department of Immunology, Mayo Clinic, Rochester, MN
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN
| |
Collapse
|
7
|
Wang X, Liu R, Chen Z, Zhang R, Mei Y, Miao X, Bai X, Dong Y. Combining Transcriptomics and Proteomics to Screen Candidate Genes Related to Bovine Birth Weight. Animals (Basel) 2024; 14:2751. [PMID: 39335340 PMCID: PMC11429316 DOI: 10.3390/ani14182751] [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: 07/18/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The placenta is a vital organ in bovine reproduction, crucial for blood supply, nutrient transport, and embryonic development. It plays an essential role in the intrauterine growth of calves. However, the molecular mechanisms governing placental function in calves remain inadequately understood. METHODS We established transcriptome and proteome databases for low-birth-weight (LB) and high-birth-weight (HB) calf placentae, identifying key genes and proteins associated with birth weight through bioinformatics analyses that included functional enrichment and protein-protein interactions (PPIs). Both mRNA and protein levels were validated. RESULTS A total of 1494 differentially expressed genes (DEGs) and 294 differentially expressed proteins (DEPs) were identified when comparing the LB group to the HB group. Furthermore, we identified 53 genes and proteins exhibiting significant co-expression across both transcriptomic and proteomic datasets; among these, 40 were co-upregulated, 8 co-downregulated, while 5 displayed upregulation at the protein level despite downregulation at the mRNA level. Functional enrichment analyses (GO and KEGG) and protein-protein interaction (PPI) analysis indicate that, at the transcriptional level, the primary factor contributing to differences in calf birth weight is that the placenta of the high-birth-weight (HB) group provides more nutrients to the fetus, characterized by enhanced nutrient transport (SLC2A1 and SLC2A11), energy metabolism (ACSL1, MICALL2, PAG2, COL14A1, and ELOVL5), and lipid synthesis (ELOVL5 and ELOVL7). In contrast, the placenta of the low-birth-weight (LB) group prioritizes cell proliferation (PAK1 and ITGA3) and angiogenesis. At the protein level, while the placentae from the HB group exhibit efficient energy production and lipid synthesis, they also demonstrate reduced immunity to various diseases such as systemic lupus erythematosus and bacterial dysentery. Conversely, the LB group placentae excel in regulating critical biological processes, including cell migration, proliferation, differentiation, apoptosis, and signal transduction; they also display higher disease immunity markers (COL6A1, TNC CD36, CD81, Igh-1a, and IGHG) compared to those of the HB group placentae. Co-expression analysis further suggests that increases in calf birth weight can be attributed to both high-efficiency energy production and lipid synthesis within the HB group placentae (ELOVL5, ELOVL7, and ACSL1), alongside cholesterol biosynthesis and metabolic pathways involving CYP11A1 and CYP17A1. CONCLUSION We propose that ELOVL5, ELOVL7, ACSL1, CYP11A1, and CYP17A1 serve as potential protein biomarkers for regulating calf birth weight through the modulation of the fatty acid metabolism, lipid synthesis, and cholesterol levels.
Collapse
Affiliation(s)
- Xiuyuan Wang
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Ruili Liu
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Zhenpeng Chen
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Renzheng Zhang
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Yanfang Mei
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiuping Miao
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Xuejin Bai
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
- Black Cattle Seed Industry Innovation Center, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| | - Yajuan Dong
- Laboratory of Animal Molecular, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
- Black Cattle Seed Industry Innovation Center, Shandong Black Cattle Breeding Engineering Technology Center, College of Animal Science, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
8
|
Couture C, Caron M, St-Onge P, Brien ME, Sinnett D, Dal Soglio D, Girard S. Identification of divergent placental profiles in clinically distinct pregnancy complications revealed by the transcriptome. Placenta 2024; 154:184-192. [PMID: 39042974 DOI: 10.1016/j.placenta.2024.07.008] [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: 04/25/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Pregnancy complications, including preeclampsia (PE), preterm birth (PTB), and intra-uterine growth restriction (IUGR) have individually been associated with inflammation but the combined comparative analysis of their placental profiles at the transcriptomic and histological levels is lacking. METHODS Bulk RNA-sequencing of human placental biopsies from uncomplicated term pregnancies (CTL) and pregnancies complicated with early-onset (EO), and late-onset (LO) PE, as well as PTB and term IUGR were used to characterize individual molecular profiles. We also applied immune-cell-specific cellular deconvolution to address local immune cell compositions and analyzed placental lesions by histology to further characterize these complications. RESULTS Transcriptome analysis revealed that clinically distinct complications differentiated themselves in unique ways compared to CTLs. Only TMEM136 was commonly modulated. Compared to CTLs, we found that PTB and IUGR were the most distinct, with LOPE being the least distinct. PTB and IUGR revealed differently enhanced inflammatory pathways, where PTB had general inflammatory responses and IUGR had immune cell activation. This inflammation was reflected in the histological profile for PTB only, whereas structural lesions were elevated in all complications. Placental lesions additionally had corresponding enhancement in inflammatory and structural biological processes. We observed that having co-complications, particularly for PTB with or without IUGR, impacted placental transcriptomes. Lastly, cellular deconvolution uncovered shared immune features among the complications. DISCUSSION Overall, we provide evidence that these pregnancy complications are not only distinct in their clinical manifestations but also in their placental profiles, which could be leveraged to understand their underlying mechanisms and could offer therapeutic targets.
Collapse
Affiliation(s)
- Camille Couture
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada; Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Maxime Caron
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montreal, Quebec, Canada
| | - Pascal St-Onge
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Daniel Sinnett
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montreal, Montreal, Quebec, Canada
| | - Dorothée Dal Soglio
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, QC, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada; Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Lv W, Xie H, Wu S, Dong J, Jia Y, Ying H. Identification of key metabolism-related genes and pathways in spontaneous preterm birth: combining bioinformatic analysis and machine learning. Front Endocrinol (Lausanne) 2024; 15:1440436. [PMID: 39229380 PMCID: PMC11368757 DOI: 10.3389/fendo.2024.1440436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 09/05/2024] Open
Abstract
Background Spontaneous preterm birth (sPTB) is a global disease that is a leading cause of death in neonates and children younger than 5 years of age. However, the etiology of sPTB remains poorly understood. Recent evidence has shown a strong association between metabolic disorders and sPTB. To determine the metabolic alterations in sPTB patients, we used various bioinformatics methods to analyze the abnormal changes in metabolic pathways in the preterm placenta via existing datasets. Methods In this study, we integrated two datasets (GSE203507 and GSE174415) from the NCBI GEO database for the following analysis. We utilized the "Deseq2" R package and WGCNA for differentially expressed genes (DEGs) analysis; the identified DEGs were subsequently compared with metabolism-related genes. To identify the altered metabolism-related pathways and hub genes in sPTB patients, we performed multiple functional enrichment analysis and applied three machine learning algorithms, LASSO, SVM-RFE, and RF, with the hub genes that were verified by immunohistochemistry. Additionally, we conducted single-sample gene set enrichment analysis to assess immune infiltration in the placenta. Results We identified 228 sPTB-related DEGs that were enriched in pathways such as arachidonic acid and glutathione metabolism. A total of 3 metabolism-related hub genes, namely, ANPEP, CKMT1B, and PLA2G4A, were identified and validated in external datasets and experiments. A nomogram model was developed and evaluated with 3 hub genes; the model could reliably distinguish sPTB patients and term labor patients with an area under the curve (AUC) > 0.75 for both the training and validation sets. Immune infiltration analysis revealed immune dysregulation in sPTB patients. Conclusion Three potential hub genes that influence the occurrence of sPTB through shadow participation in placental metabolism were identified; these results provide a new perspective for the development and targeting of treatments for sPTB.
Collapse
Affiliation(s)
- Wenqi Lv
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Han Xie
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Shengyu Wu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Jiaqi Dong
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
| | - Yuanhui Jia
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| | - Hao Ying
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, China
- Department of Clinical Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai, sChina
| |
Collapse
|
10
|
Habelrih T, Augustin TL, Mauffette-Whyte F, Ferri B, Sawaya K, Côté F, Gallant M, Olson DM, Chemtob S. Inflammatory mechanisms of preterm labor and emerging anti-inflammatory interventions. Cytokine Growth Factor Rev 2024; 78:50-63. [PMID: 39048393 DOI: 10.1016/j.cytogfr.2024.07.007] [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: 07/04/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Preterm birth is a major public health concern, requiring a deeper understanding of its underlying inflammatory mechanisms and to develop effective therapeutic strategies. This review explores the complex interaction between inflammation and preterm labor, highlighting the pivotal role of the dysregulation of inflammation in triggering premature delivery. The immunological environment of pregnancy, characterized by a fragile balance of immune tolerance and resistance, is disrupted in preterm labor, leading to a pathological inflammatory response. Feto-maternal infections, among other pro-inflammatory stimuli, trigger the activation of toll-like receptors and the production of pro-inflammatory mediators, promoting uterine contractility and cervical ripening. Emerging anti-inflammatory therapeutics offer promising approaches for the prevention of preterm birth by targeting key inflammatory pathways. From TLR-4 antagonists to chemokine and interleukin receptor antagonists, these interventions aim to modulate the inflammatory environment and prevent adverse pregnancy outcomes. In conclusion, a comprehensive understanding of the inflammatory mechanisms leading to preterm labor is crucial for the development of targeted interventions in hope of reducing the incidence of preterm birth and improving neonatal health outcomes.
Collapse
Affiliation(s)
- Tiffany Habelrih
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Thalyssa-Lyn Augustin
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Félix Mauffette-Whyte
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Béatrice Ferri
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Kevin Sawaya
- Research Center, CHU Sainte-Justine, Montreal, QC, Canada; Programmes de cycles supérieurs en sciences biomédicales, Faculté de médecine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - Mathilde Gallant
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Research Center, CHU Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
11
|
Feyaerts D, Marić I, Arck PC, Prins JR, Gomez-Lopez N, Gaudillière B, Stelzer IA. Predicting Spontaneous Preterm Birth Using the Immunome. Clin Perinatol 2024; 51:441-459. [PMID: 38705651 DOI: 10.1016/j.clp.2024.02.013] [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] [Indexed: 05/07/2024]
Abstract
Throughout pregnancy, the maternal peripheral circulation contains valuable information reflecting pregnancy progression, detectable as tightly regulated immune dynamics. Local immune processes at the maternal-fetal interface and other reproductive and non-reproductive tissues are likely to be the pacemakers for this peripheral immune "clock." This cellular immune status of pregnancy can be leveraged for the early risk assessment and prediction of spontaneous preterm birth (sPTB). Systems immunology approaches to sPTB subtypes and cross-tissue (local and peripheral) interactions, as well as integration of multiple biological data modalities promise to improve our understanding of preterm birth pathobiology and identify potential clinically actionable biomarkers.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ivana Marić
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Petra C Arck
- Department of Obstetrics and Fetal Medicine and Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251 Hamburg, Germany
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Postbus 30.001, 9700RB, Groningen, The Netherlands
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, 425 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA; Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, 300 Pasteur Drive, Palo Alto, CA 94304, USA
| | - Ina A Stelzer
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
12
|
Huang D, Ran Y, Chen R, He J, Yin N, Qi H. Identification of circRNA Expression Profile and Potential Systemic Immune Imbalance Modulation in Premature Rupture of Membranes. Anal Cell Pathol (Amst) 2024; 2024:6724914. [PMID: 38803428 PMCID: PMC11129912 DOI: 10.1155/2024/6724914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 03/08/2024] [Accepted: 04/10/2024] [Indexed: 05/29/2024] Open
Abstract
Premature rupture of membrane (PROM) refers to the rupture of membranes before the onset of labor which increases the risk of perinatal morbidity and mortality. Recently, circular RNAs (circRNAs) have emerged as promising regulators of diverse diseases. However, the circRNA expression profiles and potential circRNA-miRNA-mRNA regulatory mechanisms in PROM remain enigmatic. In this study, we displayed the expression profiles of circRNAs and mRNAs in plasma and fetal membranes of PROM and normal control (NC) groups based on circRNA microarray, the Gene Expression Omnibus database, and NCBI's Sequence Read Archive. A total of 1,459 differentially expressed circRNAs (DECs) in PROM were identified, with 406 upregulated and 1,053 downregulated. Then, we constructed the circRNA-miRNA-mRNA network in PROM, encompassing 22 circRNA-miRNA pairs and 128 miRNA-mRNA pairs. Based on the analysis of gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and gene set enrichment analysis (GSEA), DECs were implicated in immune-related pathways, with certain alterations persisting even postpartum. Notably, 11 host genes shared by DECs of fetal membrane tissue and prenatal plasma in PROM were significantly implicated in inflammatory processes and extracellular matrix regulation. Our results suggest that structurally stable circRNAs may predispose to PROM by mediating systemic immune imbalances, including peripheral leukocyte disorganization, local immune imbalance at the maternal-fetal interface, and local collagen disruption. This is the first time to decipher a landscape on circRNAs of PROM, reveals the pathogenic cause of PROM from the perspective of circRNA, and opens up a new direction for the diagnosis and treatment of PROM.
Collapse
Affiliation(s)
- Dongni Huang
- Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Yuxin Ran
- Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Ruixin Chen
- Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jie He
- Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| | - Nanlin Yin
- Center for Reproductive Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongbo Qi
- Women and Children's Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Couture C, Brien ME, Rechtzigel J, Ling S, Ledezma-Soto C, Duran Bishop G, Boufaied I, Dal Soglio D, Rey E, McGraw S, Graham CH, Girard S. Predictive biomarkers and initial analysis of maternal immune alterations in postpartum preeclampsia reveal an immune-driven pathology. Front Immunol 2024; 15:1380629. [PMID: 38745664 PMCID: PMC11091301 DOI: 10.3389/fimmu.2024.1380629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/13/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction Postpartum preeclampsia (PPPE) is an under-diagnosed condition, developing within 48 hours to 6 weeks following an uncomplicated pregnancy. The etiology of PPPE is still unknown, leaving patients vulnerable and making the identification and treatment of patients requiring postpartum care an unmet need. We aimed to understand the immune contribution to PPPE at the time of diagnosis, as well as uncover the predictive potential of perinatal biomarkers for the early postnatal identification of high-risk patients. Methods Placentas were collected at delivery from uncomplicated pregnancies (CTL) and PPPE patients for immunohistochemistry analysis. In this initial study, blood samples in PPPE patients were collected at the time of PPPE diagnosis (48h-25 days postpartum; mean 7.4 days) and compared to CTL blood samples taken 24h after delivery. Single-cell transcriptomics, flow cytometry, intracellular cytokine staining, and the circulating levels of inflammatory mediators were evaluated in the blood. Results Placental CD163+ cells and 1st trimester blood pressures can be valuable non-invasive and predictive biomarkers of PPPE with strong clinical application prospects. Furthermore, changes in immune cell populations, as well as cytokine production by CD14+, CD4+, and CD8+ cells, suggested a dampened response with an exhausted phenotype including decreased IL1β, IL12, and IFNγ as well as elevated IL10. Discussion Understanding maternal immune changes at the time of diagnosis and prenatally within the placenta in our sizable cohort will serve as groundwork for pre-clinical and clinical research, as well as guiding clinical practice for example in the development of immune-targeted therapies, and early postnatal identification of patients who would benefit from more thorough follow-ups and risk education in the weeks following an uncomplicated pregnancy.
Collapse
Affiliation(s)
- Camille Couture
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Jade Rechtzigel
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - SuYun Ling
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Cecilia Ledezma-Soto
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Ines Boufaied
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
| | - Dorothée Dal Soglio
- Department of Pathology and Cellular Biology, Université de Montréal, Montreal, QC, Canada
| | - Evelyne Rey
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| | - Serge McGraw
- Sainte-Justine Hospital Research Center, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology; Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
14
|
Wei J, Zhang L, Xu H, Luo Q. Preterm birth, a consequence of immune deviation mediated hyperinflammation. Heliyon 2024; 10:e28483. [PMID: 38689990 PMCID: PMC11059518 DOI: 10.1016/j.heliyon.2024.e28483] [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: 12/21/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Preterm birth represents a multifaceted syndrome with intricacies still present in our comprehension of its etiology. In the context of a semi-allograft, the prosperity from implantation to pregnancy to delivery hinges on the establishment of a favorable maternal-fetal immune microenvironment and a successful trilogy of immune activation, immune tolerance and then immune activation transitions. The occurrence of spontaneous preterm birth could be related to abnormalities within the immune trilogy, stemming from deviation in maternal and fetal immunity. These immune deviations, characterized by insufficient immune tolerance and early immune activation, ultimately culminated in an unsustainable pregnancy. In this review, we accentuated the role of both innate and adaptive immune reason in promoting spontaneous preterm birth, reviewed the risk of preterm birth from vaginal microbiome mediated by immune changes and the potential of vaginal microbiomes and metabolites as a new predictive marker, and discuss the changes in the role of progesterone and its interaction with immune cells in a preterm birth population. Our objective was to contribute to the growing body of knowledge in the field, shedding light on the immunologic reason of spontaneous preterm birth and effective biomarkers for early prediction, providing a roadmap for forthcoming investigations.
Collapse
Affiliation(s)
- Juan Wei
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - LiYuan Zhang
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - Heng Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| |
Collapse
|
15
|
Cifkova E, Karahoda R, Stranik J, Abad C, Kacerovsky M, Lisa M, Staud F. Metabolomic analysis of the human placenta reveals perturbations in amino acids, purine metabolites, and small organic acids in spontaneous preterm birth. EXCLI JOURNAL 2024; 23:264-282. [PMID: 38487084 PMCID: PMC10938235 DOI: 10.17179/excli2023-6785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/31/2024] [Indexed: 03/17/2024]
Abstract
Spontaneous preterm delivery presents one of the most complex challenges in obstetrics and is a leading cause of perinatal morbidity and mortality. Although it is a common endpoint for multiple pathological processes, the mechanisms governing the etiological complexity of spontaneous preterm birth and the placental responses are poorly understood. This study examined placental tissues collected between May 2019 and May 2022 from a well-defined cohort of women who experienced spontaneous preterm birth (n = 72) and healthy full-term deliveries (n = 30). Placental metabolomic profiling of polar metabolites was performed using Ultra-High Performance Liquid Chromatography/Mass Spectrometry (UHPLC/MS) analysis. The resulting data were analyzed using multi- and univariate statistical methods followed by unsupervised clustering. A comprehensive metabolomic evaluation of the placenta revealed that spontaneous preterm birth was associated with significant changes in the levels of 34 polar metabolites involved in intracellular energy metabolism and biochemical activity, including amino acids, purine metabolites, and small organic acids. We found that neither the preterm delivery phenotype nor the inflammatory response explain the reported differential placental metabolome. However, unsupervised clustering revealed two molecular subtypes of placentas from spontaneous preterm pregnancies exhibiting differential enrichment of clinical parameters. We also identified differences between early and late preterm samples, suggesting distinct placental functions in early spontaneous preterm delivery. Altogether, we present evidence that spontaneous preterm birth is associated with significant changes in the level of placental polar metabolites. Dysregulation of the placental metabolome may underpin important (patho)physiological mechanisms involved in preterm birth etiology and long-term neonatal outcomes.
Collapse
Affiliation(s)
- Eva Cifkova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Sokolska 581, 50005, Hradec Kralove, Czech Republic
| | - Miroslav Lisa
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203/8, 50005, Hradec Kralove, Czech Republic
| |
Collapse
|
16
|
Bernier E, Brien ME, Girard S. Pregnant individuals with uncomplicated pregnancies display pro-inflammatory immune changes when exposed to the COVID-19 pandemic. Am J Reprod Immunol 2024; 91:e13828. [PMID: 38374807 DOI: 10.1111/aji.13828] [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] [Revised: 12/04/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
PROBLEM The COVID-19 pandemic has been shown to have a detrimental impact on the mental health of pregnant individuals, and chronic stress can alter the immune profile. However, the effects of the COVID-19 pandemic on the immune system in pregnancy are still poorly understood. We aimed to evaluate the impact of pandemic exposure on the maternal immune profile in uncomplicated pregnancies. METHOD OF STUDY We collected blood and placenta samples from pregnant individuals exposed and unexposed to the pandemic to compare their immune and inflammatory profiles. We performed co-culture with circulating maternal immune cells and endothelial cells to assess endothelial activation. Statistical analysis was performed using unpaired t-test, Mann-Whitney, or Fisher's exact test as appropriate. RESULTS In exposed individuals, we observed a decreased proportion of Th2 cells (p < .0001) and Treg/Th17 ratio (p < .05), as well as an increased Th1/Th2 ratio (p < .0001). Levels of IL-1β (p < .01) and IL-18 (p < .01) were increased in the circulation of exposed participants, whilst other mediators were significantly decreased (IFNγ, IL-8, MCP-1, amongst others). Furthermore, we observed increased production of ICAM, hallmark of endothelial activation, when we co-cultured endothelial cells with immune cells from exposed individuals. Vaccination status impacted the cellular profile with increased proportions of Th1 and B cells in vaccinated participants. CONCLUSION Overall, we observed a pro-inflammatory bias in the circulation of pregnant individuals exposed to the COVID-19 pandemic, with otherwise uncomplicated pregnancies. Our work also supports an association between the increased risk of endothelial activation/hypertension and SARS-CoV2 infection, which might be driven in part by exposure to the pandemic and associated stressors.
Collapse
Affiliation(s)
- Elsa Bernier
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marie-Eve Brien
- Sainte-Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Jiang Y, Lai X, Liu Y, Yang C, Liu Z, Liu X, Yu T, Chen C, Khanniche A, Fan J, Lin Y, Zeng W. CD8 + T cells in fetal membranes display a unique phenotype, and their activation is involved in the pathophysiology of spontaneous preterm birth. J Pathol 2024; 262:240-253. [PMID: 38018407 DOI: 10.1002/path.6229] [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: 04/04/2023] [Revised: 08/16/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023]
Abstract
Preterm labor/birth is the leading cause of perinatal mortality and morbidity worldwide. Previous studies demonstrated that T cells were crucial for maintaining maternal-fetal immune tolerance during the first trimester of pregnancy; however, their phenotypes and functions in labor and delivery remain largely unknown. We recruited three cohorts of women at delivery for T-cell immunophenotyping in the placentas, fetal membranes, umbilical cord blood, and maternal peripheral blood. Our data showed a differential enrichment of T cells during the third trimester of human pregnancy, with CD4+ T cells being more observable within the umbilical cord blood, whereas CD8+ T cells became relatively more abundant in fetal membranes. CD4+ and CD8+ T cells derived from fetal membranes were dominated by effector memory T cells and exhibited extensive expression of activation markers but decreased expression of homing receptor. In comparison with term births, fetal membrane CD8+ T cells, especially the central memory subset, were significantly increased in frequency and showed more profound activation in spontaneous preterm birth patients. Finally, using an allogeneic mouse model, we found that T-cell-activation-induced preterm birth could be alleviated by the depletion of CD8+ T but not CD4+ T cells in vivo. Collectively, we showed that CD8+ T cells in fetal membranes displayed a unique phenotype, and their activation was involved in the pathophysiology of spontaneous preterm birth, which provides novel insights into the immune mechanisms of preterm birth and potential targets for the prevention of this syndrome. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yinan Jiang
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Xintong Lai
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuxu Liu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Cheng Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Zhicui Liu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Xiaorui Liu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tiantian Yu
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Cailian Chen
- Department of Automation, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, PR China
| | - Asma Khanniche
- Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianxia Fan
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, PR China
| | - Weihong Zeng
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
18
|
Hoffmann JA, Gründler K, Richter DU, Stubert J. Prediction of spontaneous preterm birth using CCL2 and CXCL10 in maternal serum of symptomatic high-risk pregnant women: a prospective cohort study. BMC Pregnancy Childbirth 2023; 23:697. [PMID: 37770883 PMCID: PMC10537471 DOI: 10.1186/s12884-023-06016-3] [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: 03/20/2023] [Accepted: 09/21/2023] [Indexed: 09/30/2023] Open
Abstract
INTRODUCTION CCL2 and CXCL10 are putative biomarkers for the prediction of spontaneous preterm birth. This study evaluates these markers in a cohort of pregnant high-risk women. MATERIAL AND METHODS In our prospective study, we included 109 women with signs of preterm labor between 20 + 0 and 31 + 6 weeks of gestation. Inclusion criteria were regular (< 3/30 min) or painful contractions, cervical length < 25 mm or a history of previous preterm birth (PTB). Blood samples were obtained upon first admission to our clinic. Biomarker concentrations were measured using pre-coated sandwich immunoassays (ELISA). Primary study outcome was spontaneous preterm birth < 34 weeks, secondary outcome was delivery < 37 weeks or within seven days after study inclusion. RESULTS Sixteen women (14.7%) delivered < 34 weeks and twenty women between 34 + 0 and 36 + 6 weeks (18.4%). Six patients (5.5%) gave birth within seven days after study admission. CXCL10 showed higher medium serum levels in women with PTB < 34 weeks (115 pg/ml compared to 61 pg/ml ≥ 34 weeks; p < 0.001) and < 37 weeks (103 pg/ml vs. 53 pg/ml; p < 0.001). In contrary, lower CCL2 serum levels were associated with PTB < 34 weeks (46 pg/ml vs. 73 pg/ml; p = 0.032) and birth within 7 days (25 pg/ml vs. 73 pg/ml; p = 0.008). The CXCL10/CCL2-ratio further improved the predictive model with a ROC-AUC of 0.83 (95% CI 0.73-0.93, p < 0.001) for delivery < 34 weeks. These corresponds to a sensitivity, specificity and positive predictive value of 0.67, 0.86 and 0.43 at a cut-off of 2.2. CONCLUSION Low maternal serum CCL2 levels are associated with a higher risk of preterm delivery within seven days. High CXCL10 serum levels are more associated with a high risk for preterm birth < 34 weeks. Elevated CXCL10/CCL2-ratio is showing the best predictive performance. TRIAL REGISTRATION NUMBER (DRKS-ID) DRKS00010763, Registration date: September 02, 2016.
Collapse
Affiliation(s)
- Jessica Alana Hoffmann
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany
| | - Kathleen Gründler
- Department of Obstetrics and Gynecology, HELIOS Hospital Schwerin, Schwerin, Germany
| | - Dagmar- Ulrike Richter
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany
| | - Johannes Stubert
- Department of Obstetrics and Gynecology, Rostock University Medical Centre, Rostock, Germany.
| |
Collapse
|
19
|
Lou Y, Fu Z, Tian Y, Hu M, Wang Q, Zhou Y, Wang N, Zhang Q, Jin F. Estrogen-sensitive activation of SGK1 induces M2 macrophages with anti-inflammatory properties and a Th2 response at the maternal-fetal interface. Reprod Biol Endocrinol 2023; 21:50. [PMID: 37226177 DOI: 10.1186/s12958-023-01102-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Decidual macrophages participate in immune regulation at the maternal-fetal interface. Abnormal M1/M2 polarization of decidual macrophages might predispose immune maladaptation in recurrent pregnancy loss (RPL). However, the mechanism of decidual macrophage polarization is unclear. We explored the role of Estradiol (E2)-sensitive serum-glucocorticoid regulated kinase (SGK) 1 in promoting macrophage polarization and suppressing inflammation at the maternal-fetal interface. METHODS We assessed serum levels of E2 and progesterone during first trimester of pregnancy in women with or without threatened miscarriages (ended in live birth, n = 448; or early miscarriages, n = 68). For detection of SGK1 in decidual macrophages, we performed immunofluorescence labeling and western blot analysis applying decidual samples from RPL (n = 93) and early normal pregnancy (n = 66). Human monocytic THP-1 cells were differentiated into macrophages and treated with Toll-like receptor (TLR) 4 ligand lipopolysaccharide (LPS), E2, inhibitors or siRNA for in vitro analysis. Flow cytometry analysis were conducted to detect macrophages polarization. We also applied ovariectomized (OVX) mice with hormones exploring the mechanisms underlying the regulation of SGK1 activation by E2 in the decidual macrophages in vivo. RESULTS SGK1 expression down regulation in the decidual macrophages of RPL was consistent with the lower concentration and slower increment of serum E2 from 4 to 12 weeks of gestation seen in these compromised pregnancies. LPS reduced SGK1 activities, but induced the pro-inflammatory M1 phenotype of THP-1 monocyte-derived macrophages and T helper (Th) 1 cytokines that favored pregnancy loss. E2 pretreatment promoted SGK1 activation in the decidual macrophages of OVX mice in vivo. E2 pretreatment amplified SGK1 activation in TLR4-stimulated THP-1 macrophages in vitro through the estrogen receptor beta (ERβ) and PI3K pathway. E2-sensitive activation of SGK1 increased M2 macrophages and Th2 immune responses, which were beneficial to successful pregnancy, by inducing ARG1 and IRF4 transcription, which are implicated in normal pregnancy. The experiments on OVX mice have shown that pharmacological inhibition of E2 promoted nuclear translocation of NF-κB in the decidual macrophages. Further more, pharmacological inhibition or knockdown of SGK1 in TLR4-stimulated THP-1 macrophages activated NF-κB by promoting its nuclear translocation, leading to increased secretion of pro-inflammatory cytokines involved in pregnancy loss. CONCLUSION Our findings highlighted the immunomodulatory roles of E2-activated SGK1 in Th2 immune responses by priming anti-inflammatory M2 macrophages at the maternal-fetal interface, resulting in a balanced immune microenvironment during pregnancy. Our results suggest new perspectives on future preventative strategies for RPL.
Collapse
Affiliation(s)
- Yiyun Lou
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China.
| | - Zhujing Fu
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
- Medical Department, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Ye Tian
- Medical School, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Yangtze River Delta Center for Drug Evaluation and Inspection of National Medical Products Administration, Shanghai, 201210, China
| | - Minhao Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Qijing Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yuanyuan Zhou
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Ning Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Qin Zhang
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Women's Reproductive Healthy Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University, Hangzhou, 310006, China
| |
Collapse
|
20
|
Buchanan CQ, Lawlor ML, Okafor C, Kurian SR, Philip AE, Finkle AE, McQuillan JJ, Haridas S, Koenig JM. Linked Th17 and Calgranulin Responses in Maternal-cord Blood Dyads of Preterm Gestations with Histologic Chorioamnionitis. NEWBORN (CLARKSVILLE, MD.) 2023; 2:133-141. [PMID: 37790838 PMCID: PMC10547109 DOI: 10.5005/jp-journals-11002-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Introduction Maternal-fetal immune crosstalk mechanisms are increasingly identified in the pathogenesis of gestational disorders, including histologic chorioamnionitis (HCA). Although an inflammatory Th17 immune phenotype has been described in preterm neonates with HCA, the associated maternal Th17 response is relatively unknown. To refine our understanding of Th17 biology in this context, we examined Th17 responses in maternal-cord blood dyads of preterm gestations. Materials and methods Paired maternal and cord blood (CB) samples were prospectively collected from preterm gestations (23-34 weeks) with HCA or controls. Th17-linked cell frequencies and plasma calgranulin (S100A8, S100A12) levels were determined by flow cytometry and enzyme-linked immunoassay, respectively. Results Analyses of 47 maternal-cord blood pairs showed striking parallel increases in Th17 cell frequencies as well as plasma calgranulin levels in the presence of fetal inflammation. Cord blood S100A12 levels were directly correlated with Th17 cell frequencies. In CB cultures, rh-S100A12 promoted in vitro propagation of Th17-type CD4+ cells. Conclusions Maternal and CB Th17-linked responses are dually amplified in gestations with HCA, supporting a biological role for maternal-fetal interactions in this disorder. In addition to advancing current knowledge of neonatal Th17 mechanisms, these data shed new light on their association with maternal inflammation.
Collapse
Affiliation(s)
- Christopher Q Buchanan
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Megan L Lawlor
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Chukwuebuka Okafor
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Shannon R Kurian
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrea E Philip
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Abigael E Finkle
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Jay J McQuillan
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Seema Haridas
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Joyce M Koenig
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
21
|
Weng J, Couture C, Girard S. Innate and Adaptive Immune Systems in Physiological and Pathological Pregnancy. BIOLOGY 2023; 12:402. [PMID: 36979094 PMCID: PMC10045867 DOI: 10.3390/biology12030402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023]
Abstract
The dynamic immunological changes occurring throughout pregnancy are well-orchestrated and important for the success of the pregnancy. One of the key immune adaptations is the maternal immune tolerance towards the semi-allogeneic fetus. In this review, we provide a comprehensive overview of what is known about the innate and adaptive immunological changes in pregnancy and the role(s) of specific immune cells during physiological and pathological pregnancy. Alongside this, we provided details of remaining questions and challenges, as well as future perspectives for this growing field of research. Understanding the immunological changes that occur can inform potential strategies on treatments for the optimal health of the neonate and pregnant individual both during and after pregnancy.
Collapse
Affiliation(s)
- Jessica Weng
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Camille Couture
- Department of Microbiology, Infectiology and Immunology, Universite de Montreal, Ste-Justine Hospital Research Center, Montreal, QC H3T 1C5, Canada
| | - Sylvie Girard
- Department of Obstetrics & Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Lodge-Tulloch NA, Toews AJ, Atallah A, Cotechini T, Girard S, Graham CH. Cross-Generational Impact of Innate Immune Memory Following Pregnancy Complications. Cells 2022; 11:3935. [PMID: 36497193 PMCID: PMC9741472 DOI: 10.3390/cells11233935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Pregnancy complications can have long-term negative effects on the health of the affected mothers and their children. In this review, we highlight the underlying inflammatory etiologies of common pregnancy complications and discuss how aberrant inflammation may lead to the acquisition of innate immune memory. The latter can be described as a functional epigenetic reprogramming of innate immune cells following an initial exposure to an inflammatory stimulus, ultimately resulting in an altered response following re-exposure to a similar inflammatory stimulus. We propose that aberrant maternal inflammation associated with complications of pregnancy increases the cross-generational risk of developing noncommunicable diseases (i.e., pregnancy complications, cardiovascular disease, and metabolic disease) through a process mediated by innate immune memory. Elucidating a role for innate immune memory in the cross-generational health consequences of pregnancy complications may lead to the development of novel strategies aimed at reducing the long-term risk of disease.
Collapse
Affiliation(s)
| | - Alexa J. Toews
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Aline Atallah
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|