1
|
Perera TRW, de Ruijter‐Villani M, Gibb Z, Nixon B, Sheridan A, Stout TAE, Swegen A, Skerrett‐Byrne DA. Systemic Changes in Early Pregnancy in the Mare: An Integrated Proteomic Analysis of Blood Plasma, Histotroph, and Yolk Sac Fluid at Day 14 Post-Ovulation. Proteomics Clin Appl 2025; 19:e202400095. [PMID: 39912552 PMCID: PMC11895760 DOI: 10.1002/prca.202400095] [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: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE Embryo-maternal signaling during the establishment of pregnancy in horses remains one of the biggest mysteries in large animal physiology. Early pregnancy loss represents a major source of economic loss to the breeding industry. This study aimed to investigate the systemic changes associated with early pregnancy by mapping the proteome of blood plasma at 14 days in pregnant and non-pregnant mares. EXPERIMENTAL DESIGN Plasma proteomes were analysed in commercially bred pregnant (n = 17) and non-pregnant (n = 17) Thoroughbred mares at 14 days after ovulation, using high-resolution mass spectrometry. Day 14 histotroph and yolk sac fluid were also profiled and datasets were integrated through pathway analysis. RESULTS We identified 229 total protein IDs, with 12 increased and 10 decreased significantly in pregnant versus non-pregnant plasma. To gain functional insight, these data were aligned with proteomes of 14-day pregnant mare uterine fluid (n = 4; 1358 IDs) and conceptus fluid (soluble proteins within the yolk sac fluid; n = 4; 1152 IDs), and further interrogated using gene ontology databases and pathway analysis. CONCLUSIONS AND CLINICAL RELEVANCE These analyses identified consistent systemic changes in the mare's proteome that indicate a profound and specific immune response to early pregnancy, which appears to precede the systemic endocrine response to pregnancy. Integrated pathway analysis suggests that embryo-maternal interactions in early pregnancy may mimic elements of the virus-host interaction to modulate the maternal immune response. Transthyretin (TTR) and uteroglobin (SCGB1A1) were respectively down- and upregulated in plasma while also present in uterine fluid, and are proposed to be key proteins in early pregnancy establishment. These findings contribute significantly to our knowledge of early pregnancy in the mare and identify potential new avenues for developing clinical approaches to reduce early embryo loss.
Collapse
Affiliation(s)
- Tharangani R. W. Perera
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | | | - Zamira Gibb
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Brett Nixon
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alecia Sheridan
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Tom A. E. Stout
- Department of Clinical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Aleona Swegen
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - David A. Skerrett‐Byrne
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- Institute of Experimental GeneticsHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
2
|
Jana B, Andronowska A, Całka J, Mówińska A. Biosynthetic pathway for leukotrienes is stimulated by lipopolysaccharide and cytokines in pig endometrial stromal cells. Sci Rep 2025; 15:2806. [PMID: 39843578 PMCID: PMC11754892 DOI: 10.1038/s41598-025-86787-1] [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: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
An inflammatory response is related to different inflammatory mediators generated by immune and endometrial cells. The links between lipopolysaccharide (LPS), cytokines, and leukotrienes (LTs) in endometrial stromal cells remain unclear. This study aimed to examine the influence of LPS, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-4 and IL-10 on 5-lipooxygenase (5-LO), LTA4 hydrolase (LTAH) and LTC4 synthase (LTCS) mRNA and protein abundances, and LTB4 and cysteinyl (cys)-LTs release including LTC4, by the cultured pig endometrial stromal cells, as well as on cell viability. 24-hour exposure to LPS, TNF-α, IL-4 and IL-10 up-regulated 5-LO mRNA and protein abundances. LPS increased LTAH mRNA abundance, while TNF-α, IL-1β and IL-10 augmented LTAH mRNA and protein abundances. TNF-α and IL-4 increased LTCS mRNA and protein abundances. In addition, LTCS mRNA abundance was enhanced by LPS and IL-4, while LTCS protein abundance was increased by IL-1β. Cells responded to LPS, TNF-α, IL-1β and IL-10 with increased LTB4 release. TNF-α, IL-1β and IL-4 stimulated LTC4 release. Cys-LTs release was up-regulated by LPS, TNF-α, IL-1β and IL-4. All studied cytokines augmented cell viability. In summary, LPS, TNF-α, IL-1β, IL-4 and IL-10 are potential LTs immunomodulatory agents in endometrial stromal cells. These functional interactions could be one of the mechanisms responsible for local orchestrating events in inflamed and healthy endometrium.
Collapse
Affiliation(s)
- Barbara Jana
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, Olsztyn, 10-748, Poland.
| | - Aneta Andronowska
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, Olsztyn, 10-748, Poland
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, Olsztyn, 10-719, Poland
| | - Aleksandra Mówińska
- Division of Reproductive Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, Olsztyn, 10-748, Poland
| |
Collapse
|
3
|
Li X, Li L, Ruan L. Circ_0003314 Combines with the miR-26b-5p/IL1RAP Axis to Inhibit HTR-8/SVneo Cell Proliferation, Migration, Invasion and Tube Formation and Promote Apoptosis. Appl Biochem Biotechnol 2025; 197:443-458. [PMID: 39141177 DOI: 10.1007/s12010-024-05011-4] [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] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Preeclampsia (PE) is a pregnancy-related syndrome that can lead to a variety of pathophysiological processes, such as impaired implantation. The pathogenesis of PE involves circular RNA (circRNA). The study aims to determine the role of a novel circRNA, circ_0003314, in trophoblast cell phenotypes. Circ_0003314, microRNA-26b-5p (miR-26b-5p) and IL-1 receptor accessory protein (IL1RAP) expression were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was investigated by MTT assay and 5-Ethynyl-2'-deoxyuridine assay. Cell migration and invasion were investigated by transwell assay. Cell apoptotic rate and angiogenesis were investigated by flow cytometry analysis and tube formation assay, respectively. Protein expression was detected by western blotting. The binding relationship between miR-26b-5p and circ_0003314 or IL1RAP was identified using dual-luciferase reporter assay and RNA pull-down assay. Circ_0003314 and IL1RAP expression were significantly increased, while miR-26b-5p was decreased in placental tissues of PE patients. Circ_0003314 overexpression inhibited trophoblast cell proliferation, migration, invasion and angiogenesis and induced cell apoptosis. Additionally, circ_0003314 acted as a sponge for miR-26b-5p, and miR-26b-5p bound to IL1RAP. Introduction of miR-26b-5p or silencing of IL1RAP attenuated the effects of circ_0003314 overexpression on trophoblast cell phenotypes. Further, circ_0003314 induced IL1RAP expression through miR-26b-5p in trophoblast cells. Circ_0003314 regulated trophoblast cell phenotypes by increasing IL1RAP expression through binding to miR-26b-5p.
Collapse
Affiliation(s)
- Xi'na Li
- Department of Ultrasound Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Ultrasonography, Xi'an Peoples' Hospital (Xi'an Fourth Hospital), Xi'an, 710005, Shaanxi, China
| | - Lingmin Li
- Department of Ultrasonography, Xi'an Peoples' Hospital (Xi'an Fourth Hospital), Xi'an, 710005, Shaanxi, China
| | - Litao Ruan
- Department of Ultrasound Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
4
|
Xu B, Dissanayake LV, Levchenko V, Zietara A, Kravtsova O, Staruschenko A. Deletion of Kcnj16 altered transcriptomic and metabolomic profiles of Dahl salt-sensitive rats. iScience 2024; 27:110901. [PMID: 39328933 PMCID: PMC11424968 DOI: 10.1016/j.isci.2024.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The inwardly rectifying K+ channel Kir5.1 (Kcnj16) is essential in renal salt handling and blood pressure control. However, the underlying mechanisms are not fully understood. Here, we integrated transcriptomics and metabolomics to comprehensively profile the changes in genes and metabolites in the Dahl salt-sensitive (SS) rat lacking Kcnj16 to identify potential mechanisms. Consistent with the phenotype of knockout (KO) rats, the transcriptomic profile predicted reduced blood pressure, kidney damage, and increased ion transport. Canonical pathway analysis suggested activation of metabolic-related pathways while suppression of immune response-related pathways in KO rats. Untargeted metabolomic analysis revealed different metabolic profiles between wild-type (WT) and KO rats. Integration of transcriptomic and metabolomic profiles suggested altered tricarboxylic acid (TCA) cycle, amino acid metabolism, and reactive oxygen species (ROS) metabolism that are related to SS hypertension. In conclusion, besides increased ion transport, our data suggest suppressed immune response-related and altered metabolic-related pathways of SS rats lacking Kir5.1.
Collapse
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lashodya V. Dissanayake
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Olha Kravtsova
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA
- James A. Haley Veteran’s Hospital, Tampa, FL, USA
| |
Collapse
|
5
|
Guelfi G, Dall’Aglio C, Bufalari A, Mercati F, Anipchenko P, Capaccia C, Cocci P, Palermo FA, Acuti G, Troisi A, Tomassoni D, Boiti C, Zerani M, Maranesi M. Interleukin-1 Beta (IL1B) and Nerve Growth Factor (NGF): Key Players in Rabbit Reproductive Regulation. Int J Mol Sci 2024; 25:10986. [PMID: 39456770 PMCID: PMC11506888 DOI: 10.3390/ijms252010986] [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: 09/05/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Several seminal plasma components, besides NGF, are implicated as ovulation-inducing factors in mammals. This study investigated the IL1B and its receptor IL1R1 in the testis (T), male accessory glands, prostate (P) and seminal vesicles (SV), and uterus (U) of adult rabbits using immunohistochemistry (IHC) and quantitative reverse transcription PCR (RT-qPCR). We also assessed the presence of IL1B in seminal plasma through Western blotting (WB) and examined the interaction between IL1B and NGF in vitro by measuring their production with enzyme-linked immunosorbent assay (ELISA) in the presence of NGF and IL1B alone or with their respective receptor antagonists. IHC revealed IL1B system expression in all reproductive organs studied, with IL1B and IL1R1 localized to the germinative epithelium of the T and the epithelial cells of the accessory glands and U. IL1B gene transcript levels were significantly higher (p < 0.01) in the P and SV compared to the T, while IL1R1 levels were significantly higher (p < 0.001) in the P compared to the other tissues, while IL1R1 levels were three times higher (p < 0.001) in the P. WB confirmed the presence of IL1B in seminal plasma with a 30-35 kDa band. The in vitro study demonstrated that IL1B increased (p < 0.05) basal NGF production in the U, whereas NGF had no effect on IL1B production. These findings provide evidence of the expression of the IL1B/IL1R1 system in both male and female rabbit reproductive tracts and suggest that IL1B in seminal plasma may influence uterine endocrine activity. The results propose a potential role for IL1B in ovulation, in conjunction with NGF, supporting that ovulation may involve inflammatory-like processes.
Collapse
Affiliation(s)
- Gabriella Guelfi
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Cecilia Dall’Aglio
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Antonello Bufalari
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Francesca Mercati
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Polina Anipchenko
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Camilla Capaccia
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Paolo Cocci
- School of Bioscience and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Macerata, Italy; (P.C.); (F.A.P.); (A.T.); (D.T.)
| | - Francesco Alessandro Palermo
- School of Bioscience and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Macerata, Italy; (P.C.); (F.A.P.); (A.T.); (D.T.)
| | - Gabriele Acuti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Alessandro Troisi
- School of Bioscience and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Macerata, Italy; (P.C.); (F.A.P.); (A.T.); (D.T.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Macerata, Italy; (P.C.); (F.A.P.); (A.T.); (D.T.)
| | - Cristiano Boiti
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Massimo Zerani
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| | - Margherita Maranesi
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy; (G.G.); (C.D.); (P.A.); (C.C.); (G.A.); (C.B.); (M.Z.); (M.M.)
| |
Collapse
|
6
|
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
|
7
|
Yan S, Lyu L, Wang X, Wen H, Li Y, Li J, Yao Y, Zuo C, Xie S, Wang Z, Qi X. Pro-inflammatory cytokine IL1β1 participates in promoting parturition related pathways in the ovoviviparous teleost black rockfish (Sebastes schlegelii). Biol Reprod 2023; 109:693-704. [PMID: 37593921 DOI: 10.1093/biolre/ioad100] [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: 12/20/2022] [Revised: 07/24/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023] Open
Abstract
Along with the evolution process, the reproductive strategies evolved including oviparity, viviparity and ovoviviparity, to fit the residential environment maximize the survival rate of the off spring. In mammals, the key to the initiation of parturition is the inflammatory response at the maternal-fetal interface. As a pro-inflammatory cytokine, interleukin 1 beta (IL1β) plays an important role in the process of human parturition. While less is known about IL1β1 in teleost parturition, identification of the functions of IL1β1 in inducing the parturition, black rockfish, an ovoviviparity teleost, which provides over 60% nutrition supply for over 50 000 embryos though a placenta like structure during pregnant, was employed as the research model. In the present study, based on the gene cloning, we detected the expression pattern of both Il1b1 and its receptor perinatal period, as well as the localization to the ovary by in situ hybridization. The different expression genes in transcriptomic data of perinatal primary ovarian cells treated with the recombinant IL1β1 (rIL1β1) obtained by prokaryotic expression system were analyzed. Differentially expressed genes, functional enrichment and pathway analysis mainly included immune response, signal transduction and cell death. In summary, our research provides novel insights into the potential role of IL1β1 in the parturition of ovoviviparity teleost.
Collapse
Affiliation(s)
- Shaojing Yan
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Xiaojie Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Jianshuang Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Yijia Yao
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Chenpeng Zuo
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Songyang Xie
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| | - Zhijun Wang
- Weihai Taifeng Seawater Seedling Co., LTD, Weihai, PR China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, PR China
| |
Collapse
|
8
|
Yu F, Xing J, Li L, Xiang M. CircCRIM1 mediates proliferation, migration, and invasion of trophoblast cell through regulating miR-942-5p/IL1RAP axis. Am J Reprod Immunol 2023; 90:e13699. [PMID: 37382169 DOI: 10.1111/aji.13699] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/20/2023] [Accepted: 03/16/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a severe complication that occurs during pregnancy and a main cause of perinatal mortality of mothers as well as infants, which is characterized by abnormal placental trophoblast. Previous study reported that aberrant circular RNA (circRNA) was involved in the pathogenesis and progression of PE. Herein, we aimed to investigate the role of circCRIM1 and explore the mechanism of circCRIM1 in PE. METHODS The quantitative real-time PCR (qRT-PCR) was conducted to determine the relative expression of circCRIM1, miR-942-5p, and IL1RAP in tissues and cells. Cell proliferation viability was assessed by both MTT and EdU assays. Cell cycle distribution was analyzed using flow cytometry. Transwell assay was performed to test the cell migration and invasion. The protein levels of CyclinD1, MMP9, MMP2, and IL1RAP were measured by western blot. The putative binding sites between miR-942-5p and circCRIM1 or IL1RAP 3'UTR were verified by dual-luciferase reporter gene assay. Rescue experiment was performed to confirm that miR-942-5p/IL1RAP axis was functional target of circCRIM1 in trophoblast cells. RESULTS CircCRIM1 was upregulated in placenta tissues of PE and its expression was inversely related to infant weight. Overexpression of circCRIM1 suppressed proliferation, migration, and invasion and reduced the protein levels of CyclinD1, MMP9, MMP2 of trophoblast cells, whereas its knockdown exerted the opposite effect. CircCRIM1 could interact with miR-942-5p, and introduction of miR-942-5p partially abated the inhibitory effect of circCRIM1 on trophoblast cell behaviors. IL1RAP was directly targeted and negatively regulated by miR-942-5p. miR-942-5p played its regulatory role on cell proliferation, migration, and invasion of trophoblast by IL1RAP. Further analysis showed that circCRIM1 modulated IL1RAP expression via sponging miR-942-5p. CONCLUSION The results of the present study demonstrated that circCRIM1 inhibited the proliferation, migration, and invasion of trophoblast cells through sponging miR-942-5p and up-regulating IL1RAP, providing a possible new mechanism of PE.
Collapse
Affiliation(s)
- Fen Yu
- Department of Gynecology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jie Xing
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Lingyun Li
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Mi Xiang
- Department of Obstetrics, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Lopes NA, Ambeskovic M, King SE, Faraji J, Soltanpour N, Falkenberg EA, Scheidl T, Patel M, Fang X, Metz GAS, Olson DM. Environmental Enrichment Promotes Transgenerational Programming of Uterine Inflammatory and Stress Markers Comparable to Gestational Chronic Variable Stress. Int J Mol Sci 2023; 24:ijms24043734. [PMID: 36835144 PMCID: PMC9962069 DOI: 10.3390/ijms24043734] [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: 11/16/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/15/2023] Open
Abstract
Prenatal maternal stress is linked to adverse pregnancy and infant outcomes, including shortened gestation lengths, low birth weights, cardio-metabolic dysfunction, and cognitive and behavioural problems. Stress disrupts the homeostatic milieu of pregnancy by altering inflammatory and neuroendocrine mediators. These stress-induced phenotypic changes can be passed on to the offspring epigenetically. We investigated the effects of gestational chronic variable stress (CVS) in rats using restraint and social isolation stress in the parental F0 generation and its transgenerational transmission across three generations of female offspring (F1-F3). A subset of F1 rats was housed in an enriched environment (EE) to mitigate the adverse effects of CVS. We found that CVS is transmitted across generations and induces inflammatory changes in the uterus. CVS did not alter any gestational lengths or birth weights. However, inflammatory and endocrine markers changed in the uterine tissues of stressed mothers and their offspring, suggesting that stress is transgenerationally transmitted. The F2 offspring reared in EE had increased birth weights, but their uterine gene expression patterns remained comparable to those of stressed animals. Thus, ancestral CVS induced changes transgenerationally in fetal programming of uterine stress markers over three generations of offspring, and EE housing did not mitigate these effects.
Collapse
Affiliation(s)
- Nayara A. Lopes
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mirela Ambeskovic
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Stephanie E. King
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Jamshid Faraji
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Nasrin Soltanpour
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Erin A. Falkenberg
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Taylor Scheidl
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mansi Patel
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Xin Fang
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Gerlinde A. S. Metz
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Correspondence: (G.A.S.M.); (D.M.O.); Tel.: +1-403-394-3992 (G.A.S.M.); +1-780-492-8559 (D.M.O.)
| | - David M. Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Correspondence: (G.A.S.M.); (D.M.O.); Tel.: +1-403-394-3992 (G.A.S.M.); +1-780-492-8559 (D.M.O.)
| |
Collapse
|
10
|
Fedotcheva TA, Fedotcheva NI, Shimanovsky NL. Progesterone as an Anti-Inflammatory Drug and Immunomodulator: New Aspects in Hormonal Regulation of the Inflammation. Biomolecules 2022; 12:biom12091299. [PMID: 36139138 PMCID: PMC9496164 DOI: 10.3390/biom12091299] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 11/25/2022] Open
Abstract
The specific regulation of inflammatory processes by steroid hormones has been actively studied in recent years, especially by progesterone (P4) and progestins. The mechanisms of the anti-inflammatory and immunomodulatory P4 action are not fully clear. The anti-inflammatory effects of P4 can be defined as nonspecific, associated with the inhibition of NF-κB and COX, as well as the inhibition of prostaglandin synthesis, or as specific, associated with the regulation of T-cell activation, the regulation of the production of pro- and anti-inflammatory cytokines, and the phenomenon of immune tolerance. The specific anti-inflammatory effects of P4 and its derivatives (progestins) can also include the inhibition of proliferative signaling pathways and the antagonistic action against estrogen receptor beta-mediated signaling as a proinflammatory and mitogenic factor. The anti-inflammatory action of P4 is accomplished through the participation of progesterone receptor (PR) chaperones HSP90, as well as immunophilins FKBP51 and FKBP52, which are the validated targets of clinically approved immunosuppressive drugs. The immunomodulatory and anti-inflammatory effects of HSP90 inhibitors, tacrolimus and cyclosporine, are manifested, among other factors, due to their participation in the formation of an active ligand–receptor complex of P4 and their interaction with its constituent immunophilins. Pharmacological agents such as HSP90 inhibitors can restore the lost anti-inflammatory effect of glucocorticoids and P4 in chronic inflammatory and autoimmune diseases. By regulating the activity of FKBP51 and FKBP52, it is possible to increase or decrease hormonal signaling, as well as restore it during the development of hormone resistance. The combined action of immunophilin suppressors with steroid hormones may be a promising strategy in the treatment of chronic inflammatory and autoimmune diseases, including endometriosis, stress-related disorders, rheumatoid arthritis, and miscarriages. Presumably, the hormone receptor- and immunophilin-targeted drugs may act synergistically, allowing for a lower dose of each.
Collapse
Affiliation(s)
- Tatiana A. Fedotcheva
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
- Correspondence: ; Tel.: +7-9169353196
| | - Nadezhda I. Fedotcheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str. 3, Pushchino 142290, Russia
| | - Nikolai L. Shimanovsky
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, Moscow 117997, Russia
| |
Collapse
|
11
|
Social Isolation Stress Modulates Pregnancy Outcomes and the Inflammatory Profile of Rat Uterus. Int J Mol Sci 2022; 23:ijms23116169. [PMID: 35682846 PMCID: PMC9181517 DOI: 10.3390/ijms23116169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/26/2022] [Accepted: 05/28/2022] [Indexed: 11/24/2022] Open
Abstract
Prenatal stressors have been linked to adverse pregnancy outcomes; including preterm birth (PTB). Recent work demonstrates that social isolation in mothers represents a silent stressor contributing to PTB risk. Here; we investigate the association of inflammatory and stress markers with PTB risk in Long–Evans rats exposed to social isolation stress (SIS) during preconception and pregnancy across four generations (F0-F3). Gestational length; blood glucose; corticosterone levels; and maternal and offspring weights were assessed in two SIS paradigms: transgenerational (TG) and multigenerational (MG) exposure. Maternal uterine tissues were collected 21 days after the dams gave birth. Exposure to SIS reduced pregnancy lengths in the parental generation and neonatal birth weights in the F1 and F2 generations. Interleukin (IL)-1β (Il1b) mRNA levels increased in F0 animals but decreased in the offspring of both stress lineages. Protein levels of IL-1β decreased in the TG lineage. Corticotrophin-releasing hormone receptor 1 (Crhr1) expression decreased in SIS-exposed F0 animals and increased in the TG-F2 and MG-F1 offspring. Expression of enzyme 11-β hydroxysteroid dehydrogenase-2 (11bHSD2) was enhanced in F1 animals. These findings suggest SIS has adverse consequences on the F0 mothers; but their F1–F3 progeny may adapt to this chronic stress; thus supporting the fetal programming hypothesis.
Collapse
|
12
|
Lee H, Patel V, Onushko M, Fang X, Chemtob S, Olson D. A Leukocyte Migration Assay Assists Understanding of Interleukin-1β-Induced Leukocyte Migration Into Preterm Mouse Uterus. Front Pharmacol 2022; 13:898008. [PMID: 35694257 PMCID: PMC9174527 DOI: 10.3389/fphar.2022.898008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Neutrophils and other leukocytes invade the mouse uterus at term birth, which is normal for activating the uterus for labor. To better understand the regulation of this migration at term and interleukin (IL)-1β—induced preterm birth, we developed a mouse leukocyte migration assay (mLMA) and used it with rytvela, an IL-1 receptor allosteric antagonist. The mLMA uses term peripheral blood leukocytes that migrate in a Boyden chamber in response to a chemoattractant. We tested several mouse uterine tissues after homogenization and sedimentation of debris for chemoattractant activity. The most active chemoattractant homogenate came from the mouse lower uterus on gestational day (GD) 18.5. Using flow cytometry, we demonstrated that 99% of the cells that migrate are neutrophils. IL-1β administered on GD 16 stimulated neutrophil migration and invasion into the uterus and the fetal brain along with preterm birth on GD 17. Preterm birth and the increased leukocyte invasion of the maternal uterus and fetal brain were all blocked by the co-administration of rytvela. To test where the site of IL-1β action might be, we examined the potency of lower uterine chemoattractant and the activation of leukocytes following IL-1β +/- rytvela administration. IL-1β did not increase lower uterus homogenate chemoattractant activity, but it significantly (p < 0.05) increased leukocyte activation as defined by cytokine and chemokine expression. Rytvela blocked this activation of leukocytes by IL-1β. We conclude that IL-1β stimulates preterm birth in mice by increasing leukocyte activation leading to increased uterine and fetal brain leukocyte invasion.
Collapse
Affiliation(s)
- Han Lee
- Olson Laboratory, Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Vaishvi Patel
- Olson Laboratory, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Meghan Onushko
- Olson Laboratory, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Xin Fang
- Olson Laboratory, Department of Obstetrics/Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Sylvain Chemtob
- Chemtob Laboratory, Departments of Pediatrics and Ophthalmology/Pharmacology, University of Montreal, Montreal, QC, Canada
| | - David Olson
- Olson Laboratory, Department of Physiology, University of Alberta, Edmonton, AB, Canada
- Olson Laboratory, Department of Obstetrics/Gynecology, University of Alberta, Edmonton, AB, Canada
- Olson Laboratory, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- *Correspondence: David Olson,
| |
Collapse
|
13
|
Xing H, Ding Q, Lu H, Li Q. Circ_0007611 stimulates IL-1 receptor accessory protein to inhibit trophoblast cell proliferation and induce cell apoptosis. Biol Reprod 2022; 106:1011-1021. [PMID: 35238896 DOI: 10.1093/biolre/ioac028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/14/2022] Open
Abstract
Preeclampsia (PE) is a common pregnancy disorder, and mounting evidence has revealed that circular RNA participates in PE development. However, the detailed molecular mechanism of circ_0007611 in PE progression remains unknown. RNA expressions of circ_0007611, microRNA-558 (miR-558), and IL-1 receptor accessory protein (IL1RAP) were detected by quantitative real-time polymerase chain reaction. Cell proliferation was investigated by clonogenicity, 5-Ethynyl-29-deoxyuridine, and DNA content quantitation assays. Cell apoptotic rate and angiogenesis were analyzed by cell apoptosis and tube formation assays, respectively. Protein expression was detected by western blot. The binding relationship between miR-558 and circ_0007611 or IL1RAP was identified by a dual-luciferase reporter or RNA immunoprecipitation assay. Circ_0007611 and IL1RAP expressions were significantly upregulated, while miR-558 was downregulated in the placental tissues of PE women in comparison with normal placental tissues. Functionally, circ_0007611 overexpression inhibited trophoblast cell proliferation and angiogenesis and induced cell apoptosis; however, circ_0007611 downregulation showed the opposite effects. Mechanistically, circ_0007611 acted as a miR-558 sponge, and miR-558 bound to IL1RAP. Besides, miR-558 overexpression or IL1RAP absence relieved circ_0007611-induced trophoblast cell dysfunction. Moreover, miR-558 contributed to cell proliferation and tube formation and inhibited cell apoptosis by reducing IL1RAP expression in trophoblast cells. Circ_0007611 aggravated trophoblast cell disorders by the miR-558/IL1RAP pathway in PE.
Collapse
Affiliation(s)
- Haiyan Xing
- Department of Gynaecology and Obstetrics, Jinan Fourth People's Hospital, Jinan, Shandong, China
| | - Qing Ding
- Department of Gynaecology and Obstetrics, Jinan Fourth People's Hospital, Jinan, Shandong, China
| | - Hong Lu
- Department of Gynaecology and Obstetrics, Jinan Fourth People's Hospital, Jinan, Shandong, China
| | - Qun Li
- Department of Gynaecology and Obstetrics, Jinan Fourth People's Hospital, Jinan, Shandong, China
| |
Collapse
|
14
|
Leimert KB, Xu W, Princ MM, Chemtob S, Olson DM. Inflammatory Amplification: A Central Tenet of Uterine Transition for Labor. Front Cell Infect Microbiol 2021; 11:660983. [PMID: 34490133 PMCID: PMC8417473 DOI: 10.3389/fcimb.2021.660983] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/30/2021] [Indexed: 11/23/2022] Open
Abstract
In preparation for delivery, the uterus transitions from actively maintaining quiescence during pregnancy to an active parturient state. This transition occurs as a result of the accumulation of pro-inflammatory signals which are amplified by positive feedback interactions involving paracrine and autocrine signaling at the level of each intrauterine cell and tissue. The amplification events occur in parallel until they reach a certain threshold, ‘tipping the scale’ and contributing to processes of uterine activation and functional progesterone withdrawal. The described signaling interactions all occur upstream from the presentation of clinical labor symptoms. In this review, we will: 1) describe the different physiological processes involved in uterine transition for each intrauterine tissue; 2) compare and contrast the current models of labor initiation; 3) introduce innovative models for measuring paracrine inflammatory interactions; and 4) discuss the therapeutic value in identifying and targeting key players in this crucial event for preterm birth.
Collapse
Affiliation(s)
- Kelycia B Leimert
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Magdalena M Princ
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Song XL, Zhang FF, Wang WJ, Li XN, Dang Y, Li YX, Yang Q, Shi MJ, Qi XY. LncRNA A2M-AS1 lessens the injury of cardiomyocytes caused by hypoxia and reoxygenation via regulating IL1R2. Genes Genomics 2020; 42:1431-1441. [PMID: 33057899 DOI: 10.1007/s13258-020-01007-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial ischemia and reperfusion injury (MI/RI) is a complex pathophysiological process, which can lead to severe myocardial injury. The long noncoding RNA alpha-2-macroglobulin antisense RNA 1 (A2M-AS1) has been revealed to be abnormally expressed in MI, However, its function in MI and the potential mechanism are still unclear. OBJECTIVE To evaluate the functional role of A2M-AS1 in hypoxia/reoxygenation (H/R)-induced neonatal cardiomyocytes and its potential molecular mechanism. METHODS Dataset GSE66360 was obtained from GEO database for analyzing the RNA expression of A2M-AS1 and interleukin 1 receptor type 2 (IL1R2). KEGG pathway enrichment analysis of the genes that co-expressed with A2M-AS1 was performed. Human neonatal cardiomyocytes were subjected to H/R to construct in vitro models. QRT-PCR and Western blot were adopted to test the levels of mRNA and protein. The viability and apoptosis of cardiomyocytes were tested by CCK-8 and flow cytometry assays, respectively. RESULTS The expression of A2M-AS1 was notably downregulated in H/R-treated cardiomyocytes. Overexpression of A2M-AS1 can notably enhance the cell viability of H/R-damaged cardiomyocytes, whereas knockdown of A2M-AS1 showed the opposite outcomes. Besides, a negative correlation was showed between A2M-AS1 and IL1R2 expression. In H/R-treated cardiomyocytes, overexpression of IL1R2 weakened the promoting proliferation and anti-apoptosis effects caused by overexpressing A2M-AS1, however, IL1R2-knockdown abolished the anti-proliferation and pro-apoptosis effects caused by silencing A2M-AS1. CONCLUSION This study demonstrates the potential regulatory role of A2M-AS1/ IL1R2 axis in cardiomyocytes suffered from H/R, and provides insight into the protection of MI/RI.
Collapse
Affiliation(s)
- Xue-Lian Song
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Fei-Fei Zhang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Wen-Jing Wang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xin-Ning Li
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Yi Dang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Ying-Xiao Li
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Qian Yang
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Mei-Jing Shi
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xiao-Yong Qi
- Graduate School, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Department of Cardiology Center, Hebei General Hospital, No. 348 of Heping West Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
16
|
Leimert KB, Verstraeten BSE, Messer A, Nemati R, Blackadar K, Fang X, Robertson SA, Chemtob S, Olson DM. Cooperative effects of sequential PGF2α and IL-1β on IL-6 and COX-2 expression in human myometrial cells†. Biol Reprod 2020; 100:1370-1385. [PMID: 30794283 DOI: 10.1093/biolre/ioz029] [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: 06/26/2018] [Revised: 10/17/2018] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
The change from the state of pregnancy to the state of parturition, which we call uterine transitioning, requires the actions of inflammatory mediators and results in an activated uterus capable of performing the physiology of labor. Interleukin (IL)-1β and prostaglandin (PG)F2α are two key mediators implicated in preparing the uterus for labor by regulating the expression of uterine activation proteins (UAPs) and proinflammatory cytokines and chemokines. To investigate this process, primary human myometrial smooth muscle cells (HMSMC) isolated from the lower segment of women undergoing elective cesarean sections at term (not in labor) were used to test the inflammatory cytokine and UAP outputs induced by PGF2α and IL-1β alone or in sequential combinations. PGF2α and IL-1β regulate mRNA abundance of the PGF2α receptor FP, the IL-1 receptor system, interleukin 6, and other UAPs (OXTR, COX2), driving positive feedback interactions to further amplify their own proinflammatory effects. Sequential stimulation of HMSMC by PGF2α and IL-1β in either order results in amplified upregulation of IL-6 and COX-2 mRNA and protein, compared to their effects individually. These profound increases were unique to myometrium and not observed with stimulation of human fetal membrane explants. These results suggest that PGF2α and IL-1β act cooperatively upstream in the birth cascade to maximize amplification of IL-6 and COX-2, to build inflammatory load and thereby promote uterine transition. Targeting PGF2α or IL-1β, their actions, or intermediates (e.g. IL-6) would be an effective therapeutic intervention for preterm birth prevention or delay.
Collapse
Affiliation(s)
- Kelycia B Leimert
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Angela Messer
- Department of Obstetrics, Gynecology and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Rojin Nemati
- Department of Obstetrics, Gynecology and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Kayla Blackadar
- Department of Obstetrics, Gynecology and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Xin Fang
- Department of Obstetrics, Gynecology and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sarah A Robertson
- Department of Obstetrics and Gynecology, University of Adelaide, Adelaide, South Australia, Australia
| | - Sylvain Chemtob
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine Research Center, Montréal, Quebec, Canada
| | - David M Olson
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.,Department of Obstetrics, Gynecology and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Equils O, Kellogg C, McGregor J, Gravett M, Neal-Perry G, Gabay C. The role of the IL-1 system in pregnancy and the use of IL-1 system markers to identify women at risk for pregnancy complications†. Biol Reprod 2020; 103:684-694. [PMID: 32543660 DOI: 10.1093/biolre/ioaa102] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
The interleukin (IL)-1 system plays a major role in immune responses and inflammation. The IL-1 system components include IL-1α, IL-1β, IL-1 receptor type 1 and IL-1 receptor type 2 (decoy receptor), IL-1 receptor accessory protein, and IL-1 receptor antagonist (IL-1Ra). These components have been shown to play a role in pregnancy, specifically in embryo-maternal communication for implantation, placenta development, and protection against infections. As gestation advances, maternal tissues experience increasing fetal demand and physical stress and IL-1β is induced. Dependent on the levels of IL-1Ra, which regulates IL-1β activity, a pro-inflammatory response may or may not occur. If there is an inflammatory response, prostaglandins are synthesized that may lead to myometrial contractions and the initiation of labor. Many studies have examined the role of the IL-1 system in pregnancy by independently measuring plasma, cervical, and amniotic fluid IL-1β or IL-1Ra levels. Other studies have tested for polymorphisms in IL-1β and IL-1Ra genes in women experiencing pregnancy complications such as early pregnancy loss, in vitro fertilization failure, pre-eclampsia and preterm delivery. Data from those studies suggest a definite role for the IL-1 system in successful pregnancy outcomes. However, as anticipated, the results varied among different experimental models, ethnicities, and disease states. Here, we review the current literature and propose that measurement of IL-1Ra in relation to IL-1 may be useful in predicting the risk of poor pregnancy outcomes.
Collapse
Affiliation(s)
| | - Caitlyn Kellogg
- RPI Consulting LLC, Los Angeles, CA, USA.,San Diego School of Medicine, University of California, San Diego, CA, USA
| | | | - Michael Gravett
- Department of Obstetrics and Gynecology, University of Washington Medical Center, Seattle, WA, USA
| | - Genevieve Neal-Perry
- Department of Obstetrics and Gynecology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Cem Gabay
- University of Geneva, Geneva, Switzerland
| |
Collapse
|
18
|
Lee JM, Mayall JR, Chevalier A, McCarthy H, Van Helden D, Hansbro PM, Horvat JC, Jobling P. Chlamydia muridarum infection differentially alters smooth muscle function in mouse uterine horn and cervix. Am J Physiol Endocrinol Metab 2020; 318:E981-E994. [PMID: 32315215 DOI: 10.1152/ajpendo.00513.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chlamydia trachomatis infection is a primary cause of reproductive tract diseases including infertility. Previous studies showed that this infection alters physiological activities in mouse oviducts. Whether this occurs in the uterus and cervix has never been investigated. This study characterized the physiological activities of the uterine horn and the cervix in a Chlamydia muridarum (Cmu)-infected mouse model at three infection time points of 7, 14, and 21 days postinfection (dpi). Cmu infection significantly decreased contractile force of spontaneous contraction in the cervix (7 and 14 dpi; P < 0.001 and P < 0.05, respectively), but this effect was not observed in the uterine horn. The responses of the uterine horn and cervix to oxytocin were significantly altered by Cmu infection at 7 dpi (P < 0.0001), but such responses were attenuated at 14 and 21 dpi. Cmu infection increased contractile force to prostaglandin (PGF2α) by 53-83% in the uterine horn. This corresponded with the increased messenger ribonucleic acid (mRNA) expression of Ptgfr that encodes for its receptor. However, Cmu infection did not affect contractions of the uterine horn and cervix to PGE2 and histamine. The mRNA expression of Otr and Ptger4 was inversely correlated with the mRNA expression of Il1b, Il6 in the uterine horn of Cmu-inoculated mice (P < 0.01 to P < 0.001), suggesting that the changes in the Otr and Ptger4 mRNA expression might be linked to the changes in inflammatory cytokines. Lastly, this study also showed a novel physiological finding of the differential response to PGE2 in mouse uterine horn and cervix.
Collapse
Affiliation(s)
- Jia Ming Lee
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Anne Chevalier
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Huw McCarthy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Dirk Van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
- Centenary Institute and the University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| |
Collapse
|
19
|
Leimert KB, Messer A, Gray T, Fang X, Chemtob S, Olson DM. Maternal and fetal intrauterine tissue crosstalk promotes proinflammatory amplification and uterine transition†. Biol Reprod 2020; 100:783-797. [PMID: 30379983 DOI: 10.1093/biolre/ioy232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/06/2018] [Accepted: 10/29/2018] [Indexed: 12/24/2022] Open
Abstract
Birth is a complex biological event requiring genetic, cellular, and physiological changes to the uterus, resulting in a uterus activated for completing the physiological processes of labor. We define the change from the state of pregnancy to the state of parturition as uterine transitioning, which requires the actions of inflammatory mediators and localized paracrine interactions between intrauterine tissues. Few studies have examined the in vitro interactions between fetal and maternal gestational tissues within this proinflammatory environment. Thus, we designed a co-culture model to address this gap, incorporating primary term human myometrium smooth muscle cells (HMSMCs) with human fetal membrane (hFM) explants to study interactions between the tissues. We hypothesized that crosstalk between tissues at term promotes proinflammatory expression and uterine transitioning for parturition. Outputs of 40 cytokines and chemokines encompassing a variety of proinflammatory roles were measured; all but one increased significantly with co-culture. Eighteen of the 39 cytokines increased to a higher abundance than the sum of the effect of each tissue cultured separately. In addition, COX2 and IL6 but not FP and OXTR mRNA abundance significantly increased in both HMSMCs and hFM in response to co-culture. These data suggest that synergistic proinflammatory upregulation within intrauterine tissues is involved with uterine transitioning.
Collapse
Affiliation(s)
- Kelycia B Leimert
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Angela Messer
- Departments of Obstetrics & Gynecology, and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Theora Gray
- Departments of Obstetrics & Gynecology, and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Xin Fang
- Departments of Obstetrics & Gynecology, and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - David M Olson
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.,Departments of Obstetrics & Gynecology, and Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Verstraeten BSE, McCreary JK, Weyers S, Metz GAS, Olson DM. Prenatal two-hit stress affects maternal and offspring pregnancy outcomes and uterine gene expression in rats: match or mismatch? Biol Reprod 2020; 100:195-207. [PMID: 30084951 DOI: 10.1093/biolre/ioy166] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 07/29/2018] [Indexed: 01/01/2023] Open
Abstract
Maternal stress and inflammation excesses can lead to adverse pregnancy outcomes and offspring development. We evaluated whether distinct prenatal stressors affect pregnancy, maternal and offspring outcomes, and uterine gene expression differently when combined than either alone. Long-Evans dams were exposed to psychological or/and (two-hit) immune stress (interleukin-1 beta [IL-1β]), on gestational days 12-18 and 17-delivery, respectively. Gestational length, maternal weight gain, glycaemia and corticosterone levels, offspring weight, and gender effects were recorded. Maternal and offspring uteri were collected at weaning and on postnatal day 160 correspondingly. Uterine expression of genes involved in local progesterone metabolism, neuroendocrine and immune systems were analyzed using quantitative real-time polymerase chain reaction. Maternal two-hit stress increased gestational length variation and the occurrence of adverse pregnancy outcomes while reducing gestational weight gain. Pup weight was negatively affected by prenatal stressors in a gender-specific way. In dams, IL-1β upregulated gene expression of neuroendocrine (Crh, Crhr1) and cytokine genes (Il1b, Il1rn, Il6, and Il10). Conversely, transcriptional patterns in offspring uteri were more variable with gene-specific up- or downregulation by each stressor separately, while exposure to both extensively reduced the expression of neuroendocrine (Hsd11b1), cytokine (Il1a, Il1rn, Il6), and IL-1 receptor genes. In conclusion, maternal stress affects physiological and molecular processes in dams and their offspring; two hits have different effects than single stressors. Outcomes appear generation-, gender-, and stressor-specific. Dampening of offspring uterine gene expression after exposure to multiple stressors could fit within the match/mismatch hypothesis of perinatal programming, with offspring preparing for a stressful life.
Collapse
Affiliation(s)
- Barbara S E Verstraeten
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada.,Department of Uro-Gynaecology, Ghent University, Ghent, Belgium
| | - J Keiko McCreary
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - Steven Weyers
- Department of Uro-Gynaecology, Ghent University, Ghent, Belgium
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, Alberta, Canada
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada.,Departments of Pediatrics and Physiology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Verstraeten BSE, McCreary JK, Falkenberg EA, Fang X, Weyers S, Metz GAS, Olson DM. Multiple prenatal stresses increase sexual dimorphism in adult offspring behavior. Psychoneuroendocrinology 2019; 107:251-260. [PMID: 31174163 DOI: 10.1016/j.psyneuen.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Maternal gestational stress and immune activation have independently been associated with affective and neurodevelopmental disorders across the lifespan. We investigated whether rats exposed to prenatal maternal stressors (PNMS) consisting of psychological stress, interleukin (IL)-1β or both (two-hit stress) during critical developmental windows displayed a behavioral phenotype representative of these conditions. METHODS Long-Evans dams were exposed to psychological stressors consisting of restraint stress and forced swimming from gestational day (GD)12 to 18 or to no stress (controls). From GD17 until day of delivery, these same animals were injected with saline or IL-1β as a second hit and immune stressor (5 μg/day, intraperitoneally). The behavior of F1 offspring adults was tested on the open field test, elevated plus maze and affective exploration task on postnatal days (P)90, 100 and 110 respectively. RESULTS The effects of PNMS differed depending on the specific testing environment and potentially the age at assessment, especially in female offspring. Both locomotion and anxiety-like behavioral measures were susceptible to PNMS effects. In females, psychological stress increased anxiety-like behavior, whereas IL-1β had an opposite effect, inducing exploration and risk-taking behavior on the open field test and the elevated plus maze. When present, interactions between both stressors limited the anxiogenic effect of psychological stress on its own. In contrast, prenatal psychological stress increased anxiety-like behavior in adult males overall. A similar anxiogenic effect of IL-1β was only found on the open field test while the Stress*IL-1β interaction appeared to limit the effect of either alone. Contrarily, the PNMS effects on anxiety-like behavior on the affective exploration task were highly similar between both sexes. Analysis of males and females together revealed an additive effect of Stress and IL-1β on the number of exits from the refuge, a measure of risk assessment and thus correlated with anxiety. CONCLUSION PNMS affected offspring adult behavior in a sex-dependent manner. Effects on females were more variable, whereas psychological stress mostly induced anxiety-like behavior in males. These data highlight the sexual dimorphism in vulnerability to prenatal stressors. Maternal or stress-induced programming of the stress response and neuroinflammation may play an important role in mediating stress effects on offspring adult behavior.
Collapse
Affiliation(s)
- Barbara S E Verstraeten
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada; Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - J Keiko McCreary
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Erin A Falkenberg
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada
| | - Xin Fang
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada
| | - Steven Weyers
- Department of Human Structure and Repair, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium.
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, 4401 University Drive, Lethbridge, AB T1K 3M4, Canada.
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics and Physiology, University of Alberta, 227 HMRC, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|
22
|
Wang N, Li R, Xue M. Potential regulatory network in the PSG10P/miR-19a-3p/IL1RAP pathway is possibly involved in preeclampsia pathogenesis. J Cell Mol Med 2018; 23:852-864. [PMID: 30370628 PMCID: PMC6349174 DOI: 10.1111/jcmm.13985] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE), a pregnancy-specific disorder, is a leading cause of perinatal maternal-fetal mortality and morbidity. Impaired cell migration and invasion of trophoblastic cells and an imbalanced systemic maternal inflammatory response have been proposed as potential mechanisms of PE pathogenesis. Comparative analysis between PE placentas and normal placentas profiled differentially expressed miRNAs, lncRNAs, and mRNAs, including miR-19a-3p (miRNA), PSG10P (lncRNA), and IL1RAP (mRNA). This study was conducted to investigate their potential roles in PE pathogenesis. The expression of miR-19a-3p, PSG10P, and IL1RAP was examined in PE and normal placentas using RT-qPCR. An in vitro experiment was performed in human trophoblast HET8/SVneo and TEV-1 cells cultured in normoxic and hypoxic conditions. MiR-19a-3p targets were identified using Targetscan, miRanda, and PicTar analysis as well as luciferase reporter assays. The mouse model of PE was conducted using sFlt-1 for in vivo tests. Lower levels of miR-19a-3p, but higher levels of PSG10P and IL1RAP were observed in PE placentas and the trophoblast cells in hypoxia. Luciferase reporter assays confirmed that PSG10P and IL1RAP were both direct targets of miR-19a-3p. Exposure to hypoxia inhibited cell viability, migration, and invasion of HET8/SVneo and TEV-1 cells. Knocking out PSG10P and IL1RAP or overexpressing miR-19a-3p rescued the inhibition caused by hypoxia. In vivo experiments showed that IL1RAP promoted the expression of caspase-3, a key apoptosis enzyme, but inhibited MMP9, which is responsible for degrading the extracellular matrix, suggesting a significant role of IL1RAP in cell proliferation, migration, and invasion. miR-19a-3p, PSG10P, and IL1RAP were all found to be involved in PE pathogenesis. With a common targeting region in their sequences, a regulatory network in the PSG10P/miR-19a-3p/IL1RAP pathway may contribute to PE pathogenesis during pregnancy.
Collapse
Affiliation(s)
- Nan Wang
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ruizhen Li
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Min Xue
- Department of Obstetrics and Gynecology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
23
|
Vora B, Wang A, Kosti I, Huang H, Paranjpe I, Woodruff TJ, MacKenzie T, Sirota M. Meta-Analysis of Maternal and Fetal Transcriptomic Data Elucidates the Role of Adaptive and Innate Immunity in Preterm Birth. Front Immunol 2018; 9:993. [PMID: 29867970 PMCID: PMC5954243 DOI: 10.3389/fimmu.2018.00993] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022] Open
Abstract
Preterm birth (PTB) is the leading cause of newborn deaths around the world. Spontaneous preterm birth (sPTB) accounts for two-thirds of all PTBs; however, there remains an unmet need of detecting and preventing sPTB. Although the dysregulation of the immune system has been implicated in various studies, small sizes and irreproducibility of results have limited identification of its role. Here, we present a cross-study meta-analysis to evaluate genome-wide differential gene expression signals in sPTB. A comprehensive search of the NIH genomic database for studies related to sPTB with maternal whole blood samples resulted in data from three separate studies consisting of 339 samples. After aggregating and normalizing these transcriptomic datasets and performing a meta-analysis, we identified 210 genes that were differentially expressed in sPTB relative to term birth. These genes were enriched in immune-related pathways, showing upregulation of innate immunity and downregulation of adaptive immunity in women who delivered preterm. An additional analysis found several of these differentially expressed at mid-gestation, suggesting their potential to be clinically relevant biomarkers. Furthermore, a complementary analysis identified 473 genes differentially expressed in preterm cord blood samples. However, these genes demonstrated downregulation of the innate immune system, a stark contrast to findings using maternal blood samples. These immune-related findings were further confirmed by cell deconvolution as well as upstream transcription and cytokine regulation analyses. Overall, this study identified a strong immune signature related to sPTB as well as several potential biomarkers that could be translated to clinical use.
Collapse
Affiliation(s)
- Bianca Vora
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Aolin Wang
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Idit Kosti
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| | - Hongtai Huang
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Ishan Paranjpe
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Tracey J Woodruff
- Program on Reproductive Health and the Environment, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, United States
| | - Tippi MacKenzie
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States.,Center for Maternal-Fetal Precision Medicine, University of California San Francisco, San Francisco, CA, United States.,Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Marina Sirota
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA, United States.,Department of Pediatrics, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
24
|
Ishiguro T, Takeda J, Fang X, Bronson H, Olson DM. Interleukin (IL)-1 in rat parturition: IL-1 receptors 1 and 2 and accessory proteins abundance in pregnant rat uterus at term - regulation by progesterone. Physiol Rep 2016; 4:4/14/e12866. [PMID: 27440742 PMCID: PMC4962072 DOI: 10.14814/phy2.12866] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/13/2016] [Indexed: 11/24/2022] Open
Abstract
The role of interleukin-1 (IL-1), a pro-inflammatory cytokine, in parturition is typically noted by changes in its concentrations. Studying the expression of its receptor family, IL-1 receptor (IL-1R) 1, IL-1R2, IL-1R accessory protein (IL-1RAcP), and its predominantly brain isoform, IL-1RAcPb, during late gestation in the uterus in the Long-Evans rat is another. We assessed changes in their mRNA and protein relative abundance in the uterus and compared IL-1RAcP and IL-1RAcPb mRNA abundance in uterus, cervix, ovaries, placenta, and whole blood of Long-Evans rats during late gestation or in RU486 and progesterone-treated dams using quantitative real-time PCR and western immunoblotting. IL-1R1, IL-1RAcP, and IL-1RAcPb mRNA abundance significantly increased in the uterus at delivery whereas IL-1R2 mRNA abundance significantly decreased. IL-1R1 protein increased at term and IL-1R2 protein decreased at term compared to nonpregnant uteri. IL1-RAcPb mRNA abundance was less than IL-1RAcP, but in the lower uterine segment it was the highest of all tissues examined. RU486 stimulated preterm delivery and an increase in IL-1R1 mRNA abundance whereas progesterone administration extended pregnancy and suppressed the increase in IL-1R1. These data suggest that changes in uterine sensitivity to IL-1 occur during late gestation and suggest another level of regulation for the control of delivery. The roles for IL-1RAcP and IL-1RAcPb need to be determined, but may relate to different intracellular signaling pathways.
Collapse
Affiliation(s)
- Tomohito Ishiguro
- Departments of Obstetrics and Gynecology, Physiology & Pediatrics, University of Alberta, Edmonton, Canada Departments of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan Departments of Obstetrics and Gynecology, Koshigaya Municipal Hospital, Koshigaya, Japan
| | - Jun Takeda
- Departments of Obstetrics and Gynecology, Physiology & Pediatrics, University of Alberta, Edmonton, Canada Departments of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Xin Fang
- Departments of Obstetrics and Gynecology, Physiology & Pediatrics, University of Alberta, Edmonton, Canada
| | - Heather Bronson
- Departments of Obstetrics and Gynecology, Physiology & Pediatrics, University of Alberta, Edmonton, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Physiology & Pediatrics, University of Alberta, Edmonton, Canada
| |
Collapse
|