1
|
Mousavi SO, Reshi QUA, Godakumara K, Kodithuwakku S, Fazeli A. Extracellular vesicles as mediators of stress response in embryo-maternal communication. Front Cell Dev Biol 2024; 12:1440849. [PMID: 39161594 PMCID: PMC11330882 DOI: 10.3389/fcell.2024.1440849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Introduction: The pivotal role of extracellular vesicles (EVs) in facilitating effective communication between the embryo and maternal cells during the preimplantation stage of pregnancy has been extensively explored. Nonetheless, inquiries persist regarding the alterations in EV cargo from endometrial cells under stress conditions and its potential to elicit specific stress responses in trophoblast cells. Thus, the aim of this study was to elucidate the involvement of EV miRNA miRNAs in transmitting stress signals from maternal cells to trophoblasts. Methods: The receptive endometrial epithelium analogue RL95-2 cells were subjected to stress induction with 200 µM CoCl2 for 24 h before EV isolation. JAr trophoblast spheroids, which serve as embryos, were subjected to treatment with stressed or unstressed EVs derived from RL95-2 cells for 24 h. Transcriptomic alterations in the treated JAr spheroids as well as in the untreated group, as a negative control, were investigated by mRNA sequencing. Furthermore, the changes in EV miRNAs were assessed by sequencing EV samples. Results: A comprehensive analysis comparing the miRNA profiles between stressed and unstressed EVs revealed significant changes in 25 miRNAs. Furthermore, transcriptomic analysis of JAr spheroids treated with stressed RL95-2EVs versus unstressed EVs or the untreated group demonstrated 6 and 27 differentially expressed genes, respectively. Pathway enrichment analysis showed that stressed EVs induce alterations in gene expression in trophoblast cells, which is partially mediated by EV microRNAs. Discussion: Our results suggest that EVs can transfer stress signals from endometrial cells to the embryo. These discoveries shed new light on the mechanism underlying implantation failures under stress conditions. Unraveling the role of EVs in transmitting stress signals, can extend our knowledge to pave the way for targeted interventions to manage stress-related implantation failures.
Collapse
Affiliation(s)
- Seyed Omid Mousavi
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| | - Qurat Ul Ain Reshi
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| | - Suranga Kodithuwakku
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, Sri Lanka
| | - Alireza Fazeli
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
2
|
Lin RC, Chao YY, Su MT, Tsai HL, Tsai PY, Wang CY. Upregulation of miR-20b-5p inhibits trophoblast invasion by blocking autophagy in recurrent miscarriage. Cell Signal 2024; 113:110934. [PMID: 37871665 DOI: 10.1016/j.cellsig.2023.110934] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/01/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Recurrent miscarriage is defined as more than three pregnancy failures occurring before 20 weeks of gestation. Poor differentiation of the endometrial stroma or defective trophoblast cell invasion at the maternal-fetal interface leads to recurrent miscarriages. Several miRNAs, including miR-20b-5p, are aberrantly regulated in recurrent miscarriages; however, the underlying molecular mechanisms remain unclear. Primary cilia are antenna-like organelles that coordinate signaling during development and differentiation. Defective primary cilia formation leads to complications, such as recurrent miscarriage or preeclampsia. Here, we demonstrated that miR-20b-5p inhibited trophoblast cell invasion by blocking primary cilia formation. Mechanistically, miR-20b-5p targeted and inhibited ATG16L1 and ATG7 expression, thereby blocking autophagy. Defective autophagy reduced primary cilia formation and stopped ERK activation, which is a crucial signaling pathway for trophoblast invasion. Aspirin is used to prevent recurrent miscarriages in clinical settings. Treatment with aspirin inhibited miR-20b-5p levels, thus restoring primary cilia formation and trophoblast invasion. Thus, our findings uncovered the molecular mechanism by which miR-20b-5p suppressed primary cilia formation and trophoblast invasion by reducing the expression of ATG16L1 and ATG7. Moreover, we found that the defective phenotypes could be rescued by aspirin in recurrent miscarriages.
Collapse
Affiliation(s)
- Ruei-Ci Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yu-Ying Chao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Hui-Ling Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Obstetrics and Gynecology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
3
|
Li X, Chang Y, Shen W, Huang G, Hu N, Lv H, Jin M. miR-138 from ADSC Exo accelerates wound healing by targeting SIRT1/PTEN pathway to promote angiogenesis and fibrosis. Cell Signal 2023; 111:110843. [PMID: 37544635 DOI: 10.1016/j.cellsig.2023.110843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Affiliation(s)
- Xue Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuzhen Chang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Weijun Shen
- Department of Anesthesiology, Tenth People's Hospital of Tongji University, No 301 Middle Yan Chang Road, Shanghai 200072, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Nan Hu
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, 21000, Jiangsu, China.
| | - Haihong Lv
- Department of endocrinology, The First Hospital of Lanzhou University, #1 Donggang West Road Road, Lanzhou, 730000, Gansu, China.
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
4
|
Álvarez-Silvares E, Fernández-Cruz T, Bermudez-González M, Rubio-Cid P, Almeida A, Pinto E, Seoane-Pillado T, Martínez-Carballo E. Placental levels of essential and non-essential trace element in relation to neonatal weight in Northwestern Spain: application of generalized additive models. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:62566-62578. [PMID: 36943567 DOI: 10.1007/s11356-023-26560-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/16/2023] [Indexed: 05/10/2023]
Abstract
Adequate gestational progression depends to a great extent on placental development, which can modify maternal and neonatal outcomes. Any environmental toxicant, including metals, with the capacity to affect the placenta can alter the development of the pregnancy and its outcome. The objective of this study was to correlate the placenta levels of 14 essential and non-essential elements with neonatal weight. We examined relationships between placental concentrations of arsenic, cadmium, cobalt, copper, mercury, lithium, manganese, molybdenum, nickel, lead, rubidium, selenium, strontium, and zinc from 79 low obstetric risk pregnant women in Ourense (Northwestern Spain, 42°20'12.1″N 7°51.844'O) with neonatal weight. We tested associations between placental metal concentrations and neonatal weight by conducting multivariable linear regressions using generalized linear models (GLM) and generalized additive models (GAM). While placental Co (p = 0.03) and Sr (p = 0.048) concentrations were associated with higher neonatal weight, concentrations of Li (p = 0.027), Mo (p = 0.049), and Se (p = 0.02) in the placenta were associated with lower newborn weight. Our findings suggest that the concentration of some metals in the placenta may affect fetal growth.
Collapse
Affiliation(s)
- Esther Álvarez-Silvares
- Obstetrics and Gynaecology Department, Complexo Hospitalario Universitario de Ourense, C/ Ramón Puga 54, 32005, Ourense, Spain.
| | - Tania Fernández-Cruz
- Food and Health Omics, Analytical and Food Chemistry Department, Campus da Auga, Faculty of Sciences, University of Vigo, 32004, Ourense, Spain
| | - Mónica Bermudez-González
- Obstetrics and Gynaecology Department, Complexo Hospitalario Universitario de Ourense, C/ Ramón Puga 54, 32005, Ourense, Spain
| | - Paula Rubio-Cid
- Obstetrics and Gynaecology Department, Complexo Hospitalario Universitario de Ourense, C/ Ramón Puga 54, 32005, Ourense, Spain
| | - Agostinho Almeida
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Edgar Pinto
- LAQV/REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- Department of Environmental Health, School of Health, P. Porto, 4200-072, Porto, Portugal
| | | | - Elena Martínez-Carballo
- Food and Health Omics, Analytical and Food Chemistry Department, Campus da Auga, Faculty of Sciences, University of Vigo, 32004, Ourense, Spain
| |
Collapse
|
5
|
Toh P, Nicholson JL, Vetter AM, Berry MJ, Torres DJ. Selenium in Bodily Homeostasis: Hypothalamus, Hormones, and Highways of Communication. Int J Mol Sci 2022; 23:15445. [PMID: 36499772 PMCID: PMC9739294 DOI: 10.3390/ijms232315445] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
The ability of the body to maintain homeostasis requires constant communication between the brain and peripheral tissues. Different organs produce signals, often in the form of hormones, which are detected by the hypothalamus. In response, the hypothalamus alters its regulation of bodily processes, which is achieved through its own pathways of hormonal communication. The generation and transmission of the molecules involved in these bi-directional axes can be affected by redox balance. The essential trace element selenium is known to influence numerous physiological processes, including energy homeostasis, through its various redox functions. Selenium must be obtained through the diet and is used to synthesize selenoproteins, a family of proteins with mainly antioxidant functions. Alterations in selenium status have been correlated with homeostatic disturbances in humans and studies with animal models of selenoprotein dysfunction indicate a strong influence on energy balance. The relationship between selenium and energy metabolism is complicated, however, as selenium has been shown to participate in multiple levels of homeostatic communication. This review discusses the role of selenium in the various pathways of communication between the body and the brain that are essential for maintaining homeostasis.
Collapse
Affiliation(s)
- Pamela Toh
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Jessica L. Nicholson
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Alyssa M. Vetter
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
- School of Human Nutrition, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marla J. Berry
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Daniel J. Torres
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| |
Collapse
|
6
|
Cheng W, Zhang L, Sa P, Luo J, Li M. Transcriptomic analysis reveals the effects of maternal selenium deficiency on placental transport, hormone synthesis, and immune response in mice. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6674774. [PMID: 36002020 DOI: 10.1093/mtomcs/mfac062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/19/2022] [Indexed: 11/14/2022]
Abstract
Selenium deficiency has been considered to increase the risk of gestational complications. Our previous work showed that maternal selenium deficiency suppressed proliferation, induced autophagy dysfunction and apoptosis in the placenta of mice. However, other effects of maternal selenium deficiency on the placenta and the underlying mechanisms remain unclear. In the present study, dietary selenium deficiency in dams significantly suppressed glutathione peroxidase (GSH-Px) activity, total antioxidant capacity (T-AOC), and increased malondialdehyde (MDA) content in the placentae, confirming the oxidative stress in the placenta. By transcriptome sequencing analysis, the DEGs were involved in many biological processes, including ion transport, lipid metabolic process, immune response, transmembrane transport, and others. According to the KEGG analysis, the DEGs were primarily enriched in metabolic pathways, PI3K-Akt signaling pathway, and others. Among these, the steroid hormone biosynthesis pathway enriched the most DEGs. Hsd3b1, an ER enzyme involved in progesterone synthesis, was validated downregulated. Consistently, the progesterone content in the serum of the selenium-deficient group was decreased. Ion transporters and transmembrane transporters, such as Heph, Trf, Slc39a8, Slc23a1, Atp7b, and Kcnc1, were reduced in the selenium-deficient placentae. Immune response-related genes, including Ccl3, Ccl8, Cxcl10, and Cxcl14, were increased in the selenium-deficient placentae, along with an increase in macrophage number. These results suggested that maternal selenium deficiency may impair progesterone biosynthesis, reduce nutrient transporters expression, and promote immune response by increasing the oxidative stress of the placentae. This present study provides a novel insight into the possible cause of placenta disorder during pregnancy.
Collapse
Affiliation(s)
- Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peiyue Sa
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jing Luo
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
7
|
BushenHuoxue Recipe for the Treatment of Prethrombotic State of ACA-Positive Recurrent Miscarriage via the Regulation of the PI3K-AKT Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2385534. [PMID: 35198031 PMCID: PMC8860511 DOI: 10.1155/2022/2385534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022]
Abstract
Background Although the Bushen Huoxue (BSHX) recipe is commonly used for the effective treatment of the prethrombotic state of recurrent abortions, its mechanism of action is unclear. In this article, we investigated the therapeutic effects of BSHX on anti-cardiolipin antibody (ACA) positive recurrent miscarriage mice and the molecular mechanism involved in the treatment of the prethrombotic state of ACA-positive recurrent miscarriages based on the PI3K-Akt signaling pathway, to provide a scientific basis for clinical practice. Methods An ACA-positive recurrent miscarriage mouse model and normal pregnancy mouse model were adopted in this experiment. Seventy CBA/J female mice were induced to establish the ACA-positive recurrent model; the mice were mated with DBA/2 male mice. Of these mice, 50 became pregnant, which were randomly divided into a BSHX high-dose group (BH, 2.52 g/kg), BSHX medium-dose group (BM, 1.26 g/kg), BSHX low-dose group (BL, 0.63 g/kg), model group (M, distilled water), and an aspirin enteric-coated tablet group; each group had 10 mice. In addition, 16 CBA/J female mice were induced to establish the normal pregnant mouse model; the mice were mated with BALB/C male mice. Of these mice, 10 became pregnant, which were used as the blank control group (C) and received distilled water by gavage. Stillbirth and abortion rates were recorded for each group, and the uterine tissue, urine, and serum were collected. The serum expression levels of ACA, interleukin-6 (IL-6), progesterone ,estradiol, and endometrial histological changes were compared between the groups. Metabolomics was performed on the urine and uterine tissues of both groups using UHPLC-QTOF/MS, and the expression levels of PI3K, p-PI3K, AKT, and p-AKT proteins in the uterine tissues were detected using Western blot. Results Compared with the model pregnancy group, the BSHX high-dose group, BSHX medium-dose group, and BSHX low-dose group all had a lower absorption rate of mouse embryos, improved uterine histopathological morphology, significantly reduced serum levels of ACA and IL-6, increased serum levels of progesterone and estradiol, and significantly upregulated uterine levels of p-AKT, PI3K, and p-PI3K proteins. The metabolomic results showed that the metabolic levels in the urine and uterine tissues were significantly altered in the mouse model of ACA-positive recurrent abortion. The results also suggested that the pathogenesis of ACA-positive recurrent abortion may be associated with metabolic pathways, such as pentose, glucuronide, lysine degradation, and steroid hormone biosynthesis. Conclusion The BSHX recipe improved the uterine histopathological morphology of pregnant mice and promoted vascular formation in uterine tissues. The mechanisms involved the reduction in serum ACA and IL-6 levels, the increment in serumprogesterone and estradiol levels, the upregulation of the levels of p-AKT, PI3K, and p-PI3K proteins, and the activation of the PI3K-Akt signaling pathway. These data will be useful for effective drug research and development.
Collapse
|
8
|
Li M, Cheng W, Zhang L. Maternal selenium deficiency suppresses proliferation, induces autophagy dysfunction and apoptosis in the placenta of mice. Metallomics 2021; 13:6406492. [PMID: 34669944 DOI: 10.1093/mtomcs/mfab058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022]
Abstract
Selenium deficiency is thought to be associated with the occurrence of gestational complications. However, the underlying mechanism of selenium deficiency impairs placental function remains unclear. In this study, female mice were separately supplemented with a Se-deficient (0.02 mg/kg Se) or control diet (0.2 mg/kg Se) for 12 weeks before mating and throughout gestation. Maternal liver and placentas were collected at embryonic day 15.5 and analyzed for Se content. Oxidative stress status, proliferation capability, autophagy, and apoptosis of the placenta were determined. We found that maternal selenium deficiency decreased placental Se concentration and some antioxidant selenoproteins expressions. The concentrations of catalase and glutathione in selenium-deficient placentas were reduced, along with an increase in hydrogen peroxide (H2O2) content. Selenium deficiency inhibited the expression of proliferating cell nuclear antigen. Autophagosomes, autophagolysosomes, and upregulation of autophagy-related protein microtubule-associated protein 1 light chain 3 alpha II (LC3B), Beclin1, PTEN-induced putative kinase 1 (PINK1), and Parkin were found in the selenium-deficient trophoblasts. Autophagic substrate p62/sequestosome 1 was surprisingly increased, indicating autophagy flux dysfunction. Selenium deficiency increased expressions of B cell leukemia/lymphoma 2 associated X protein (Bax), cleaved caspase-9/-3, and decreased the B cell leukemia/lymphoma 2 (Bcl2) level. Moreover, typical apoptotic ultrastructure and apoptosis-positive cells were observed in the selenium-deficient placenta. Our results suggested that maternal selenium deficiency impaired placental proliferation, induced autophagy dysfunction and apoptosis via increasing oxidative stress, and the Akt/mechanistic target of rapamycin (mTOR) pathway involved in this process. This study revealed a novel mechanism by which maternal selenium deficiency caused impairment of the placenta.
Collapse
Affiliation(s)
- Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
9
|
Ma Y, Wu H, Liang X, Zhang C, Ma Y, Wei Y, Li J, Chen H. Identification of downstream targets and signaling pathways of long non-coding RNA NR_002794 in human trophoblast cells. Bioengineered 2021; 12:6617-6628. [PMID: 34516352 PMCID: PMC8806843 DOI: 10.1080/21655979.2021.1974808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Preeclampsia (PE) is a huge threat to pregnant women. Our previous study demonstrated that long non-coding RNA (lncRNA) NR_002794 was highly expressed in placentas of PE patients and could regulate the phenotypes of trophoblast cells. However, the downstream regulatory mechanisms of NR_002794 remain unknown. In this text, some potential downstream targets or signaling pathways of NR_002794 were identified through RNA sequencing (RNA-seq) and bioinformatics analysis in SWAN71 trophoblast cells. Western blot assay demonstrated that NR_002794 inactivated protein kinase B (AKT) and extracellular signal-regulated kinase 1/2 (ERK1/2) pathways and activated cell apoptotic signaling in SWAN71 cells. Both RNA-seq and reverse transcription-quantitative PCR (RT-qPCR) outcomes showed that NR_002794 up-regulation could notably inhibit the expression of C-C motif chemokine ligand 4 like 2 (CCL4L2), interleukin 15 receptor subunit alpha (IL15RA), interleukin 32 (IL32), and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), while NR_002794 knockdown induced these gene expressions in SWAN71 cells. CCK-8, BrdU, Transwell, wound healing, and flow cytometry analyses showed that NR_002794 inhibited cell proliferation and migration and induced cell apoptosis through down-regulating TIE1 in SWAN71 cells. In conclusion, lncRNA NR_002794 could exert its functions by regulating AKT and ERK1/2 pathways and TIE1 expression in human trophoblast cells.
Collapse
Affiliation(s)
- Yinyao Ma
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Hua Wu
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Xuxia Liang
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Chun Zhang
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Yanhua Ma
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Yanfen Wei
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Jing Li
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| | - Hui Chen
- Department of Obstetrics, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, P.R. China
| |
Collapse
|
10
|
Adu-Gyamfi EA, Lamptey J, Chen XM, Li FF, Li C, Ruan LL, Yang XN, Liu TH, Wang YX, Ding YB. Iodothyronine deiodinase 2 (DiO 2) regulates trophoblast cell line cycle, invasion and apoptosis; and its downregulation is associated with early recurrent miscarriage. Placenta 2021; 111:54-68. [PMID: 34166926 DOI: 10.1016/j.placenta.2021.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/26/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Trophoblast development is a crucial event in placentation and pregnancy complications but its underlying mechanisms remain unclear. Thus, we aimed at investigating the role of DiO2 in trophoblast cell line decisions and assessing its placental villous expression in early recurrent miscarriage (ERM) patients. METHODS The placental villous expression of DiO2 was determined with immunofluorescence. Cell proliferation was measured with the CCK8 kit while cell-cycle and apoptosis were studied with flow-cytometry. Cell migration and invasion were measured with wound-healing and transwell assays, respectively. Gene expression was then assessed with RT-qPCR and western blotting. RESULTS DiO2 is expressed in the CTB, PCT, DCT and STB of the placenta. Its overexpression arrested trophoblast cell line proliferation at the G1 phase of the cell-cycle by downregulating cyclin-D1 and PCNA, while promoting apoptosis via increased caspase-3 activity and inhibition of the AKT and ERK1/2 signaling pathways. Also, it augmented trophoblast cell line migration and invasion via the upregulation of N-cadherin, vimentin, fascin-1, twist-1 and other epithelial-mesenchymal transition genes. DiO2 knockdown elicited the opposite effects. Surprisingly, each of these effects of DiO2 manipulation was not mediated by thyroid hormone metabolism. Assessment of the ERM placental villi revealed a downregulation of DiO2, N-cadherin, vimentin, fascin-1 and twist-1. The expression of E-cadherin remained unchanged in these placentae. DISCUSSION During placentation, DiO2 may inhibit trophoblast proliferation while facilitating their differentiation into an invasive phenotype; and that its downregulation may contribute to the shallow trophoblast invasion that precedes ERM. Hence, DiO2 is a potential therapeutic target against ERM.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Genetics, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Jones Lamptey
- Department of Genetics, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xue-Mei Chen
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fang-Fang Li
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Cong Li
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Ling-Ling Ruan
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xue-Niu Yang
- First Affiliated Hospital of Chongqing Medical University, Chongqing, 400020, People's Republic of China
| | - Tai-Hang Liu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Ying-Xiong Wang
- Department of Genetics, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Yu-Bin Ding
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, 400016, People's Republic of China; Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
11
|
Hu XQ, Zhang L. Hypoxia and Mitochondrial Dysfunction in Pregnancy Complications. Antioxidants (Basel) 2021; 10:antiox10030405. [PMID: 33800426 PMCID: PMC7999178 DOI: 10.3390/antiox10030405] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Hypoxia is a common and severe stress to an organism's homeostatic mechanisms, and hypoxia during gestation is associated with significantly increased incidence of maternal complications of preeclampsia, adversely impacting on the fetal development and subsequent risk for cardiovascular and metabolic disease. Human and animal studies have revealed a causative role of increased uterine vascular resistance and placental hypoxia in preeclampsia and fetal/intrauterine growth restriction (FGR/IUGR) associated with gestational hypoxia. Gestational hypoxia has a major effect on mitochondria of uteroplacental cells to overproduce reactive oxygen species (ROS), leading to oxidative stress. Excess mitochondrial ROS in turn cause uteroplacental dysfunction by damaging cellular macromolecules, which underlies the pathogenesis of preeclampsia and FGR. In this article, we review the current understanding of hypoxia-induced mitochondrial ROS and their role in placental dysfunction and the pathogenesis of pregnancy complications. In addition, therapeutic approaches selectively targeting mitochondrial ROS in the placental cells are discussed.
Collapse
|
12
|
Abstract
Selenium (Se), an essential trace element, is inserted as selenocysteine into an array of functional proteins and forms the core of various enzymes that play a cardinal role in antioxidant defense mechanisms, in redox regulation, and in thyroid hormone metabolism. Variations in plasma Se are due to nutritional habits, geographic and ethnic differences, and probably to genetic polymorphisms, the latter still to be conclusively established. Se concentrations were reported to be low in women of reproductive age in the UK, decreasing further during pregnancy, this resulting in low plasma and placental antioxidant enzyme activities. Since low serum Se levels have been found in women with preeclampsia, it has been hypothesized that low maternal Se status during early gestation may be an indicator of preterm birth. Moreover, it is documented that Se administration during pregnancy tendentially reduced the markers of thyroid autoimmunity and the incidence of maternal hypothyroidism in the postpartum period. Importantly, low Se levels in pregnant women affect fetal growth and augment the risk of delivering a small-for-gestational age infant by reducing placental antioxidant defense, while low Se in the third trimester is thought to indicate increased demands by the placenta, an issue which requires further confirmation. There is evidently a need for double-blind, placebo-controlled studies to better determine the efficacy and safety of Se supplementation in pregnancy at high risk for complications, and for measurement of Se levels or of selenoprotein P, the most reliable parameter of Se status, particularly in selenopenic regions.
Collapse
Affiliation(s)
- Leonidas H Duntas
- Evgenideion Hospital, Unit of Endocrinology, Metabolism and Diabetes, Thyroid Section, University of Athens, 20 Papadiamantopoulou Str, 11528 Athens, Greece
| |
Collapse
|
13
|
Stanishevska NV. Selenoproteins and their emerging roles in signaling pathways. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The functional activity of selenoproteins has a wide range of effects on complex pathogenetic processes, including teratogenesis, immuno-inflammatory, neurodegenerative. Being active participants and promoters of many signaling pathways, selenoproteins support the lively interest of a wide scientific community. This review is devoted to the analysis of recent data describing the participation of selenoproteins in various molecular interactions mediating important signaling pathways. Data processing was carried out by the method of complex analysis. For convenience, all selenoproteins were divided into groups depending on their location and function. Among the group of selenoproteins of the ER membrane, selenoprotein N affects the absorption of Ca2+ by the endoplasmic reticulum mediated by oxidoreductin (ERO1), a key player in the CHOP/ERO1 branch, a pathogenic mechanism that causes myopathy. Another selenoprotein of the ER membrane selenoprotein K binding to the DHHC6 protein affects the IP3R receptor that regulates Ca2+ flux. Selenoprotein K is able to affect another protein of the endoplasmic reticulum CHERP, also appearing in Ca2+ transport. Selenoprotein S, associated with the lumen of ER, is able to influence the VCP protein, which ensures the incorporation of selenoprotein K into the ER membrane. Selenoprotein M, as an ER lumen protein, affects the phosphorylation of STAT3 by leptin, which confirms that Sel M is a positive regulator of leptin signaling. Selenoprotein S also related to luminal selenoproteins ER is a modulator of the IRE1α-sXBP1 signaling pathway. Nuclear selenoprotein H will directly affect the suppressor of malignant tumours, p53 protein, the activation of which increases with Sel H deficiency. The same selenoprotein is involved in redox regulation. Among the cytoplasmic selenoproteins, abundant investigations are devoted to SelP, which affects the PI3K/Akt/Erk signaling pathway during ischemia/reperfusion, is transported into the myoblasts through the plasmalemma after binding to the apoER2 receptor, and into the neurons to the megaline receptor and in general, selenoprotein P plays the role of a pool that stores the necessary trace element and releases it, if necessary, for vital selenoproteins. The thioredoxin reductase family plays a key role in the invasion and metastasis of salivary adenoid cystic carcinoma through the influence on the TGF-β-Akt/GSK-3β pathway during epithelial-mesenchymal transition. The deletion of thioredoxin reductase 1 affects the levels of messengers of the Wnt/β-catenin signaling pathway. No less studied is the glutathione peroxidase group, of which GPX3 is able to inhibit signaling in the Wnt/β-catenin pathway and thereby inhibit thyroid metastasis, as well as suppress protein levels in the PI3K/Akt/c-fos pathway. A key observation is that in cases of carcinogenesis, a decrease in GPX3 and its hypermethylation are almost always found. Among deiodinases, deiodinase 3 acts as a promoter of the oncogenes BRAF, MEK or p38, while stimulating a decrease in the expression of cyclin D1. The dependence of the level of deiodinase 3 on the Hedgehog (SHH) signaling pathway is also noted. Methionine sulfoxide reductase A can compete for the uptake of ubiquitin, reduce p38, JNK and ERK promoters of the MAPK signaling pathway; methionine sulfoxide reductase B1 suppresses MAPK signaling messengers, and also increases PARP and caspase 3.
Collapse
|
14
|
Chen Y, Ding H, Wei M, Zha W, Guan S, Liu N, Li Y, Tan Y, Wang Y, Wu F. MSC-Secreted Exosomal H19 Promotes Trophoblast Cell Invasion and Migration by Downregulating let-7b and Upregulating FOXO1. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:1237-1249. [PMID: 32069774 PMCID: PMC7026285 DOI: 10.1016/j.omtn.2019.11.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/13/2019] [Accepted: 11/24/2019] [Indexed: 12/12/2022]
Abstract
Exosomes perform important functions for intercellular communication through extracellular signaling pathways, leading to the regulation of important biological processes, including cell proliferation, but also systemic dysfunctions such as preeclampsia (PE). However, the inhibitory effects of mesenchymal stem cell (MSCs)-derived exosomes in PE remain largely unknown. Thus, we assessed the possibility that exosomes could transport long non-coding RNA H19 and the correlation between H19 and the apoptosis of trophoblast cells. The expression of microRNA let-7b and forkhead box protein O1 (FOXO1) was characterized in placental tissues of PE patients. Gain- and loss-of-function experiments were performed to examine the roles of FOXO1 and let-7b in trophoblast cells. Interactions between let-7b and H19 as well as between let-7b and FOXO1 were confirmed by a dual-luciferase reporter assay, RNA pull-down, and RNA immunoprecipitation. HTR-8/SVneo cells were co-cultured with exosomes derived from MSCs overexpressing H19, followed by invasion, migration, and apoptosis assessments of trophoblast cells. We found that let-7b was highly expressed and FOXO1 was poorly expressed in placental tissues of PE patients. Furthermore, H19 acts as a competitive endogenous RNA against let-7b, and let-7b directly targeted FOXO1. Moreover, H19 could be transferred to trophoblast cells via MSC-secreted exosomes. MSC-derived exosomes overexpressing H19 decreased let-7b, increased FOXO1, and activated the protein kinase B (AKT) signaling pathway, thus increasing invasion and migration and inhibiting apoptosis of trophoblast cells. These results suggest that MSC-derived exosomes overexpressing H19 may be a novel direction for therapeutic strategies against PE.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Haiyan Ding
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Min Wei
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wenhui Zha
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Shuang Guan
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ning Liu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yang Li
- Center of Reproductive Medicine, Center of Prenatal Diagnosis, The First Hospital of JiLin University, Changchun 130041, P.R. China
| | - Yuan Tan
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yan Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fuju Wu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China.
| |
Collapse
|
15
|
Long non-coding RNA MALAT1 enhances the apoptosis of cardiomyocytes through autophagy inhibition by regulating TSC2-mTOR signaling. Biol Res 2019; 52:58. [PMID: 31783925 PMCID: PMC6883637 DOI: 10.1186/s40659-019-0265-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Our previous study showed that knockdown of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) attenuated myocardial apoptosis in mouse acute myocardial infarction (AMI). This study aims to explore whether MALAT1 enhanced cardiomyocyte apoptosis via autophagy regulation and the underlying mechanisms of MALAT1 regulating autophagy. METHODS Cardiomyocytes were isolated from neonatal mice and then stimulated with hypoxia/reoxygenation (H/R) injury to mimic AMI. The autophagy level was assessed using GFP-LC3 immunofluorescence and western blot analysis of autophagy-related proteins. RNA pull-down and RNA immunoprecipitation (RIP) was performed to analyze the binding of MALAT1 and EZH2. Chromatin immunoprecipitation (ChIP) assay was performed to analyze the binding of TSC2 promoter and EZH2. The cell apoptosis was evaluated using TUNEL staining and western blot analysis of apoptosis-related proteins. RESULTS H/R injury increased MALAT1 expression in cardiomyocytes. Furthermore, MALAT1 overexpression inhibited, whereas MALAT1 knockdown enhanced the autophagy of cardiomyocytes. Moreover, MALAT1 overexpression recruited EZH2 to TSC2 promoter regions to elevate H3K27me3 and epigenetically inhibited TSC2 transcription. Importantly, TSC2 overexpression suppressed mTOR signaling and then activated the autophagy. Further results showed that MALAT1 inhibited proliferation and enhanced apoptosis of cardiomyocytes through inhibiting TSC2 and autophagy. CONCLUSION These findings demonstrate that the increased MALAT1 expression induced by H/R injury enhances cardiomyocyte apoptosis through autophagy inhibition by regulating TSC2-mTOR signaling.
Collapse
|
16
|
Hallman M, Haapalainen A, Huusko JM, Karjalainen MK, Zhang G, Muglia LJ, Rämet M. Spontaneous premature birth as a target of genomic research. Pediatr Res 2019; 85:422-431. [PMID: 30353040 DOI: 10.1038/s41390-018-0180-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 01/23/2023]
Abstract
Spontaneous preterm birth is a serious and common pregnancy complication associated with hormonal dysregulation, infection, inflammation, immunity, rupture of fetal membranes, stress, bleeding, and uterine distention. Heredity is 25-40% and mostly involves the maternal genome, with contribution of the fetal genome. Significant discoveries of candidate genes by genome-wide studies and confirmation in independent replicate populations serve as signposts for further research. The main task is to define the candidate genes, their roles, localization, regulation, and the associated pathways that influence the onset of human labor. Genomic research has identified some candidate genes that involve growth, differentiation, endocrine function, immunity, and other defense functions. For example, selenocysteine-specific elongation factor (EEFSEC) influences synthesis of selenoproteins. WNT4 regulates decidualization, while a heat-shock protein family A (HSP70) member 1 like, HSPAIL, influences expression of glucocorticoid receptor and WNT4. Programming of pregnancy duration starts before pregnancy and during placentation. Future goals are to understand the interactive regulation of the pathways in order to define the clocks that influence the risk of prematurity and the duration of pregnancy. Premature birth has a great impact on the duration and the quality of life. Intensification of focused research on causes, prediction and prevention of prematurity is justified.
Collapse
Affiliation(s)
- Mikko Hallman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.
| | - Antti Haapalainen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Johanna M Huusko
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, OH, USA
| | - Minna K Karjalainen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Ge Zhang
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, OH, USA
| | - Louis J Muglia
- Division of Human Genetics, Center for Prevention of Preterm Birth, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, March of Dimes Prematurity Research Center Ohio Collaborative, Cincinnati, OH, USA
| | - Mika Rämet
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
17
|
Wang Z, Li R, Zhong R. Extracellular matrix promotes proliferation, migration and adhesion of airway smooth muscle cells in a rat model of chronic obstructive pulmonary disease via upregulation of the PI3K/AKT signaling pathway. Mol Med Rep 2018; 18:3143-3152. [PMID: 30066869 PMCID: PMC6102654 DOI: 10.3892/mmr.2018.9320] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrix (ECM) creates the tissue microenvironment and serves a role in airway wall remodeling in chronic obstructive pulmonary disease (COPD). However, the biological function of ECM in COPD remains to be elucidated. In the present study, 24 healthy Sprague Dawley rats were randomized to normal and COPD groups. COPD was established by intratracheal injection with lipopolysaccharide over 30 days. Subsequently, airway smooth muscle cells (ASMCs) were isolated from rats and served as a model to assess the effects of three ECM components, including collagen type I, laminin and collagen type III (COL‑3). Functional analysis in vitro, using cell counting kit‑8, flow cytometry, wound healing and cell adhesion assays indicated that the ECM components could promote cell proliferation, cell cycle progression, migration and adhesion ability, respectively. Furthermore, as demonstrated by ELISA, treatment with ECM components increased levels of C‑X‑C motif chemokine ligand 1 (CXCL1), CXCL8 and interleukin‑6 in ASMCs. Expression of transforming growth factor β1 (TGFβ1), fibroblast growth factor‑1 (FGF‑1) and tissue inhibitor of metalloproteinase 1 (TIMP1) was increased, and expression of matrix metalloproteinase‑9 (MMP‑9) was decreased following treatment with ECM components, as demonstrated by reverse transcription‑quantitative polymerase chain reaction and western blot analysis. Additionally, specific activation of phosphoinositide 3‑kinase (PI3K) signaling, using insulin‑like growth factor‑1 (IGF‑1), promoted cell proliferation and cell cycle progression, increased expression of TGFβ1, FGF‑1, PI3K, AKT, phospho‑AKT, serine/threonine‑protein kinase mTOR (mTOR), phospho‑mTOR and TIMP1, promoted cell migration capacity and reduced the expression level of MMP‑9 in cells from COPD rats. Consistently, PI3K inhibitor LY294002 exerted the opposite effect to IGF‑1. In conclusion, ECM proteins promoted proliferation, migration and adhesion of ASMCs form rat models of COPD through activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zhengyan Wang
- Department of Respiratory Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Rui Li
- Department of Orthopedics, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| | - Rui Zhong
- Second Affiliated Hospital of Hubei University of Medicine, Suizhou, Hubei 442000, P.R. China
| |
Collapse
|
18
|
The Free Radical Diseases of Prematurity: From Cellular Mechanisms to Bedside. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7483062. [PMID: 30140369 PMCID: PMC6081521 DOI: 10.1155/2018/7483062] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/28/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
During the perinatal period, free radicals (FRs) are involved in several physiological roles such as the cellular responses to noxia, the defense against infectious agents, the regulation of cellular signaling function, and the induction of a mitogenic response. However, the overproduction of FRs and the insufficiency of an antioxidant mechanism result in oxidative stress (OS) which represents a deleterious process and an important mediator of damage to the placenta and the developing fetus. After birth, OS can be magnified by other predisposing conditions such as hypoxia, hyperoxia, ischemia, hypoxia ischemia-reperfusion, inflammation, and high levels of nonprotein-bound iron. Newborns are particularly susceptible to OS and oxidative damage due to the increased generation of FRs and the lack of adequate antioxidant protection. This impairment of the oxidative balance has been thought to be the common factor of the so-called “free radical related diseases of prematurity,” including retinopathy of prematurity, bronchopulmonary dysplasia, intraventricular hemorrhage, periventricular leukomalacia, necrotizing enterocolitis, kidney damage, and oxidative hemolysis. In this review, we provide an update focused on the factors influencing these diseases refining the knowledge about the role of OS in their pathogenesis and the current evidences of such relationship. Mechanisms governing FR formation and subsequent OS may represent targets for counteracting tissue damage.
Collapse
|
19
|
Wang W, Bai G, Zhang Y, Zhang T, Li C, Yuan Y, Liu S, Wang C. HBxAg suppresses cell apoptosis and promotes the secretion of placental hormones in human placental trophoblasts via activation of the EGFR/Akt pathway. Cell Biol Int 2017; 42:237-247. [PMID: 29052908 DOI: 10.1002/cbin.10891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 10/07/2017] [Indexed: 12/11/2022]
Abstract
The aim of this study is to investigate the role of Hepatitis B virus x (HBx) in the growth and secretion of human placental trophoblasts. Firstly, placenta tissues were collected from pregnant HBV carriers with various viral loads. The results of immunohistochemical technique showed that the HBx protein and pEGFR protein levels were both markedly increased with the viral load elevation. Then, a placental trophoblast cell strain (JEG-3-HBx), which stably expressed HBx mRNA and protein, was established with the pcDNA-HBx transfection followed by the G418 selection. The JEG-3-HBx strain displayed distinct activation of the EGFR/AKT pathway, a lower level of cell apoptosis, and higher secretion levels of placental hormones, including human chorionic gonadotropin (hCG), progesterone, estrogen and β-endorphin. Subsequently, HBx siRNA was used to silence the HBx gene in the JEG-3-HBx strain. Our data showed that the HBx siRNA transfection markedly suppressed the activation of the EGFR/AKT pathway, promoted cell apoptosis, and reduced the secretion of the placental hormones. Finally, EGF was applied to simulate the JEG-3-HBx strain with or without the HBx siRNA transfection. EGF treatment counteracted the reduction of cell apoptosis and the suppression of hormone secretion caused by HBx siRNA in the cell strain. In conclusion, the pEGFR protein was robustly upregulated in HBx-infected human placenta tissues and trophoblast cells. HBx reduces cell apoptosis and promotes the secretion of placental hormones in human placental trophoblast cells via activation of the EGFR/Akt pathway.
Collapse
Affiliation(s)
- Weimin Wang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Guiqin Bai
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Yuting Zhang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Ting Zhang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Chen Li
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Yongxing Yuan
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Sixue Liu
- College of Life Science and Technology, Xi'an Jiaotong University, Xi'an City, China
| | - Caili Wang
- Department of Gynecology and Obstetrics, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| |
Collapse
|