1
|
Kosykh AV, Tereshina MB, Gurskaya NG. Potential Role of AGR2 for Mammalian Skin Wound Healing. Int J Mol Sci 2023; 24:ijms24097895. [PMID: 37175601 PMCID: PMC10178616 DOI: 10.3390/ijms24097895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
The limited ability of mammals to regenerate has garnered significant attention, particularly in regard to skin wound healing (WH), which is a critical step for regeneration. In human adults, skin WH results in the formation of scars following injury or trauma, regardless of severity. This differs significantly from the scarless WH observed in the fetal skin of mammals or anamniotes. This review investigates the role of molecular players involved in scarless WH, which are lost or repressed in adult mammalian WH systems. Specifically, we analyze the physiological role of Anterior Gradient (AGR) family proteins at different stages of the WH regulatory network. AGR is activated in the regeneration of lower vertebrates at the stage of wound closure and, accordingly, is important for WH. Mammalian AGR2 is expressed during scarless WH in embryonic skin, while in adults, the activity of this gene is normally inhibited and is observed only in the mucous epithelium of the digestive tract, which is capable of full regeneration. The combination of AGR2 unique potencies in postnatal mammals makes it possible to consider it as a promising candidate for enhancing WH processes.
Collapse
Affiliation(s)
- Anastasiya V Kosykh
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Maria B Tereshina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Nadya G Gurskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| |
Collapse
|
2
|
Basak S, Duttaroy AK. Maternal PUFAs, Placental Epigenetics, and Their Relevance to Fetal Growth and Brain Development. Reprod Sci 2023; 30:408-427. [PMID: 35676498 DOI: 10.1007/s43032-022-00989-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/24/2022] [Indexed: 12/17/2022]
Abstract
Dietary polyunsaturated fatty acids (PUFAs), especially omega-3 (n-3) and n-6 long-chain (LC) PUFAs, are indispensable for the fetus' brain supplied by the placenta. Despite being highly unsaturated, n-3 LCPUFA-docosahexaenoic acid (DHA) plays a protective role as an antioxidant in the brain. Deficiency of DHA during fetal development may cause irreversible damages in neurodevelopment programming. Dietary PUFAs can impact placental structure and functions by regulating early placentation processes, such as angiogenesis. They promote remodeling of uteroplacental architecture to facilitate increased blood flow and surface area for nutrient exchange. The placenta's fatty acid transfer depends on the uteroplacental vascular development, ensuring adequate maternal circulatory fatty acids transport to fulfill the fetus' rapid growth and development requirements. Maternal n-3 PUFA deficiency predominantly leads to placental epigenetic changes than other fetal developing organs. A global shift in DNA methylation possibly transmits epigenetic instability in developing fetuses due to n-3 PUFA deficiency. Thus, an optimal level of maternal omega-3 (n-3) PUFAs may protect the placenta's structural and functional integrity and allow fetal growth by controlling the aberrant placental epigenetic changes. This narrative review summarizes the recent advances and underpins the roles of maternal PUFAs on the structure and functions of the placenta and their relevance to fetal growth and brain development.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India.
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
3
|
Zhou D, Xu X, Liu Y, Liu H, Cheng X, Gu Y, Xu Y, Zhu L. MiR-195-5p facilitates the proliferation, migration, and invasion of human trophoblast cells by targeting FGF2. J Obstet Gynaecol Res 2022; 48:2122-2133. [PMID: 35716001 DOI: 10.1111/jog.15298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/12/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Preeclampsia (PE), the most significant adverse exposure to cardiovascular risk during pregnancy, is one of the three major factors contributing to maternal and fetal mortality and the leading cause of preterm birth. Recently, various miRNAs have been reported to participate in PE occurrence and development. Nevertheless, the regulatory impact of miR-195-5p in PE is still indistinct. METHODS Quantitative realtime-PCR (qRT-PCR), western blot, and fluorescence in situ hybridization (FISH) assay were performed to examine miR-195-5p and FGF2 expressions in PE serum samples or HTR-8/SVneo and TEV-1 cells. CCK8, flow cytometry, wound scratch, and transwell assays were conducted to determine cell viability, cycle, apoptosis, migration, and invasion. Dual-luciferase reporter assay unveiled the relationship between miR-195-5p and FGF2. Migration-related and invasion-related protein expressions were measured by western blot assay. RESULTS miR-195-5p was prominently downregulated while FGF2 was increased in serum samples from PE patients and hypoxia-treated human trophoblast cells. FGF2 was predicted as a downstream target of miR-195-5p and targeted association was verified by dual-luciferase reporter assay. Functional experiments elaborated that miR-195-5p could facilitate trophoblast cell proliferation and metastasis but hinder cell cycle and apoptosis. Inversely, overexpressing of FGF2 could reverse the effects of miR-195-5p on trophoblast cell growth. DISCUSSION miR-195-5p was decreased in PE serum samples and cell lines, serving as a potential biomarker in protecting PE exacerbation by targeting FGF2.
Collapse
Affiliation(s)
- Dachun Zhou
- Department of Gynaecology and Obstetrics, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| | - Xiaoying Xu
- Department of Gynaecology and Obstetrics, Haian People's Hospital, Haian, Jiangsu Province, China
| | - Yuanlin Liu
- Department of Gynaecology and Obstetrics, Shanghai First Maternity and Infant Hospital, Shanghai, China.,School of Medicine, Tongji University, Shanghai, China
| | - Haiyun Liu
- Department of Gynaecology and Obstetrics, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| | - Xiaoyan Cheng
- Department of Gynaecology and Obstetrics, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| | - Yannan Gu
- Department of Gynaecology and Obstetrics, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| | - Yuanyuan Xu
- Department of Ultrasound, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| | - Lingling Zhu
- Department of Gynaecology and Obstetrics, Nantong Maternal and Child Health Care Hospital, Nantong, Jiangsu Province, China
| |
Collapse
|
4
|
Fakhr Y, Koshti S, Habibyan YB, Webster K, Hemmings DG. Tumor Necrosis Factor-α Induces a Preeclamptic-like Phenotype in Placental Villi via Sphingosine Kinase 1 Activation. Int J Mol Sci 2022; 23:ijms23073750. [PMID: 35409108 PMCID: PMC8998215 DOI: 10.3390/ijms23073750] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
Preeclampsia (PE) involves inadequate placental function. This can occur due to elevated pro-inflammatory tumor necrosis factor-α (TNF-α). In other tissues, TNF-α signals via sphingosine kinase 1 (SphK1). SphK1 hinders syncytial formation. Whether this occurs downstream of TNF-α signaling is unclear. We hypothesized that placental SphK1 levels are higher in PE and elevated TNF-α decreases syncytial function, increases syncytial shedding, and increases cytokine/factor release via SphK1 activity. Term placental biopsies were analyzed for SphK1 using immunofluorescence and qRT-PCR. Term placental explants were treated after 4 days of culture, at the start of syncytial regeneration, with TNF-α and/or SphK1 inhibitors, PF-543. Syncytialization was assessed by measuring fusion and chorionic gonadotropin release. Cell death and shedding were measured by lactate dehydrogenase release and placental alkaline phosphatase-positive shed particles. Forty-two cytokines were measured using multiplex assays. Placental SphK1 was increased in PE. Increased cell death, shedding, interferon-α2, IFN-γ-induced protein 10, fibroblast growth factor 2, and platelet-derived growth factor-AA release induced by TNF-α were reversed upon SphK1 inhibition. TNF-α increased the release of 26 cytokines independently of SphK1. TNF-α decreased IL-10 release and inhibiting SphK1 reversed this effect. Inhibiting SphK1 alone decreased TNF-α release. Hence, SphK1 partially mediates the TNF-α-induced PE placental phenotype, primarily through cell damage, shedding, and specific cytokine release.
Collapse
Affiliation(s)
- Yuliya Fakhr
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Saloni Koshti
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Yasaman Bahojb Habibyan
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Kirsten Webster
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
| | - Denise G. Hemmings
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T5H 3V9, Canada; (Y.F.); (S.K.); (Y.B.H.); (K.W.)
- Women and Children’s Health Research Institute, Edmonton, AB T6G 1C9, Canada
- Department of Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence: ; Tel.: +1-(780)-492-2098
| |
Collapse
|
5
|
Park JY, Mani S, Clair G, Olson HM, Paurus VL, Ansong CK, Blundell C, Young R, Kanter J, Gordon S, Yi AY, Mainigi M, Huh DD. A microphysiological model of human trophoblast invasion during implantation. Nat Commun 2022; 13:1252. [PMID: 35292627 PMCID: PMC8924260 DOI: 10.1038/s41467-022-28663-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Successful establishment of pregnancy requires adhesion of an embryo to the endometrium and subsequent invasion into the maternal tissue. Abnormalities in this critical process of implantation and placentation lead to many pregnancy complications. Here we present a microenigneered system to model a complex sequence of orchestrated multicellular events that plays an essential role in early pregnancy. Our implantation-on-a-chip is capable of reconstructing the three-dimensional structural organization of the maternal-fetal interface to model the invasion of specialized fetal extravillous trophoblasts into the maternal uterus. Using primary human cells isolated from clinical specimens, we demonstrate in vivo-like directional migration of extravillous trophoblasts towards a microengineered maternal vessel and their interactions with the endothelium necessary for vascular remodeling. Through parametric variation of the cellular microenvironment and proteomic analysis of microengineered tissues, we show the important role of decidualized stromal cells as a regulator of extravillous trophoblast migration. Furthermore, our study reveals previously unknown effects of pre-implantation maternal immune cells on extravillous trophoblast invasion. This work represents a significant advance in our ability to model early human pregnancy, and may enable the development of advanced in vitro platforms for basic and clinical research of human reproduction.
Collapse
Affiliation(s)
- Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Heather M Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vanessa L Paurus
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Charles K Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Cassidy Blundell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Young
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott Gordon
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Y Yi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Uchakin PN, Sakhalkar VS, Dane FC, Uchakina ON, Sheed JN, Uphouse WT, Sakhalkar OV. Azithromycin Reduces Markers of Vascular Damage in Pediatric Patients With Sickle Cell Disease. J Hematol 2021; 10:178-186. [PMID: 34527114 PMCID: PMC8425801 DOI: 10.14740/jh827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
Background Immunomodulatory effects of macrolides in chronic inflammation are well known. In this study, we tested our hypothesis that azithromycin (AZT) can decrease inflammation in pediatric patients with sickle cell disease (SCD). Methods The use of AZT as an anti-inflammatory agent was evaluated in double-blind, placebo-controlled, cross-over study for 8 weeks of treatment with 8 weeks of washout. Blood samples were collected before (PRE) and after (POST) each 8-week treatment period. Repeated measures analysis of variance (ANOVA) with post hoc multiple comparison procedures and Chi-square test were used for statistical analysis of the data. Complete blood count, distribution of the lymphocyte subsets, and plasma levels of markers of vascular damage were analyzed. Results A significant decrease in the number of leucocytes and granulocytes was observed in AZT group following treatment. An opposite dynamic was observed in placebo group; numbers of granulocytes significantly increased at POST interval. All markers of vascular damage were reduced in AZT group at POST interval with overall significance (P = 0.026). The most prominent significant changes were observed in levels of myeloid-related protein 8/14 (MRP8/14), lipocalin A (NGAL), matrix metalloproteinases (MMP) 9, and insulin-like growth factor-binding protein (IGFBP) 4. Plasma level of C-reactive protein (CRP) was significantly decreased in AZT group as well. Conclusions Data suggested that AZT may be beneficial in management of microvascular injury in SCD.
Collapse
Affiliation(s)
- Peter N Uchakin
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA.,Department of Internal Medicine, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Vishwas S Sakhalkar
- Division of Pediatric Hematology/Oncology, Mercer University School of Medicine, Beverly Knight Olson Children's Hospital and HOPE Clinic at Atrium Health-Navicent, Macon, GA 31201, USA
| | - Francis C Dane
- Department of Psychology, Radford University, Roanoke, VA 24013, USA
| | - Olga N Uchakina
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | | | | | | |
Collapse
|
7
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
8
|
Chen W, He S, Xiang D. Hypoxia-induced retinal pigment epithelium cell-derived bFGF promotes the migration and angiogenesis of HUVECs through regulating TGF-β1/smad2/3 pathway. Gene 2021; 790:145695. [PMID: 33964379 DOI: 10.1016/j.gene.2021.145695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/18/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022]
Abstract
Hypoxia promotes the secretion of basic fibroblast growth factor (bFGF) in retinal pigment epithelium (RPE), which plays an important part in retinopathy of prematurity (ROP). This study preliminarily explored the effect of hypoxia-induced RPE-derived bFGF on the biological functions of human umbilical vein endothelial cells (HUVECs). After cell culture in hypoxia conditions, the cell viability, apoptosis, and the expressions of bFGF and vascular endothelial growth factor A (VEGFA) in human RPEs were detected by 3-(4, 5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, western blot, RT-qPCR, or ELISA. The HUVECs were transfected with siRNA for bFGF (sibFGF) or transforming growth factor-β1 (TGF-β1) (siTGF-β1) and grown in the supernatant RPE under normoxia conditions or hypoxia conditions to further determine the cell viability, migration, angiogenesis, and the expressions of TGF-β1, p-smad2/3, and smad2/3 in the cells by performing MTT, transwell, tube formation, Western blot, or RT-qPCR. Hypoxia culture decreased the cell viability and promoted the apoptosis as well as the expressions of bFGF and VEGFA in RPEs. In both normoxia and hypoxia conditions, RPE-derived bFGF increased the cell viability, migration, angiogenesis, and the expressions of TGF-β1 and p-smad2/3 in the HUVECs, with hypoxia-induced RPE-derived bFGF showing a stronger effect than bFGF induced by normoxia. However, sibFGF reversed the effects caused by RPE-derived bFGF. Moreover, siTGF-β1 decreased the high cell viability, migration and angiogenesis of HUVECs, and downregulated the expressions of TGF-β1 and phosphorylated (p)-smad2/3 upregulated by hypoxia-induced RPE-derived bFGF. Hypoxia-induced RPE-derived bFGF could promote the migration and angiogenesis of HUVECs through regulating TGF-β1/smad2/3 pathway.
Collapse
Affiliation(s)
- Wensi Chen
- Department of Pediatric Ophthalmology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China
| | - Shiping He
- Department of Ophthalmology, Aier Eye Hospital, China
| | - Daoman Xiang
- Department of Pediatric Ophthalmology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, China.
| |
Collapse
|
9
|
Li X, Zhao J, Yan T, Mu J, Lin Y, Chen J, Deng H, Meng X. Cyanidin-3-O-glucoside and cisplatin inhibit proliferation and downregulate the PI3K/AKT/mTOR pathway in cervical cancer cells. J Food Sci 2021; 86:2700-2712. [PMID: 33908630 DOI: 10.1111/1750-3841.15740] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/11/2021] [Accepted: 03/24/2021] [Indexed: 12/27/2022]
Abstract
Natural compounds have been increasingly investigated as substances enhancing the effect of drugs and reducing drug-related adverse reactions. The objective of this study was to determine how a combination of cisplatin (DDP) with cyanidin-3-O-glucoside (C3G) affected malignancy features of cervical cancer cells. The results demonstrated that the proliferation of HeLa cells treated with 5 µg/ml DDP, 400 µg/ml C3G, or a combination of both (5 µg/ml DDP and 400 µg/ml C3G) was inhibited by 17.43%, 34.98%, and 63.38%, respectively. The IC50 values for DDP and the DDP/C3G combination treatments in HeLa cells were 18.53 and 6.435 µg/ml, respectively. Flow cytometry analysis indicated that treatment with DDP, C3G, or the combination induced G1 cell cycle arrest and apoptosis in HeLa cells. Furthermore, after treatment, cyclin D1 and Bcl-2 levels decreased; Bax, cleaved caspase-3, p53, and TIMP-1 were activated; and the PI3K/AKT/mTOR signaling pathway was modulated. These anticancer effects were enhanced in cells treated with the combination of DDP and C3G compared to those treated with DDP or C3G alone. Our study indicates that C3G increases the antitumor activity of DDP, suggesting a potential strategy to reduce adverse effects associated with chemotherapy in cervical cancer. PRACTICAL APPLICATION: Natural biologically active food ingredients are suggested to have a potential to enhance the effect of chemotherapy in cancer. We believe that our study makes a significant contribution to the literature because it revealed, for the first time, that C3G could increase the antitumor activity of DDP, suggesting a potential strategy to reduce adverse effects associated with chemotherapy in cervical cancer.
Collapse
Affiliation(s)
- Xu Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Jin Zhao
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Tingcai Yan
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Jingjing Mu
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Yang Lin
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Jing Chen
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Haotian Deng
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Xianjun Meng
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| |
Collapse
|
10
|
Liu E, Zhou Y, Li J, Zhang D. MicroRNA‑491‑5p inhibits trophoblast cell migration and invasion through targeting matrix metalloproteinase‑9 in preeclampsia. Mol Med Rep 2020; 22:5033-5040. [PMID: 33174053 PMCID: PMC7646938 DOI: 10.3892/mmr.2020.11604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/04/2020] [Indexed: 01/03/2023] Open
Abstract
Insufficient invasion of trophoblasts is correlated with the development of preeclampsia (PE). MicroRNA (miR)-491-5p has been reported to be implicated in human cancer cell invasion; however, whether miR-491-5p is involved in the development of PE remains largely unclear. The aim of the present study was to investigate the role of miR-491-5p in trophoblastic invasion in vitro and to determine its underlying mechanism of action. The expression levels of miR-491-5p were validated using reverse transcription-quantitative PCR. The effects of miR-491-5p on trophoblast cell invasion were evaluated in vitro. Then, the association between miR-491-5p and its downstream target was investigated in both cell lines and clinical specimens. miR-491-5p expression levels were observed to be significantly increased in the placental tissues from patients with PE. The invasive capacity of HTR-8/SVneo trophoblast cells was suppressed following the upregulation of miR-491-5p and increased following the inhibition of miR-491-5p. Matrix metalloproteinase-9 (MMP-9), a well-known regulator of trophoblast cell invasion, was discovered to be a direct target of miR-491-5p in HTR-8/SVneo trophoblast cells. Moreover, miR-491-5p expression levels were found to be inversely correlated with MMP-9 expression levels in placental tissues from patients with PE. The overexpression of MMP-9 partly attenuated the inhibitory effects of miR-491-5p on HTR-8/SVneo trophoblast cells invasion. Collectively, these findings suggested that the aberrant expression of miR-491-5p may contribute to PE through suppressing trophoblast invasion, thus highlighting the novel roles of miR-491-5p in the molecular pathogenesis of PE. The present study also showed that the miR-491-5p/MMP-9 axis may be an effective biomarker or a viable drug target for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Enling Liu
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Yuxiu Zhou
- Department of Immunity, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Jun Li
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Donghong Zhang
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
11
|
Oxidative stress and mitochondrial dysfunction in early-onset and late-onset preeclampsia. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165961. [PMID: 32916282 DOI: 10.1016/j.bbadis.2020.165961] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Preeclampsia is a pregnancy-specific syndrome with multisystem involvement which leads to foetal, neonatal, and maternal morbidity and mortality. This syndrome is characterized by the onset of clinical signs and symptoms and delivery before (early-onset preeclampsia, eoPE), or after (late-onset preeclampsia, loPE), the 34 weeks of gestation. Preeclampsia is a mitochondrial disorder where its differential involvement in eoPE and loPE is unclear. Mitochondria regulate cell metabolism and are a significant source of reactive oxygen species (ROS). The syncytiotrophoblast in eoPE and loPE show altered mitochondrial structure and function resulting in ROS overproduction, oxidative stress, and cell damage and death. Mitochondrial dysfunction in eoPE may result from altered expression of several molecules, including dynamin-related protein 1 and mitofusins, compared with loPE where these factors are either reduced or unaltered. Equally, mitochondrial fusion/fission dynamics seem differentially modulated in eoPE and loPE. It is unclear whether the electron transport chain and oxidative phosphorylation are differentially altered in these two subgroups of preeclampsia. However, the activity of complex IV (cytochrome c oxidase) and the expression of essential proteins involved in the electron transport chain are reduced, leading to lower oxidative phosphorylation and mitochondrial respiration in the preeclamptic placenta. Interventional studies in patients with preeclampsia using the coenzyme Q10, a key molecule in the electron transport chain, suggest that agents that increase the antioxidative capacity of the placenta may be protective against preeclampsia development. In this review, the mitochondrial dysfunction in both eoPE and loPE is summarized. Therapeutic approaches are discussed in the context of contributing to the understanding of mitochondrial dysfunction in eoPE and loPE.
Collapse
|
12
|
Tan Y, Qiao Y, Chen Z, Liu J, Guo Y, Tran T, Tan KS, Wang DY, Yan Y. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Front Cell Dev Biol 2020; 8:223. [PMID: 32300593 PMCID: PMC7142218 DOI: 10.3389/fcell.2020.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
The fibroblast growth factor 2 (FGF2) is a potent mitogenic factor belonging to the FGF family. It plays a role in airway remodeling associated with chronic inflammatory airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Recently, research interest has been raised in the immunomodulatory function of FGF2 in asthma and COPD, through its involvement in not only the regulation of inflammatory cells but also its participation as a mediator between immune cells and airway structural cells. Herein, this review provides the current knowledge on the biology of FGF2, its expression pattern in asthma and COPD patients, and its role as an immunomodulatory factor. The potential that FGF2 is involved in regulating inflammation indicates that FGF2 could be a therapeutic target for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Yuanyang Tan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | | | - Zhuanggui Chen
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- Department of Respiratory Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yanrong Guo
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, University Health System, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
13
|
Sheikh AQ, Saddouk FZ, Ntokou A, Mazurek R, Greif DM. Cell Autonomous and Non-cell Autonomous Regulation of SMC Progenitors in Pulmonary Hypertension. Cell Rep 2019; 23:1152-1165. [PMID: 29694892 PMCID: PMC5959296 DOI: 10.1016/j.celrep.2018.03.043] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 02/26/2018] [Accepted: 03/11/2018] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension is a devastating disease characterized by excessive vascular muscularization. We previously demonstrated primed platelet-derived growth factor receptor β+ (PDGFR-β+)/smooth muscle cell (SMC) marker+ progenitors at the muscular-unmuscular arteriole border in the normal lung, and in hypoxia-induced pulmonary hypertension, a single primed cell migrates distally and expands clonally, giving rise to most of the pathological smooth muscle coating of small arterioles. Little is known regarding the molecular mechanisms underlying this process. Herein, we show that primed cell expression of Kruppel-like factor 4 and hypoxia-inducible factor 1-α(HIF1-α) are required, respectively, for distal migration and smooth muscle expansion in a sequential manner. In addition, the HIF1-α/PDGF-B axis in endothelial cells non-cell autonomously regulates primed cell induction, proliferation, and differentiation. Finally, myeloid cells transdifferentiate into or fuse with distal arteriole SMCs during hypoxia, and Pdgfb deletion in myeloid cells attenuates pathological muscularization. Thus, primed cell autonomous and non-cell autonomous pathways are attractive therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Abdul Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Fatima Zahra Saddouk
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Aglaia Ntokou
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Renata Mazurek
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Daniel M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
14
|
Chaudhary P, Babu GS, Sobti RC, Gupta SK. HGF regulate HTR-8/SVneo trophoblastic cells migration/invasion under hypoxic conditions through increased HIF-1α expression via MAPK and PI3K pathways. J Cell Commun Signal 2019; 13:503-521. [PMID: 30684191 DOI: 10.1007/s12079-019-00505-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) is reported to be down-regulated in pregnancy complications like intrauterine growth retardation and preeclampsia, which are associated with abnormal trophoblast migration/invasion. In this study, role of HGF and associated signaling pathways has been investigated in HTR-8/SVneo trophoblastic cells migration/invasion under normoxia (20% O2) and hypoxia (2% O2). HTR-8/SVneo cells exposed to hypoxia showed increase in migration and invasion as compared to cells incubated under normoxic conditions. The migration/invasion under both normoxic and hypoxic conditions was further enhanced after treatment with HGF. Subsequent to treatment with HGF, a significant increase in expression of MMP2 & MMP3 under normoxia and MMP1 & MMP9 under hypoxia was observed. Treatment of HTR-8/SVneo cells with HGF under hypoxia also led to decrease in TIMP1. Treatment of the cells with HGF led to activation of mitogen activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways. Inhibition of MAPK by U0126 and PI3K by LY294002 led to concomitant decrease in the HGF-mediated migration/invasion of HTR-8/SVneo cells. HGF treatment under hypoxia also led to a significant increase in hypoxia inducible factor (HIF-1α) expression. Additionally, inhibition of HIF-1α by siRNA led to decrease in HGF-mediated migration of HTR-8/SVneo cells under hypoxic conditions. Inhibition of HGF activated MAPK and PI3K signaling led to reduction in HIF-1α expression under hypoxia. In conclusion, HGF facilitates HTR-8/SVneo cell migration/invasion by activation of MAPK/PI3K signaling pathways and increased expression of MMPs. HIF-1α has a role in HGF-mediated increase in migration under hypoxic conditions.
Collapse
Affiliation(s)
- Piyush Chaudhary
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.,Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Gosipatala Sunil Babu
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Ranbir Chander Sobti
- Department of Biotechnology, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Satish Kumar Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.
| |
Collapse
|
15
|
Wie JH, Ko HS, Choi SK, Park IY, Kim A, Kim HS, Shin JC. Effects of Oncostatin M on Invasion of Primary Trophoblasts under Normoxia and Hypoxia Conditions. Yonsei Med J 2018; 59:879-886. [PMID: 30091322 PMCID: PMC6082983 DOI: 10.3349/ymj.2018.59.7.879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 06/29/2018] [Accepted: 07/03/2018] [Indexed: 11/27/2022] Open
Abstract
PURPOSE To investigate the effect of oncostatin M (OSM) on protein expression levels and enzymatic activities of matrix metalloprotainase (MMP)-2 and MMP-9 in primary trophoblasts and the invasiveness thereof under normoxia and hypoxia conditions. MATERIALS AND METHODS Protein expression levels and enzymatic activities of MMP-2 and MMP-9 in primary trophoblasts under normoxia and hypoxia conditions were examined by Western blot and zymography, respectively. Effects of exogenous OSM on the in vitro invasion activity of trophoblasts according to oxygen concentration were also determined. Signal transducer and activator of transcription 3 (STAT3) siRNA was used to determine whether STAT3 activation in primary trophoblasts was involved in the effect of OSM. RESULTS OSM enhanced protein expression levels and enzymatic activities of MMP-2 and MMP-9 in term trophoblasts under hypoxia condition, compared to normoxia control (p<0.05). OSM-induced MMP-2 and MMP-9 enzymatic activities were significantly suppressed by STAT3 siRNA silencing under normoxia and hypoxia conditions (p<0.05). Hypoxia alone or OSM alone did not significantly increase the invasiveness of term trophoblasts. However, the invasion activity of term trophoblasts was significantly increased by OSM under hypoxia, compared to that without OSM treatment under normoxia. CONCLUSION OSM might be involved in the invasiveness of extravillous trophoblasts under hypoxia conditions via increasing MMP-2 and MMP-9 enzymatic activities through STAT3 signaling. Increased MMP-9 activity by OSM seems to be more important in primary trophoblasts.
Collapse
Affiliation(s)
- Jeong Ha Wie
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun Sun Ko
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sae Kyung Choi
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Yang Park
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ahyoung Kim
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Shik Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Chul Shin
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
16
|
Sánchez-Santos A, Martínez-Hernández MG, Contreras-Ramos A, Ortega-Camarillo C, Baiza-Gutman LA. Hyperglycemia-induced mouse trophoblast spreading is mediated by reactive oxygen species. Mol Reprod Dev 2018; 85:303-315. [PMID: 29392783 DOI: 10.1002/mrd.22965] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 01/26/2018] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
During embryo implantation, the outer layer of the blastocyst interacts with the endometrium giving rise to the development of the trophoblast cell lineage. The cells in this lineage participate in the penetration of endometrium due to their motility and invasive properties. The mechanisms that regulate the differentiation and invasive ability of these cells are essential for the establishment and maintenance of an efficient exchange between maternal and fetal tissues during pregnancy. In this context, hyperglycemia can induce oxidative stress causing alterations in the placenta. This study evaluated the role of reactive oxygen species (ROS) in the actions of high glucose concentration (HG) on trophoblast spreading and the expression of extracellular proteases in cultured mouse conceptuses. Blastocysts from gestational day 4 (GD4) were cultured until GD7 in HAM-F10 medium and further treated for 48 hr with HG (25 mM glucose) from GD7 to GD9. This treatment induced larger trophoblast outgrowths and increased ROS concentration, which was associated with increased expression levels of urokinase-type plasminogen activator (PLAU), plasminogen activator inhibitor 1 (PAI-1), and matrix metalloproteinase 9 (MMP-9). These effects were prevented by treatment with the non-specific antioxidant N-acetylcysteine (NAC) or apocynin, an inhibitor of NADPH oxidase. Our data suggest that the HG-induced trophoblast spreading and the expression of PLAU, PAI-1, and MMP-9 were mediated by the production of ROS via NADPH oxidase activity. Our results shed light on placental alterations in gestational diabetes mellitus.
Collapse
Affiliation(s)
- Alejandra Sánchez-Santos
- Laboratorio de Biología del Desarrollo, Unidad de Morfología y Función, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, C.P. Ciudad de México, México
| | - María G Martínez-Hernández
- Laboratorio de Biología del Desarrollo, Unidad de Morfología y Función, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Alejandra Contreras-Ramos
- Departamento de Biología del Desarrollo y Teratogénesis Experimental, Hospital Infantil de México Federico Gómez, México DF, México
| | - Clara Ortega-Camarillo
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México DF, México
| | - Luis A Baiza-Gutman
- Laboratorio de Biología del Desarrollo, Unidad de Morfología y Función, FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, C.P. Ciudad de México, México
| |
Collapse
|
17
|
Novel Regenerative Therapies Based on Regionally Induced Multipotent Stem Cells in Post-Stroke Brains: Their Origin, Characterization, and Perspective. Transl Stroke Res 2017; 8:515-528. [PMID: 28744717 DOI: 10.1007/s12975-017-0556-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/07/2017] [Accepted: 07/18/2017] [Indexed: 12/16/2022]
Abstract
Brain injuries such as ischemic stroke cause severe neural loss. Until recently, it was believed that post-ischemic areas mainly contain necrotic tissue and inflammatory cells. However, using a mouse model of cerebral infarction, we demonstrated that stem cells develop within ischemic areas. Ischemia-induced stem cells can function as neural progenitors; thus, we initially named them injury/ischemia-induced neural stem/progenitor cells (iNSPCs). However, because they differentiate into more than neural lineages, we now refer to them as ischemia-induced multipotent stem cells (iSCs). Very recently, we showed that putative iNSPCs/iSCs are present within post-stroke areas in human brains. Because iNSPCs/iSCs isolated from mouse and human ischemic tissues can differentiate into neuronal lineages in vitro, it is possible that a clearer understanding of iNSPC/iSC profiles and the molecules that regulate iNSPC/iSC fate (e.g., proliferation, differentiation, and survival) would make it possible to perform neural regeneration/repair in patients following stroke. In this article, we introduce the origin and traits of iNSPCs/iSCs based on our reports and recent viewpoints. We also discuss their possible contribution to neurogenesis through endogenous and exogenous iNSPC/iSC therapies following ischemic stroke.
Collapse
|
18
|
Menkhorst E, Winship A, Van Sinderen M, Dimitriadis E. Human extravillous trophoblast invasion: intrinsic and extrinsic regulation. Reprod Fertil Dev 2017; 28:406-15. [PMID: 25163485 DOI: 10.1071/rd14208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 12/12/2022] Open
Abstract
During the establishment of pregnancy, a human blastocyst implants into the uterine endometrium to facilitate the formation of a functional placenta. Implantation involves the blastocyst adhering to the uterine luminal epithelium before the primitive syncytiotrophoblast and subsequently specialised cells, the extravillous trophoblast (EVT), invade into the decidua in order to engraft and remodel uterine spiral arteries, creating the placental blood supply at the end of the first trimester. Defects in EVT invasion lead to abnormal placentation and thus adverse pregnancy outcomes. The local decidual environment is thought to play a key role in regulating trophoblast invasion. Here we describe the major cell types present in the decidua during the first trimester of pregnancy and review what is known about their regulation of EVT invasion. Overall, the evidence suggests that in a healthy pregnancy almost all cell types in the decidua actively promote EVT invasion and, further, that reduced EVT invasion towards the end of the first trimester is regulated, in part, by the reduced invasive capacity of EVTs shown at this time.
Collapse
Affiliation(s)
- E Menkhorst
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - A Winship
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - M Van Sinderen
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - E Dimitriadis
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| |
Collapse
|
19
|
Su MT, Tsai PY, Tsai HL, Chen YC, Kuo PL. miR-346 and miR-582-3p-regulated EG-VEGF expression and trophoblast invasion via matrix metalloproteinases 2 and 9. Biofactors 2017; 43:210-219. [PMID: 27619846 DOI: 10.1002/biof.1325] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/14/2016] [Accepted: 08/09/2016] [Indexed: 12/31/2022]
Abstract
Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an important regulator for embryo implantation and placental development, and is clinically associated with several obstetric disorders related to insufficient or inappropriate trophoblast invasion, such as recurrent abortion, preeclampsia, and intrauterine fetal growth restriction. This study was performed to identify the microRNAs targeting EG-VEGF, and evaluate the regulatory effect on trophoblast biology. miR-346 and miR-582-3p were initially identified via bioinformatic tools, and their specific binding sites on the EG-VEGF 3'UTR were further confirmed using dual luciferase and a co-transfection assays. miR-346 and miR-582-3p were demonstrated not only to suppress EG-VEGF expression, but also inhibit trophoblast invasion and migration in the JAR and HTR-8/SVneo cell lines. We further evaluated the effect of microRNAs in HTR-8/SVneo cells coexpressing EG-VEGF and miR-346 or miR-582-3p on matrix metalloproteinase (MMP 2 and MMP 9) and the tissue inhibitors of metalloproteinase (TIMP 1 and TIMP 2) using RT-PCR, western blotting and gelatin zymography. TIMP 1 and TIMP 2 were not affected by the two microRNAs, whereas the expressions and activities of MMP 2 and MMP 9 were significantly downregulated, which in turn inhibited the invasion ability of trophoblasts. In conclusion, miR-346 and miR-582-3p regulate EG-VEGF-induced trophoblast invasion through repressing MMP 2 and MMP 9, and may become novel diagnostic biomarkers or therapeutic targets for EG-VEGF-related obstetric disorders. © 2016 BioFactors, 43(2):210-219, 2017.
Collapse
Affiliation(s)
- Mei-Tsz Su
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Yin Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ling Tsai
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chi Chen
- Department of Economics, National Cheng Kung University, Tainan, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
20
|
Jiang K, Chen X, Zhao G, Wu H, Mi J, Qiu C, Peng X, Deng G. IFN-τ Plays an Anti-Inflammatory Role in Staphylococcus aureus-Induced Endometritis in Mice Through the Suppression of NF-κB Pathway and MMP9 Expression. J Interferon Cytokine Res 2017; 37:81-89. [DOI: 10.1089/jir.2016.0058] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xiuying Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Junxian Mi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
21
|
Shomer E, Katzenell S, Zipori Y, Rebibo-Sabbah A, Brenner B, Aharon A. Microvesicles of pregnant women receiving low molecular weight heparin improve trophoblast function. Thromb Res 2015; 137:141-147. [PMID: 26639203 DOI: 10.1016/j.thromres.2015.11.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Microvesicles including exosomes and microparticles, participate in the placental-maternal crosstalk in normal pregnancies and gestational vascular complications (GVC). Low molecular weight heparin (LMWH) is known to reduce the risk of placenta-mediated pregnancy complications. This study was aimed to characterize microvesicles of pregnant women receiving LMWH and explore microvesicle involvement in trophoblast and endothelial cell function. MATERIALS AND METHODS Microvesicles were isolated from blood samples obtained from non-pregnant women, healthy pregnant women (HP) and pregnant woman treated with LMWH. Microvesicle protein contents were assessed by protein array and ELISA. Microvesicle effects on early stage trophoblasts, term trophoblasts and endothelial cell migration, angiogenesis and apoptosis were evaluated. RESULTS Microvesicles derived from the group treated with LMWH contained higher levels of several proangiogenic proteins compared to those of HP women. Exposure of endothelial cells to circulating microvesicles derived from HP and LMWH treated groups induced significantly higher cell migration and branch tube formation compared to untreated cells. The effect of microvesicles from HP- and LMWH groups on early-stage trophoblast migration was similar. Microvesicles derived from these two study groups significantly decreased early-stage trophoblast apoptosis, while microvesicles derived from the HP-group (but not from the LMWH-group) significantly increased the term trophoblast apoptosis (TUNEL assay) compared to untreated cells. CONCLUSION Therapy with LMWH affects patients' microvesicle content, leading to normalization of invasion, angiogenesis activity and survival of endothelial and trophoblast cells in vitro. The effects of LMWH on microvesicles may point to an additional mechanism of heparin action in high-risk pregnancy.
Collapse
Affiliation(s)
- Einat Shomer
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Rambam Health Care Campus, Haifa, Israel
| | - Sarah Katzenell
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Rambam Health Care Campus, Haifa, Israel
| | - Yaniv Zipori
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Annie Rebibo-Sabbah
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Benjamin Brenner
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Anat Aharon
- Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
22
|
Nakagomi T, Nakano-Doi A, Kawamura M, Matsuyama T. Do Vascular Pericytes Contribute to Neurovasculogenesis in the Central Nervous System as Multipotent Vascular Stem Cells? Stem Cells Dev 2015; 24:1730-9. [PMID: 25900222 DOI: 10.1089/scd.2015.0039] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increasing evidence suggests that multipotent stem cells are harbored within a vascular niche inside various organs. Although a precise phenotype of resident vascular stem cells (VSCs) that can function as multipotent stem cells remains unclear, accumulating evidence shows that multipotent VSCs are likely vascular pericytes (PCs) that localize within blood vessels. These PCs are multipotent, possessing the ability to differentiate into various cell types, including vascular lineage cells. In addition, brain PCs are unique: They are derived from neural crest and can differentiate into neural lineage cells. Because PCs in the central nervous system (CNS) can contribute to both neurogenesis and vasculogenesis, they may mediate the reparative process of neurovascular units that are constructed by neural and vascular cells. Here, we describe the activity of PCs when viewed as multipotent VSCs, primarily regarding their neurogenic and vasculogenic potential in the CNS. We also discuss similarities between PCs and other candidates for multipotent VSCs, including perivascular mesenchymal stem cells, neural crest-derived stem cells, adventitial progenitor cells, and adipose-derived stem cells.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Akiko Nakano-Doi
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| | - Miki Kawamura
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan .,2 Department of Neurology, Osaka University Graduate School of Medicine , Osaka, Japan
| | - Tomohiro Matsuyama
- 1 Institute for Advanced Medical Sciences, Hyogo College of Medicine , Hyogo, Japan
| |
Collapse
|
23
|
Nakagomi T, Kubo S, Nakano-Doi A, Sakuma R, Lu S, Narita A, Kawahara M, Taguchi A, Matsuyama T. Brain vascular pericytes following ischemia have multipotential stem cell activity to differentiate into neural and vascular lineage cells. Stem Cells 2015; 33:1962-74. [PMID: 25694098 DOI: 10.1002/stem.1977] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 01/27/2015] [Indexed: 01/01/2023]
Abstract
Brain vascular pericytes (PCs) are a key component of the blood-brain barrier (BBB)/neurovascular unit, along with neural and endothelial cells. Besides their crucial role in maintaining the BBB, increasing evidence shows that PCs have multipotential stem cell activity. However, their multipotency has not been considered in the pathological brain, such as after an ischemic stroke. Here, we examined whether brain vascular PCs following ischemia (iPCs) have multipotential stem cell activity and differentiate into neural and vascular lineage cells to reconstruct the BBB/neurovascular unit. Using PCs extracted from ischemic regions (iPCs) from mouse brains and human brain PCs cultured under oxygen/glucose deprivation, we show that PCs developed stemness presumably through reprogramming. The iPCs revealed a complex phenotype of angioblasts, in addition to their original mesenchymal properties, and multidifferentiated into cells from both a neural and vascular lineage. These data indicate that under ischemic/hypoxic conditions, PCs can acquire multipotential stem cell activity and can differentiate into major components of the BBB/neurovascular unit. Thus, these findings support the novel concept that iPCs can contribute to both neurogenesis and vasculogenesis at the site of brain injuries.
Collapse
Affiliation(s)
| | - Shuji Kubo
- Department of Genetics, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Rika Sakuma
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Shan Lu
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan.,Department of Neurology of Hangzhou First People's Hospital, Hangzhou, People's Republic of China
| | - Aya Narita
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan
| | - Maiko Kawahara
- Institute for Advanced Medical Sciences, Nishinomiya, Hyogo, Japan.,Graduate School of Science and Technology, Kwansei Gakuin University, Sanda, Hyogo, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Hyogo, Japan
| | | |
Collapse
|
24
|
Concise Review: Are Stimulated Somatic Cells Truly Reprogrammed into an ES/iPS-Like Pluripotent State? Better Understanding by Ischemia-Induced Multipotent Stem Cells in a Mouse Model of Cerebral Infarction. Stem Cells Int 2015; 2015:630693. [PMID: 25945100 PMCID: PMC4402558 DOI: 10.1155/2015/630693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/22/2015] [Indexed: 02/07/2023] Open
Abstract
Following the discovery of pluripotent stem (PS) cells such as embryonic stem (ES) and induced pluripotent stem (iPS) cells, there has been a great hope that injured tissues can be repaired by transplantation of ES/iPS-derived various specific types of cells such as neural stem cells (NSCs). Although PS cells can be induced by ectopic expression of Yamanaka's factors, it is known that several stimuli such as ischemia/hypoxia can increase the stemness of somatic cells via reprogramming. This suggests that endogenous somatic cells acquire stemness during natural regenerative processes following injury. In this study, we describe whether somatic cells are converted into pluripotent stem cells by pathological stimuli without ectopic expression of reprogramming factors based on the findings of ischemia-induced multipotent stem cells in a mouse model of cerebral infarction.
Collapse
|
25
|
Conditioned medium of adipose-derived stromal cell culture in three-dimensional bioreactors for enhanced wound healing. J Surg Res 2015; 194:8-17. [DOI: 10.1016/j.jss.2014.10.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 10/29/2014] [Accepted: 10/30/2014] [Indexed: 01/09/2023]
|
26
|
The interplay between glucose and fatty acids on tube formation and fatty acid uptake in the first trimester trophoblast cells, HTR8/SVneo. Mol Cell Biochem 2014; 401:11-9. [DOI: 10.1007/s11010-014-2287-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/15/2014] [Indexed: 10/24/2022]
|
27
|
Jia RZ, Rui C, Li JY, Cui XW, Wang X. CDX1 restricts the invasion of HTR-8/SVneo trophoblast cells by inhibiting MMP-9 expression. Placenta 2014; 35:450-4. [DOI: 10.1016/j.placenta.2014.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 04/10/2014] [Accepted: 04/21/2014] [Indexed: 12/19/2022]
|
28
|
Hatt L, Brinch M, Singh R, Møller K, Lauridsen RH, Uldbjerg N, Huppertz B, Christensen B, Kølvraa S. Characterization of Fetal Cells from the Maternal Circulation by Microarray Gene Expression Analysis - Could the Extravillous Trophoblasts Be a Target for Future Cell-Based Non-Invasive Prenatal Diagnosis? Fetal Diagn Ther 2013; 35:218-27. [DOI: 10.1159/000356073] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/01/2013] [Indexed: 12/16/2022]
|
29
|
Lipocalin2 enhances the matrix metalloproteinase-9 activity and invasion of extravillous trophoblasts under hypoxia. Placenta 2013; 34:1036-43. [DOI: 10.1016/j.placenta.2013.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 07/24/2013] [Accepted: 08/06/2013] [Indexed: 11/21/2022]
|
30
|
Matjila M, Millar R, van der Spuy Z, Katz A. The differential expression of Kiss1, MMP9 and angiogenic regulators across the feto-maternal interface of healthy human pregnancies: implications for trophoblast invasion and vessel development. PLoS One 2013; 8:e63574. [PMID: 23696833 PMCID: PMC3656049 DOI: 10.1371/journal.pone.0063574] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 04/04/2013] [Indexed: 11/18/2022] Open
Abstract
Genes involved in invasion of trophoblast cells and angiogenesis are crucial in determining pregnancy outcome. We therefore studied expression profiles of these genes in both fetal and maternal tissues to enhance our understanding of feto-maternal dialogue. We investigated the expression of genes involved in trophoblast invasion, namely Kiss1, Kiss1 Receptor (Kiss1R) and MMP9 as well as the expression of angiogenic ligands Vascular Endothelial Growth Factor-A (VEGF-A) and Prokineticin-1 (PROK1) and their respective receptors (VEGFR1, VEGFR2 and PROK1R) across the feto-maternal interface of healthy human pregnancies. The placenta, placental bed and decidua parietalis were sampled at elective caesarean delivery. Real-time RT-PCR was used to investigate transcription, while immunohistochemistry and western blot analyses were utilized to study protein expression. We found that the expression of Kiss1 (p<0.001), Kiss1R (p<0.05) and MMP9 (p<0.01) were higher in the placenta compared to the placental bed and decidua parietalis. In contrast, the expression of VEGF-A was highest in the placental bed (p<0.001). While VEGFR1 expression was highest in the placenta (p<0.01), the expression of VEGFR2 was highest in the placental bed (p<0.001). Lastly, both PROK1 (p<0.001) and its receptor PROK1R (p<0.001) had highest expression in the placenta. Genes associated with trophoblast invasion were highly expressed in the placenta which could suggest that the influence on invasion capacity may largely be exercised at the fetal level. Furthermore, our findings on angiogenic gene expression profiles suggest that angiogenesis may be regulated by two distinct pathways with the PROK1/PROK1R system specifically mediating angiogenesis in the fetus and VEGFA/VEGFR2 ligand-receptor pair predominantly mediating maternal angiogenesis.
Collapse
Affiliation(s)
- Mushi Matjila
- MRC/UCT Receptor Biology Unit, Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Department of Obstetrics and Gynaecology, Groote Schuur Hospital, Cape Town, South Africa
- * E-mail: (MM); (AK)
| | - Robert Millar
- MRC/UCT Receptor Biology Unit, Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Mammalian Research Institute, University of Pretoria, Pretoria, South Africa
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland
| | - Zephne van der Spuy
- Department of Obstetrics and Gynaecology, Groote Schuur Hospital, Cape Town, South Africa
| | - Arieh Katz
- MRC/UCT Receptor Biology Unit, Division of Medical Biochemistry, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- * E-mail: (MM); (AK)
| |
Collapse
|
31
|
Zhang Z, Zhang L, Jia L, Cui S, Shi Y, Chang A, Zeng X, Wang P. AP-2α suppresses invasion in BeWo cells by repression of matrix metalloproteinase-2 and -9 and up-regulation of E-cadherin. Mol Cell Biochem 2013; 381:31-9. [PMID: 23660954 DOI: 10.1007/s11010-013-1685-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/02/2013] [Indexed: 11/26/2022]
Abstract
Preeclampsia complicates 5-10% of pregnancies and is a leading cause of maternal/fetal morbidity and mortality. Although the cause is unknown, the reduced migration/invasion of extravillous trophoblasts is generally regarded as a key feature of preeclampsia genesis. The present study examined the expression of activator protein-2α (AP-2α), tissue inhibitor of metalloproteinase 2 (TIMP-2), matrix metalloproteinase-2 (MMP-2), matrix metalloproteinase-9 (MMP-9), and E-cadherin in severe preeclamptic placentas and normal placentas using real-time PCR and immunohistochemistry. The expression levels of AP-2α, TIMP-2, and E-cadherin were elevated, while MMP-2 and MMP-9 levels were decreased in severe preeclamptic placentas when compared with normal placentas. To explore the underlying molecular mechanisms, BeWo cells were transfected with an AP-2α-expression construct as well as a siRNA against AP-2α. The over-expression of AP-2α decreased the invasive abilities of BeWo cells. AP-2α induction was followed by the induction of TIMP-2 and E-cadherin and a significant reduction of MMP-2 and MMP-9. Whereas in AP-2α-silencing BeWo cells, we observed the decreased expression of TIMP-2 and E-cadherin and the increased expression of MMP-2 and MMP-9. We presume that AP-2α may suppress trophoblast invasion by repression of MMP-2 and MMP-9 and up-regulation of E-cadherin, thus leading to shallow placentation in severe preeclampsia.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Choi JH, Lee HJ, Yang TH, Kim GJ. Effects of hypoxia inducible factors-1α on autophagy and invasion of trophoblasts. Clin Exp Reprod Med 2012; 39:73-80. [PMID: 22816073 PMCID: PMC3398120 DOI: 10.5653/cerm.2012.39.2.73] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Revised: 03/21/2012] [Accepted: 03/30/2012] [Indexed: 11/22/2022] Open
Abstract
Objective This study was undertaken to determine the effect of hypoxia inducible factor (HIF)-1α on the cell death, autophagy, and invasion of trophoblasts. Methods To understand the effect of HIF-1α, we inhibited HIF-1α using siRNA under normoxia and hypoxia conditions. Invasion assay and zymography were performed to determine changes in the invasion ability of HIF-1α. Western blotting and immunofluorescence were performed to determine some of the signal events involved in apoptosis and autophagy. Results There was no difference in cell death through the inhibition of HIF-1α expression by siRNA; however, the expression of LC3 and autophagosome formation increased. On the other hand, autophagy was increased, and the invasive ability of trophoblast cells decreased according to the inhibition of HIF-1α expression by siRNA. These experimental results mean that HIF-1α genes regulate the invasive ability of trophoblasts by increasing autophagy. Conclusion This study contributes important data for understanding the mechanism of early pregnancy implantation and the invasive ability of trophoblasts by defining the relationship between the roles of HIF-1α and autophagy.
Collapse
Affiliation(s)
- Jong-Ho Choi
- Department of Biomedical Science, CHA University, Seoul, Korea
| | | | | | | |
Collapse
|
33
|
Overexpression of Calreticulin in Pre-eclamptic Placentas: Effect on Apoptosis, Cell Invasion and Severity of Pre-eclampsia. Cell Biochem Biophys 2012; 63:183-9. [DOI: 10.1007/s12013-012-9350-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|