1
|
Liu Q, Jiang M, Lu X, Hong J, Sun Y, Yang C, Chen Y, Chai X, Tang H, Liu X. Prenatal triphenyl phosphate exposure impairs placentation and induces preeclampsia-like symptoms in mice. ENVIRONMENTAL RESEARCH 2024; 257:119159. [PMID: 38754605 DOI: 10.1016/j.envres.2024.119159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/11/2024] [Accepted: 05/14/2024] [Indexed: 05/18/2024]
Abstract
Triphenyl phosphate (TPhP) is an organophosphate flame retardant that is widely used in many commercial products. The United States Environmental Protection Agency has listed TPhP as a priority compound that requires health risk assessment. We previously found that TPhP could accumulate in the placentae of mice and impair birth outcomes by activating peroxisome proliferator-activated receptor gamma (PPARγ) in the placental trophoblast. However, the underlying mechanism remains unknown. In this study, we used a mouse intrauterine exposure model and found that TPhP induced preeclampsia (PE)-like symptoms, including new on-set gestational hypertension and proteinuria. Immunofluorescence analysis showed that during placentation, PPARγ was mainly expressed in the labyrinth layer and decidua of the placenta. TPhP significantly decreased placental implantation depth and impeded uterine spiral artery remodeling by activating PPARγ. The results of the in vitro experiments confirmed that TPhP inhibited extravillous trophoblast (EVT) cell migration and invasion by activating PPARγ and inhibiting the PI3K-AKT signaling pathway. Overall, our data demonstrated that TPhP could activate PPARγ in EVT cells, inhibit cell migration and invasion, impede placental implantation and uterine spiral artery remodeling, then induce PE-like symptom and impair birth outcomes. Although the exposure doses used in this study was several orders of magnitude higher than human daily intake, our study highlights the placenta as a potential target organ of TPhP worthy of further research.
Collapse
Affiliation(s)
- Qian Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, Guangdong, China; School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Mengzhu Jiang
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, Guangdong, China; School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Xiaoxun Lu
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jiabin Hong
- The Third People's Hospital of Zhuhai, Zhuhai, 519000, Guangdong, China
| | - Yanqin Sun
- Department of Pathology, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Chun Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuting Chen
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Xingxing Chai
- Dongguan Key Laboratory for Development and Application of Experimental Animal Resources in Biomedical Industry, Laboratory Animal Center, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huanwen Tang
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Xiaoshan Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, Guangdong, China; School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
2
|
Zhang K, Zhang H, Wang B, Gao S, Sun C, Jia C, Cui J. NR2F1 overexpression alleviates trophoblast cell dysfunction by inhibiting GDF15/MAPK axis in preeclampsia. Hum Cell 2024; 37:1405-1420. [PMID: 39007956 DOI: 10.1007/s13577-024-01095-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
Abnormal functions of trophoblast cells are associated with the pathogenesis of preeclampsia (PE). Nuclear receptor subfamily 2 group F member 1 (NR2F1) acts as a transcriptionally regulator in many diseases, but its role in PE remains unknown. Hypoxia/reoxygenation (H/R)-stimulated HTR-8/SVneo cells were used to mimic PE injury in vitro. NR2F1 overexpression alleviated trophoblast apoptosis, as evidenced by the decreased number of TUNEL-positive cells and the downregulation of caspase 3 and caspase 9 expression in cells. NR2F1 overexpression increased the invasion and migration ability of HTR-8/SVneo cells, accompanied by increased protein levels of matrix metalloproteinase (MMP)-2 and MMP-9. mRNA-seq was applied to explore the underlying mechanism of NR2F1, identifying growth differentiation factor 15 (GDF15) as the possible downstream effector. Dual-luciferase reporter, ChIP-qPCR, and DNA pull-down assays confirmed that NR2F1 bound to the promoter of GDF15 and transcriptionally inhibited its expression. GDF15 overexpression increased apoptosis and decreased the ability of invasion and migration in HTR-8/SVneo cells expressing NR2F1. MAPK pathway was involved in the regulation of PE. Administration of p38 inhibitor, ERK inhibitor, and JNK inhibitor reversed the effect of simultaneous overexpression NR2F1 and GDF15 on trophoblast apoptosis, invasion, and migration. Our findings demonstrated that NR2F1 overexpression inhibited trophoblast apoptosis and promoted trophoblast invasion and migration. NR2F1 might negatively regulate GDF15 expression by binding to its promoter region, which further inhibited MAPK signaling pathway in PE. Our study highlights that NR2F1 might sever as a potential target in PE.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hailing Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Bing Wang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shanshan Gao
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Caiping Sun
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Cong Jia
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jinquan Cui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, No. 2, Jingba Road, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
3
|
Chen Z, Li Z, Zong Y, Xia B, Luo S, Deng G, Gao J. Exosome-delivered miR-410-3p reverses epithelial-mesenchymal transition, migration and invasion of trophoblasts in spontaneous abortion. J Cell Mol Med 2024; 28:e18097. [PMID: 38164738 PMCID: PMC10844701 DOI: 10.1111/jcmm.18097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 11/15/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Current studies have indicated that insufficient trophoblast epithelial-mesenchymal transition (EMT), migration and invasion are crucial for spontaneous abortion (SA) occurrence and development. Exosomal miRNAs play significant roles in embryonic development and cellular communication. Hereon, we explored the roles of serum exosomes derived from SA patients on trophoblast EMT, migration and invasion. Exosomes were isolated from normal control (NC) patients with abortion for unplanned pregnancy and SA patients, then characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting. Exosomal miRNA profiles were identified by miRNA sequencing. The effects of serum exosomes on trophoblast migration and invasion were detected by scratch wound healing and transwell assays, and other potential mechanisms were revealed by quantitative real-time PCR (RT-PCR), western blotting and dual-luciferase reporter assay. Finally, animal experiments were used to explore the effects of exosomal miR-410-3p on embryo absorption in mice. The serum exosomes from SA patients inhibited trophoblast EMT and reduced their migration and invasion ability in vitro. The miRNA sequencing showed that miR-410-3p was upregulated in SA serum exosomes. The functional experiments showed that SA serum exosomes restrained trophoblast EMT, migration and invasion by releasing miR-410-3p. Mechanistically, SA serum exosomal miR-410-3p inhibited trophoblast cell EMT, migration and invasion by targeting TNF receptor-associated factor 6 (TRAF6) at the post-transcriptional level. Besides, SA serum exosomal miR-410-3p inhibited the p38 MAPK signalling pathway by targeting TRAF6 in trophoblasts. Moreover, milk exosomes loaded with miR-410-3p mimic reached the maternal-fetal interface and aggravated embryo absorption in female mice. Clinically, miR-410-3p and TRAF6 expression were abnormal and negatively correlated in the placental villi of SA patients. Our findings indicated that exosome-derived miR-410-3p plays an important role between SA serum and trophoblasts in intercellular communication, suggesting a novel mechanism by which serum exosomal miRNA regulates trophoblasts in SA patients.
Collapse
Affiliation(s)
- Zhen‐yue Chen
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Zhen Li
- The Second Clinical College of Guangzhou University of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yun Zong
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Bo Xia
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Song‐ping Luo
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Gao‐pi Deng
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Jie Gao
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
4
|
Xiong M, Wang Q, Zhang X, Wen L, Zhao A. Decidual stromal cells-derived exosomes incurred insufficient migration and invasion of trophoblast by disturbing of β-TrCP-mediated snail ubiquitination and degradation in unexplained recurrent spontaneous abortion. Eur J Med Res 2024; 29:39. [PMID: 38195659 PMCID: PMC10775448 DOI: 10.1186/s40001-023-01598-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Exosomes released from decidual stromal cells (DSC-exos) play a crucial role in facilitating the epithelial-mesenchymal transition (EMT) of trophoblasts and insufficient trophoblasts EMT are associated with URSA (unexplained recurrent spontaneous abortion). However, the mechanisms underlying DSC-exos inducing EMT is not completely understood. METHODS DSC-exos of normal pregnant women (N-DSC-exos) and URSA patients (URSA-DSC-exos) were extracted and characterized. Characterization of the isolated DSC-exos was performed using with TEM (transmission electron microscopy), NTA (nanoparticle tracking analysis), and WB (western blot) techniques. Subsequently, these DSC-exos were co-cultured with trophoblasts cell lines (HTR-8/SVneo). The influence of both N-DSC-exos and URSA-DSC-exos on trophoblasts proliferation, invasion and migration, as well as on the expression of EMT-related proteins, was evaluated through a series of assays including CCK8 assays, wound healing assays, transwell assays, and western blot, respectively. Then rescue experiments were performed by β-TrCP knockdown or β-TrCP overexpressing trophoblasts with snail-siRNA transfection or β-TrCP overexpressing Lentivirus infection, respectively. Finally, animal experiments were employed to explore the effect of N-DSC-exos on embryo absorption in mice. RESULTS We found increased β-TrCP expression in the villus of URSA patients when compared to the normal pregnant women, alongside reduction in the levels of both snail and N-cadherin within URSA patients. N-DSC-exos can promote the EMT of the trophoblast by inhibiting β-TrCP-mediated ubiquitination and degradation of transcription factor snail. Moreover the capacity to promote EMT was found to be more potent in N-DSC-exos than URSA-DSC-exos. Down-regulation of snail or overexpression of β-TrCP can reverse the effects of N-DSC-exos on trophoblast. Finally, in vivo experiment suggested that N-DSC-exos significantly reduced the embryo resorption rate of spontaneous abortion mouse model. CONCLUSIONS Our findings indicate that URSA-DSC-exos caused insufficient migration and invasion of trophoblast because of disturbing of β-TrCP-mediated ubiquitination and degradation of EMT transcription factor snail. Elucidating the underlying mechanism of this dysregulation may shed light on the novel pathways through which DSC-exos influence trophoblast function, thereby contributing to our understanding of their role in URSA.
Collapse
Affiliation(s)
- Miao Xiong
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai, 200127, People's Republic of China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, People's Republic of China
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Qiaohong Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai, 200127, People's Republic of China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, People's Republic of China
| | - Xiaoxin Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai, 200127, People's Republic of China
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, People's Republic of China
| | - Liping Wen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, People's Republic of China
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai, 200127, People's Republic of China.
- Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Chen J, Song T, Yang S, Meng Q, Han X, Wu Z, Cheng JC, Fang L. Snail mediates GDF-8-stimulated human extravillous trophoblast cell invasion by upregulating MMP2 expression. Cell Commun Signal 2023; 21:93. [PMID: 37143106 PMCID: PMC10158255 DOI: 10.1186/s12964-023-01107-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/19/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Extravillous trophoblast (EVT) cell invasion is a tightly regulated process that requires for a normal pregnancy. The epithelial-mesenchymal transition (EMT) has been implicated in EVT cell invasion. Growth differentiation factor-8 (GDF-8), a member of the transforming growth factor-beta (TGF-β) superfamily, is expressed in the human placenta and promotes EVT cell invasion by upregulating the expression of matrix metalloproteinase 2 (MMP2). However, the underlying molecular mechanism of GDF-8-induced MMP2 expression remains undetermined. Therefore, the present study aims to examine the role of Snail and Slug, the EMT-related transcriptional regulators, in GDF-8-stimulated MMP2 expression and cell invasion in HTR-8/SVneo human EVT cell line and primary cultures of human EVT cells. METHODS HTR-8/SVneo and primary cultures of human EVT cells were used to examine the effect of GDF-8 on MMP2 expression and explore the underlying mechanism. For gene silencing and overexpression, the HTR-8/SVneo cell line was used to make the experiments more technically feasible. The cell invasiveness was measured by Matrigel-coated transwell invasion assay. RESULTS GDF-8 stimulated MMP2 expression in both HTR-8/SVneo and primary EVT cells. The stimulatory effect of GDF-8 on MMP2 expression was blocked by the inhibitor of TGF-β type-I receptors, SB431542. Treatment with GDF-8 upregulated Snail and Slug expression in both HTR-8/SVneo and primary EVT cells. The stimulatory effects of GDF-8 on Snail and Slug expression were blocked by pretreatment of SB431542 and siRNA-mediated knockdown of SMAD4. Interestingly, using the siRNA knockdown approach, our results showed that Snail but not Slug was required for the GDF-8-induced MMP2 expression and cell invasion in HTR-8/SVneo cells. The reduction of MMP2 expression in the placentas with preeclampsia (PE) was also observed. CONCLUSIONS These findings discover the physiological function of GDF-8 in the human placenta and provide important insights into the regulation of MMP2 expression in human EVT cells. Video Abstract.
Collapse
Affiliation(s)
- Jiaye Chen
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Tinglin Song
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Sizhu Yang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Qingxue Meng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Xiaoyu Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Ze Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
6
|
Atigan A, Tan S, Cetin H, Guler OT, Ozdamar S, Karakaya YA. CD97 expression level and its effect on cell adhesion in Preeclampsia. BMC Pregnancy Childbirth 2022; 22:967. [PMID: 36572878 PMCID: PMC9791749 DOI: 10.1186/s12884-022-05280-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/02/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES Cellular interactions and cell adhesion underlie preeclampsia (PE). The aim of the current study is to investigate the role of cell adhesion molecules such as CD97, neural (N)-cadherin, epithelial (E) -cadherin and integrin beta-4 in PE. METHODS This prospective study included 20 pregnant women with PE and a control group of 16 healthy pregnant women who were matched for age, gestational age, gravida and parity. Standard blood tests and placental cell adhesion molecule immunohistochemical staining were examined. RESULTS The creatinine, uric acid and lactate dehydrogenase (LDH) levels from standard blood tests were found to be statistically higher in the PE group (p = 0.002, p = 0.000, p = 0.001; respectively). In the PE group, the CD97 maternal serum level was statistically significantly lower, as was its immunohistochemical expression in placental sections (p = 0.028, p = 0.000; respectively). The E-cadherin expression score was statistically higher in the PE group compared to the control group (3,65 ± 1,84 vs 2,06 ± 1,76 respectively; p = 0.003). The N-cadherin expression score was statistically lower in the PE group compared to the control group (1,50 ± 0,82 vs 2,43 ± 1,59 respectively; p = 0.049). Integrin beta-4 was not statistically different between groups. CONCLUSIONS Cellular interaction may be responsible for PE as in cancer. A balance in intercellular communication, as researched in cancer therapy, may offer the solution in PE.
Collapse
Affiliation(s)
- Ayhan Atigan
- grid.440448.80000 0004 0384 3505Department of Obstetrics and Gynecology, Faculty of Medicine, Karabuk University, School of Medicine, Karabuk, Turkey
| | - Semih Tan
- grid.411742.50000 0001 1498 3798Department of Histology and Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Hulya Cetin
- grid.411742.50000 0001 1498 3798Department of Histology and Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Omer Tolga Guler
- grid.411742.50000 0001 1498 3798Department of Obstetrics and Gynecology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Saim Ozdamar
- grid.411742.50000 0001 1498 3798Department of Histology and Embryology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Yeliz Arman Karakaya
- grid.411742.50000 0001 1498 3798Department of Pathology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
7
|
Lamptey J, Czika A, Aremu JO, Pervaz S, Adu-Gyamfi EA, Otoo A, Li F, Wang YX, Ding YB. The role of fascin in carcinogenesis and embryo implantation. Exp Cell Res 2021; 409:112885. [PMID: 34662557 DOI: 10.1016/j.yexcr.2021.112885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 01/02/2023]
Abstract
The cytoskeleton, with its actin bundling proteins, plays crucial roles in a host of cellular function, such as cancer metastasis, antigen presentation and trophoblast migration and invasion, as a result of cytoskeletal remodeling. A key player in cytoskeletal remodeling is fascin. Upregulation of fascin induces the transition of epithelial phenotypes to mesenchymal phenotypes through complex interaction with transcription factors. Fascin expression also regulates mitochondrial F-actin to promote oxidative phosphorylation (OXPHOS) in some cancer cells. Trophoblast cells, on the other hand, exhibit similar physiological functions, involving the upregulation of genes crucial for its migration and invasion. Owing to the similar tumor-like characteristics among cancer and trophoblats, we review recent studies on fascin in relation to cancer and trophoblast cell biology; and based on existing evidence, link fascin to the establishment of the maternal-fetal interface.
Collapse
Affiliation(s)
- Jones Lamptey
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China; Kumasi Centre for Collaborative Research in Tropical Medicine, KCCR, UPO, Kumasi, Ghana.
| | - Armin Czika
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - John Ogooluwa Aremu
- Department of Human Anatomy and Histoembryology, Harbin Medical University, Harbin, People's Republic of China
| | - Sadaf Pervaz
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Enoch Appiah Adu-Gyamfi
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Antonia Otoo
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying-Xiong Wang
- School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China.
| | - Yu-Bin Ding
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
8
|
Jie Q, Sun F, Li Q, Zhu J, Wei Y, Yang H, Long P, Wang Z, Yang X, Li D, Huang L, Ma Y. Downregulated ribosomal protein L39 inhibits trophoblast cell migration and invasion by targeting E-cadherin in the placenta of patients with preeclampsia. FASEB J 2021; 35:e21322. [PMID: 33710681 DOI: 10.1096/fj.202002061r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/06/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022]
Abstract
Early-onset preeclampsia (PE) is a pregnancy complication that can lead to severe adverse maternal and fetal outcomes. However, the mechanisms underlying the development of early-onset PE are not fully understood. Ribosomal protein L39 (RPL39) is a member of the S39E family of ribosomal proteins that plays an important role in stem cell self-renewal, cancer metastasis, and chemoresistance. In this study, we aimed to explore the potential function of RPL39 in placental trophoblast cells. We analyzed the expression of RPL39 in early-onset PE and normal placental tissues using real-time PCR, western blot analysis, and immunohistochemistry. The results showed that RPL39 was markedly downregulated in early-onset PE placental tissues. RPL39 knockdown inhibited trophoblast cell proliferation, migration, and invasion, as well as placental explant outgrowth. Flow cytometry analysis suggested that knockdown of RPL39 resulted in cell cycle arrest at the G0/G1 phase, but had no significant effect on cell apoptosis. We also found that RPL39 knockdown could alter cell morphology. We then measured the expression of the epithelial cell marker E-cadherin following knockdown of RPL39 in Bewo and HTR8/SVneo cells. RPL39 knockdown increased the expression of E-cadherin. Furthermore, E-cadherin expression was upregulated in placental explant outgrowth tissues transfected with RPL39 small interfering RNA. In conclusion, RPL39 plays an essential role in proliferation, invasion, and migration of trophoblast cells by targeting E-cadherin. Our findings provide novel insight into the mechanisms underlying the occurrence of early-onset PE.
Collapse
Affiliation(s)
- Qiuling Jie
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fei Sun
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qi Li
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Juan Zhu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yunjian Wei
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Huamei Yang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ping Long
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhen Wang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiaohui Yang
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Dan Li
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanlin Ma
- Department of Hainan Provincial Key Laboratory for human reproductive medicine and Genetic Research, Key Laboratory of Tropical Translational Medicine of Ministry of Education, Reproductive Medicine, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
9
|
Marinello WP, Mohseni ZS, Cunningham SJ, Crute C, Huang R, Zhang JJ, Feng L. Perfluorobutane sulfonate exposure disrupted human placental cytotrophoblast cell proliferation and invasion involving in dysregulating preeclampsia related genes. FASEB J 2020; 34:14182-14199. [PMID: 32901980 DOI: 10.1096/fj.202000716rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
We reported that maternal PFBS, an emerging pollutant, exposure is positively associated with preeclampsia which can result from aberrant trophoblasts invasion and subsequent placental ischemia. In this study, we investigated the effects of PFBS on trophoblasts proliferation/invasion and signaling pathways. We exposed a human trophoblast line, HTR8/SVneo, to PFBS. Cell viability, proliferation, and cell cycle were evaluated by the MTS assay, Ki-67 staining, and flow cytometry, respectively. We assessed cell migration and invasion with live-cell imaging-based migration assay and matrigel invasion assay, respectively. Signaling pathways were examined by Western blot, RNA-seq, and qPCR. PFBS exposure interrupted cell proliferation and invasion in a dose-dependent manner. PFBS (100 μM) did not cause cell death but instead significant cell proliferation without cell cycle disruption. PFBS (10 and 100 μM) decreased cell migration and invasion, while PFBS (0.1 μM) significantly increased cell invasion but not migration. Further, RNA-seq analysis identified dysregulated HIF-1α target genes that are relevant to cell proliferation/invasion and preeclampsia, while Western Blot data showed the activation of HIF-1α, but not Notch, ERK1/2, (PI3K)AKT, and P38 pathways. PBFS exposure altered trophoblast cell proliferation/invasion which might be mediated by preeclampsia-related genes, suggesting a possible association between prenatal PFBS exposure and adverse placentation.
Collapse
Affiliation(s)
- William P Marinello
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Zahra S Mohseni
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Sarah J Cunningham
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Christine Crute
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Jun J Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Farah O, Nguyen C, Tekkatte C, Parast MM. Trophoblast lineage-specific differentiation and associated alterations in preeclampsia and fetal growth restriction. Placenta 2020; 102:4-9. [PMID: 33218578 DOI: 10.1016/j.placenta.2020.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 12/26/2022]
Abstract
The human placenta is a poorly-understood organ, but one that is critical for proper development and growth of the fetus in-utero. The epithelial cell type that contributes to primary placental functions is called "trophoblast," including two main subtypes, villous and extravillous trophoblast. Cytotrophoblast and syncytiotrophoblast comprise the villous compartment and contribute to gas and nutrient exchange, while extravillous trophoblast invade and remodel the uterine wall and vessels, in order to supply maternal blood to the growing fetus. Abnormal differentiation of trophoblast contributes to placental dysfunction and is associated with complications of pregnancy, including preeclampsia (PE) and fetal growth restriction (FGR). This review describes what is known about the cellular organization of the placenta during both normal development and in the setting of PE/FGR. It also explains known trophoblast lineage-specific markers and pathways regulating their differentiation, and how these are altered in the setting of PE/FGR, focusing on studies which have used human placental tissues. Finally, it also highlights remaining questions and needed resources to advance this field.
Collapse
Affiliation(s)
- Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Calvin Nguyen
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chandana Tekkatte
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
11
|
Lu W, Ma YY, Shao QQ, Liang J, Qi TT, Huang Y, Wang QJ. ROS/p53/miR‑335‑5p/Sp1 axis modulates the migration and epithelial to mesenchymal transition of JEG‑3 cells. Mol Med Rep 2019; 21:1208-1216. [PMID: 31894323 PMCID: PMC7003020 DOI: 10.3892/mmr.2019.10901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/18/2019] [Indexed: 11/07/2022] Open
Abstract
Differential expression of microRNA (miR)-335-5p, a key tumor suppressor, has been detected in pre-eclampsia (PE) placentas. However, the role of miR-335-5p in the pathogenesis of PE and the factor modulating its aberrant expression remain unknown. The present study used JEG-3 cells in vitro to investigate these mechanisms. The role of miR-335-5p in proliferation, apoptosis and migration of JEG-3 cells was investigated using MTT, Annexin V-FITC/PI, Transwell migration and wound healing assays, respectively. miR-335-5p expression levels were analyzed using reverse transcription-quantitative PCR. The expression levels of E-cadherin, N-cadherin, Snail, specificity protein 1 (Sp1) and p53 were assessed using western blot analysis. Cell viability analysis was performed using the Cell Counting Kit-8 assay. The intracellular reactive oxygen species (ROS) levels were detected using a 2,7-dichlorodihydrofluorescein diacetate assay. The present results suggested that miR-335-5p did not affect the proliferation or apoptotic rate of JEG-3 cells. Overexpression of miR-335-5p significantly inhibited the migration of JEG-3 cells, decreased the expression levels of Sp1, N-cadherin and Snail, and increased E-cadherin expression. Sp1 silencing produced similar results in JEG-3 cells. H2O2 significantly increased the intracellular ROS levels and miR-335-5p expression, whereas N-acetyl-cysteine pretreatment prior to H2O2 treatment reversed the increases in miR-335-5p expression. Knockdown of p53 significantly decreased the expression levels of miR-335-5p in JEG-3 cells and in H2O2-treated cells. The present results suggested that miR-335-5p expression levels in trophoblast cells could be increased by ROS in a p53-dependent manner, leading to the downregulation of Sp1 and subsequent inhibition of epithelial to mesenchymal transition and cell migration. The present results may provide novel evidence on the etiology of PE.
Collapse
Affiliation(s)
- Wei Lu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yu-Yan Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qian-Qian Shao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jie Liang
- Central Sterile Supply Department, People's Hospital of Fangzi, Weifang, Shandong 261200, P.R. China
| | - Tong-Tong Qi
- School of Pharmaceutical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yi Huang
- School of Pharmaceutical Science, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing-Jie Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Institute of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
12
|
Abdulghani M, Song G, Kaur H, Walley JW, Tuteja G. Comparative Analysis of the Transcriptome and Proteome during Mouse Placental Development. J Proteome Res 2019; 18:2088-2099. [PMID: 30986076 DOI: 10.1021/acs.jproteome.8b00970] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The condition of the placenta is a determinant of the short- and long-term health of the mother and the fetus. However, critical processes occurring in early placental development, such as trophoblast invasion and establishment of placental metabolism, remain poorly understood. To gain a better understanding of the genes involved in regulating these processes, we utilized a multiomics approach, incorporating transcriptome, proteome, and phosphoproteome data generated from mouse placental tissue collected at two critical developmental time points. We found that incorporating information from both the transcriptome and proteome identifies genes associated with time point-specific biological processes, unlike using the proteome alone. We further inferred genes upregulated on the basis of the proteome data but not the transcriptome data at each time point, leading us to identify 27 genes that we predict to have a role in trophoblast migration or placental metabolism. Finally, using the phosphoproteome data set, we discovered novel phosphosites that may play crucial roles in the regulation of placental transcription factors. By generating the largest proteome and phosphoproteome data sets in the developing placenta, and integrating transcriptome analysis, we uncovered novel aspects of placental gene regulation.
Collapse
Affiliation(s)
- Majd Abdulghani
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Gaoyuan Song
- Department of Plant Pathology and Microbiology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Haninder Kaur
- Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Justin W Walley
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Plant Pathology and Microbiology , Iowa State University , Ames , Iowa 50011-1079 , United States
| | - Geetu Tuteja
- Interdepartmental Genetics and Genomics , Iowa State University , Ames , Iowa 50011-1079 , United States.,Department of Genetics, Development, and Cell Biology , Iowa State University , Ames , Iowa 50011-1079 , United States
| |
Collapse
|
13
|
Tseng AM, Mahnke AH, Wells AB, Salem NA, Allan AM, Roberts VH, Newman N, Walter NA, Kroenke CD, Grant KA, Akison LK, Moritz KM, Chambers CD, Miranda RC. Maternal circulating miRNAs that predict infant FASD outcomes influence placental maturation. Life Sci Alliance 2019; 2:2/2/e201800252. [PMID: 30833415 PMCID: PMC6399548 DOI: 10.26508/lsa.201800252] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
Maternal gestational circulating microRNAs, predictive of adverse infant outcomes, including growth deficits, following prenatal alcohol exposure, contribute to placental pathology by impairing the EMT pathway in trophoblasts. Prenatal alcohol exposure (PAE), like other pregnancy complications, can result in placental insufficiency and fetal growth restriction, although the linking causal mechanisms are unclear. We previously identified 11 gestationally elevated maternal circulating miRNAs (HEamiRNAs) that predicted infant growth deficits following PAE. Here, we investigated whether these HEamiRNAs contribute to the pathology of PAE, by inhibiting trophoblast epithelial–mesenchymal transition (EMT), a pathway critical for placental development. We now report for the first time that PAE inhibits expression of placental pro-EMT pathway members in both rodents and primates, and that HEamiRNAs collectively, but not individually, mediate placental EMT inhibition. HEamiRNAs collectively, but not individually, also inhibited cell proliferation and the EMT pathway in cultured trophoblasts, while inducing cell stress, and following trophoblast syncytialization, aberrant endocrine maturation. Moreover, a single intravascular administration of the pooled murine-expressed HEamiRNAs, to pregnant mice, decreased placental and fetal growth and inhibited the expression of pro-EMT transcripts in the placenta. Our data suggest that HEamiRNAs collectively interfere with placental development, contributing to the pathology of PAE, and perhaps also, to other causes of fetal growth restriction.
Collapse
Affiliation(s)
- Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Alan B Wells
- Clinical and Translational Research Institute, University of California San Diego, San Diego, CA, USA.,Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Nihal A Salem
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Andrea M Allan
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, USA
| | - Victoria Hj Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Natali Newman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Nicole Ar Walter
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Christopher D Kroenke
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lisa K Akison
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Christina D Chambers
- Clinical and Translational Research Institute, University of California San Diego, San Diego, CA, USA .,Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | | |
Collapse
|
14
|
Butler TM, Pater JA, MacPhee DJ. Integrin linked kinase regulates syncytialization of BeWo trophoblast cells. Biol Reprod 2017; 96:673-685. [PMID: 28339614 DOI: 10.1095/biolreprod.116.145748] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/27/2017] [Indexed: 12/31/2022] Open
Abstract
During placental development, mononuclear villous cytotrophoblast cells differentiate and fuse with the overlying syncytiotrophoblast. This process requires the dissolution of E-cadherin (CDH1)-containing adherens junctions in cytotrophoblast. Integrin linked kinase (ILK) can downregulate CDH1 through poly (ADP-ribose) polymerase 1 (PARP1) and Snail-1 (SNAI1) during epithelial-mesenchymal transition. ILK is known to be expressed in cytotrophoblast; thus, the role of a potential ILK-PARP1-SNAI1 pathway in aiding trophoblast syncytialization via the downregulation of CDH1 was examined. The spatiotemporal expression of PARP1, SNAI1, and CDH1 were determined in first and early second trimester chorionic villi, term villi, and BeWo cells by immunofluorescence analysis. PARP1 and SNAI1 were highly detectable in villous cytotrophoblast nuclei of human chorionic villi and SNAI1 expression, in particular, also persisted in syncytiotrophoblast. In BeWo cells undergoing syncytialization, PARP1 and SNAI1 increasingly localized to cell nuclei in correlation with decreased CDH1 expression. Using luciferase reporter assays, it was determined that PARP1 and SNAI1 promoter activities were significantly higher in BeWo cells during syncytialization compared to the activities in proliferating cells. Overexpression of wild type or constitutively active ILK also resulted in significantly increased PARP1 and SNAI1 promoter activities while dominant negative ILK overexpression significantly reduced promoter activities. Lastly, siRNA-mediated depletion of ILK expression in BeWo cells undergoing syncytialization resulted in significantly reduced SNAI1 expression and a significant reduction in the incidence of syncytialization correlating with increased CDH1 expression. These results demonstrate that ILK aids trophoblast syncytialization via the downregulation of CDH1, perhaps through an ILK-PARP1-SNAI1 pathway.
Collapse
Affiliation(s)
- Trina M Butler
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Newfoundland and Labrador Canada
| | - Justin A Pater
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Daniel J MacPhee
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, Newfoundland and Labrador Canada.,Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,One Reproductive Health Research Group, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
15
|
Chen Y, Khoo SK, Leach R, Wang K. MTA3 Regulates Extravillous Trophoblast Invasion Through NuRD Complex. AIMS MEDICAL SCIENCE 2017; 4:17-27. [PMID: 28959722 PMCID: PMC5613952 DOI: 10.3934/medsci.2017.1.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extravillous trophoblast (EVT) invasion is required for remodeling uterine tertiary arteries and placenta development during pregnancy. Compromised EVT invasion may contribute to the pathology of placenta-related diseases. Metastasis -associated protein 3 (MTA3) is one of the subunits of nucleosome remodeling and deacetylation (NuRD) complex that represses transcription in a histone deacetylase-dependent manner. MTA3 is reported to be down-regulated in preeclamptic placentas, suggesting its potential role in EVT invasion. Here, we investigate the role of MTA3 in EVT invasion by studying its molecular mechanisms in EVT cells. First, we confirmed MTA3 expression in the EVT cells in human placenta using immunohistochemistry. We then used lentivirus-mediated MTA3 short hairpin RNA (shRNA) to knock down MTA3 expression in EVT-derived HTR8/SVneo cells and found higher invasion capacity in MTA3 knockdown cells. Using quantitative real-time PCR, we showed higher expression of invasion-related genes matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), and transcription factor Snail in MTA3 knockdown compared with control cells. Co-immunoprecipitation-Western blot assay showed the protein-protein interaction of histone deacetylase 1 (HDAC1), a subunit of NuRD, with MTA3 in HTR8/SVneo cells. Co-immunoprecipitation-Mass spectrometry assay further identified 71 proteins interacting with MTA3, including NuRD subunits, heterochromatin proteins, epigenetics modifiers and transcription factors. This result not only indicated the involvement of NuRD complex in MTA3’s function, but also demonstrated the complicated multiple co-players in MTA3 and NuRD complex mediated transcription repression in EVT. In summary, our data demonstrates that MTA3 regulates EVT invasion and related gene expression via NuRD complex in EVT.
Collapse
Affiliation(s)
- Ying Chen
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Sok Kean Khoo
- Department of Cell and Molecular Biology, Grand Valley State University, Grand Rapids, MI, USA
| | - Richard Leach
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Kai Wang
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
16
|
Polejaeva IA, Rutigliano HM, Wells KD. Livestock in biomedical research: history, current status and future prospective. Reprod Fertil Dev 2017; 28:112-24. [PMID: 27062879 DOI: 10.1071/rd15343] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Livestock models have contributed significantly to biomedical and surgical advances. Their contribution is particularly prominent in the areas of physiology and assisted reproductive technologies, including understanding developmental processes and disorders, from ancient to modern times. Over the past 25 years, biomedical research that traditionally embraced a diverse species approach shifted to a small number of model species (e.g. mice and rats). The initial reasons for focusing the main efforts on the mouse were the availability of murine embryonic stem cells (ESCs) and genome sequence data. This powerful combination allowed for precise manipulation of the mouse genome (knockouts, knockins, transcriptional switches etc.) leading to ground-breaking discoveries on gene functions and regulation, and their role in health and disease. Despite the enormous contribution to biomedical research, mouse models have some major limitations. Their substantial differences compared with humans in body and organ size, lifespan and inbreeding result in pronounced metabolic, physiological and behavioural differences. Comparative studies of strategically chosen domestic species can complement mouse research and yield more rigorous findings. Because genome sequence and gene manipulation tools are now available for farm animals (cattle, pigs, sheep and goats), a larger number of livestock genetically engineered (GE) models will be accessible for biomedical research. This paper discusses the use of cattle, goats, sheep and pigs in biomedical research, provides an overview of transgenic technology in farm animals and highlights some of the beneficial characteristics of large animal models of human disease compared with the mouse. In addition, status and origin of current regulation of GE biomedical models is also reviewed.
Collapse
Affiliation(s)
- Irina A Polejaeva
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Heloisa M Rutigliano
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Kevin D Wells
- Division of Animal Sciences, Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
17
|
Du L, Kuang L, He F, Tang W, Sun W, Chen D. Mesenchymal-to-epithelial transition in the placental tissues of patients with preeclampsia. Hypertens Res 2016; 40:67-72. [DOI: 10.1038/hr.2016.97] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 04/17/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
|
18
|
Jurkovic I, Gecek I, Skrtic A, Zmijanac Partl J, Nikuseva Martic T, Serman A, Galesic Ljubanovic D, Serman L. ELF5 transcription factor expression during gestation in humans and rats - an immunohistochemical analysis. J Matern Fetal Neonatal Med 2016; 30:1261-1266. [PMID: 27384067 DOI: 10.1080/14767058.2016.1210596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The purpose of this study was to measure immunohistochemically the expression of ELF5 protein in term human and rat placentas and in human placentas associated with gestational diabetes (GD) and intrauterine growth restriction (IUGR). METHODS The results were quantitated stereologically using the stereological variable of volume density. A semiquantitative analysis was performed independently by a certified pathologist. RESULTS Total expression of ELF5 protein was higher in pathological pregnancies than in corresponding control term placentas, with both methods of quantifications showing similar results. In addition, ELF5 expression was also higher in connective tissue and blood vessels in chorionic villi in IUGR placentas (but not in GD placentas) compared to healthy controls. ELF5 is higher in placenta as a whole and in most of its components in both pathologies. The two exceptions are chorionic plates in IUGR and decidua in GD, where its expression is lower than in healthy controls. CONCLUSIONS We have shown that IUGR and GD are associated with significantly increased levels of ELF5 protein in placentas, which suggests that ELF5 may play an important role in normal placentation. However, these are term placentas and to study ELF5 in premature births would give better insight into human placentation in health and disease.
Collapse
Affiliation(s)
- Ivana Jurkovic
- a Department of Biology , School of Medicine, University of Zagreb , Zagreb , Croatia
| | - Iva Gecek
- b Department of Pathology , University Hospital Dubrava , Zagreb , Croatia
| | - Anita Skrtic
- c Department of Pathology , School of Medicine, University of Zagreb , Zagreb , Croatia
| | - Jasenka Zmijanac Partl
- d Department of Gynecology and Obstetrics , University Hospital Merkur , Zagreb , Croatia , and
| | | | - Alan Serman
- e Department of Gynecology and Obstetrics , School of Medicine, University of Zagreb , Zagreb , Croatia
| | | | - Ljiljana Serman
- a Department of Biology , School of Medicine, University of Zagreb , Zagreb , Croatia
| |
Collapse
|
19
|
E Davies J, Pollheimer J, Yong HEJ, Kokkinos MI, Kalionis B, Knöfler M, Murthi P. Epithelial-mesenchymal transition during extravillous trophoblast differentiation. Cell Adh Migr 2016; 10:310-21. [PMID: 27070187 DOI: 10.1080/19336918.2016.1170258] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
A successful pregnancy depends on the intricate and timely interactions of maternal and fetal cells. Placental extravillous cytotrophoblast invasion involves a cellular transition from an epithelial to mesenchymal phenotype. Villous cytotrophoblasts undergo a partial epithelial to mesenchymal transition (EMT) when differentiating into extravillous cytotrophoblasts and gain the capacity to migrate and invade. This review summarizes our current knowledge regarding known regulators of EMT in the human placenta, including the inducers of EMT, upstream transcription factors that control EMT and the downstream effectors, cell adhesion molecules and their differential expression and functions in pregnancy pathologies, preeclampsia (PE) and fetal growth restriction (FGR). The review also describes the research strategies that were used for the identification of the functional role of EMT targets in vitro. A better understanding of molecular pathways driven by placental EMT and further elucidation of signaling pathways underlying the developmental programs may offer novel strategies of targeted therapy for improving feto-placental growth in placental pathologies including PE and FGR.
Collapse
Affiliation(s)
- Jessica E Davies
- a Department of Obstetrics and Gynecology , The University of Melbourne , Parkville , Victoria , Australia.,b Department of Maternal-Fetal Medicine Pregnancy Research Centre , The Royal Women's Hospital , Parkville , Victoria , Australia
| | - Jürgen Pollheimer
- c Department of Obstetrics and Fetal-Maternal Medicine , Reproductive Biology Unit, Medical University of Vienna , Vienna , Austria
| | - Hannah E J Yong
- a Department of Obstetrics and Gynecology , The University of Melbourne , Parkville , Victoria , Australia.,b Department of Maternal-Fetal Medicine Pregnancy Research Centre , The Royal Women's Hospital , Parkville , Victoria , Australia
| | - Maria I Kokkinos
- b Department of Maternal-Fetal Medicine Pregnancy Research Centre , The Royal Women's Hospital , Parkville , Victoria , Australia
| | - Bill Kalionis
- a Department of Obstetrics and Gynecology , The University of Melbourne , Parkville , Victoria , Australia.,b Department of Maternal-Fetal Medicine Pregnancy Research Centre , The Royal Women's Hospital , Parkville , Victoria , Australia
| | - Martin Knöfler
- c Department of Obstetrics and Fetal-Maternal Medicine , Reproductive Biology Unit, Medical University of Vienna , Vienna , Austria
| | - Padma Murthi
- a Department of Obstetrics and Gynecology , The University of Melbourne , Parkville , Victoria , Australia.,b Department of Maternal-Fetal Medicine Pregnancy Research Centre , The Royal Women's Hospital , Parkville , Victoria , Australia.,d Department of Medicine , School of Clinical Sciences, Monash University , Clayton , Victoria , Australia
| |
Collapse
|
20
|
Zhang D, Liu H, Zeng J, Miao X, Huang W, Chen H, Huang Y, Li Y, Ye D. Glucocorticoid exposure in early placentation induces preeclampsia in rats via interfering trophoblast development. Gen Comp Endocrinol 2016; 225:61-70. [PMID: 26407501 DOI: 10.1016/j.ygcen.2015.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/18/2015] [Accepted: 09/17/2015] [Indexed: 12/29/2022]
Abstract
In pregnancy, placenta can be exposed to glucocorticoids (GCs) via several ways, which may disturb placentation and adversely affect pregnancy. Preeclampsia (PE) is thought to be attributed, in part, to impaired trophoblast development. The purpose of the present study was to confirm that GC exposure in early placentation could lead to PE in rats, with the mechanisms involving dysregulated trophoblast development. In the study, pregnant rats were administered with 2.5mg/kg Dex subcutaneously once per day from gestational day 7 to 13. Maternal systolic blood pressure and urinary albumin were increased, while both fetus and placenta were restricted after GC exposure relative to the control group. GC exposure also contributed to placental abnormalities and renal impairment. Moreover, placental oxidative damage was increased along with placental hypoxia-inducible factor 1-alpha (HIF1A) overexpression after GC treatment. Mechanically, GC induced PE in rat partially through inhibiting trophoblast proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), which involved phospho-extracellular signal regulated kinase (p-ERK) downregulation. Furthermore, GC receptor was required for the inhibition of GC on trophoblast proliferation, migration, invasion and EMT in vitro. These findings suggest that GC exposure in early placentation could contribute to PE in pregnant rats, with the mechanisms involving inhibition of trophoblast proliferation, migration, invasion and EMT by GC.
Collapse
Affiliation(s)
- Dongxin Zhang
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China; Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Haojing Liu
- Department of Internal Medicine, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Ji Zeng
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Xili Miao
- Department of Clinical Laboratory, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| | - Wei Huang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Yinping Huang
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325003, People's Republic of China
| | - Yongsheng Li
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Duyun Ye
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China.
| |
Collapse
|
21
|
Li Y, Klausen C, Zhu H, Leung PCK. Activin A Increases Human Trophoblast Invasion by Inducing SNAIL-Mediated MMP2 Up-Regulation Through ALK4. J Clin Endocrinol Metab 2015; 100:E1415-27. [PMID: 26305619 DOI: 10.1210/jc.2015-2134] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT Activin A increases matrix metalloproteinase (MMP) 2 expression and cell invasion in human trophoblasts, but whether the expression of MMP2 is essential for the proinvasive effect of activin A has yet to be determined. Moreover, the identity of the activin receptor-like kinase (ALK; TGF-β type I receptors) and downstream transcription factors (eg, SNAIL and SLUG) mediating the effects of activin on MMP2 expression and trophoblast cell invasion remains unknown. OBJECTIVE To elucidate the role of MMP2 in activin A-induced human trophoblast cell invasion as well as the involvement of ALK4 and SNAIL. DESIGN HTR8/SVneo immortalized human extravillous cytotrophoblast (EVT) cells and primary cultures of human first-trimester EVT cells were used as study models. Small interfering RNA (siRNA)-mediated knockdown approaches were used to investigate the molecular determinants of activin A-mediated functions. MAIN OUTCOME MEASURES Levels of mRNA and protein were examined by reverse transcription-quantitative real-time PCR and Western blot, respectively. Cell invasiveness was measured by Matrigel-coated transwell assays. RESULTS Treatment of HTR8/SVneo cells with activin A increased the production of SNAIL, SLUG, and MMP2 without altering that of MMP9, TIMP1, TIMP2, TWIST, RUNX2, ZEB1, or ZEB2. Similarly, activin A up-regulated the mRNA and protein levels of SNAIL and MMP2 in primary EVT cells. Knockdown of SNAIL attenuated activin A-induced MMP2 up-regulation in HTR8/SVneo and primary EVT cells. In HTR8/SVneo cells, activin A-induced production of SNAIL and MMP2 was abolished by pretreatment with the TGF-β type I receptor (ALK4/5/7) inhibitor SB431542 or siRNA targeting ALK4, SMAD2/3, or common SMAD4. Likewise, knockdown of ALK4 or SMAD4 abolished the stimulatory effects of activin A on SNAIL and MMP2 expression in primary EVT cells. Importantly, activin A-induced HTR8/SVneo and primary EVT cell invasion were attenuated by siRNA-mediated depletion of ALK4 or MMP2. CONCLUSION Activin A induces human trophoblast cell invasion by inducing SNAIL-mediated MMP2 expression through ALK4 in a SMAD2/3-SMAD4-dependent manner.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Hua Zhu
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
22
|
Tang C, Mei L, Pan L, Xiong W, Zhu H, Ruan H, Zou C, Tang L, Iguchi T, Wu X. Hedgehog signaling through GLI1 and GLI2 is required for epithelial-mesenchymal transition in human trophoblasts. Biochim Biophys Acta Gen Subj 2015; 1850:1438-48. [PMID: 25888497 DOI: 10.1016/j.bbagen.2015.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 03/31/2015] [Accepted: 04/08/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) is critical for human placental development, trophoblastic differentiation, and pregnancy-associated diseases. Here, we investigated the effects of hedgehog (HH) signaling on EMT in human trophoblasts, and further explored the underlying mechanism. METHODS Human primary cytotrophoblasts and trophoblast-like JEG-3 cells were used as in vitro models. Quantitative real-time RT-PCR and Western blot analysis were performed to examine mRNA and protein levels, respectively. Lentiviruses expressing short hairpin RNA were used to knock down the target genes. Reporter assays and chromatin immunoprecipitation were performed to determine the transactivity. Cell migration, invasion and colony formation were accessed by wound healing, Matrigel-coated transwell, and colony formation assays, respectively. RESULTS Activation of HH signaling induced the transdifferentiation of cytotrophoblasts and trophoblast-like JEG-3 cells from epithelial to mesenchymal phenotypes, exhibiting the decreases in E-Cadherin expression as well as the increases in vimentin expression, invasion, migration and colony formation. Knockdown of GLI1 and GLI2 but not GLI3 attenuated HH-induced transdifferentiation, whereas GLI1 was responsible for the expression of HH-induced key EMT regulators including Snail1, Slug, and Twist, and both GLI1 and GLI2 acted directly as transcriptional repressor of CDH1 gene encoding E-Cadherin. CONCLUSION HH through GLI1 and GLI2 acts as critical signals in supporting the physiological function of mature placenta. GENERAL SIGNIFICANCE HH signaling through GLI1 and GLI2 could be required for the maintenance of human pregnancy.
Collapse
Affiliation(s)
- Chao Tang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liu Mei
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liyu Pan
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenyi Xiong
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haibin Zhu
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongfeng Ruan
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaochun Zou
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lanfang Tang
- The Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Takuma Iguchi
- Department of Toxicology, Osaka University, Suita, Osaka, Japan
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
23
|
Tran HD, Luitel K, Kim M, Zhang K, Longmore GD, Tran DD. Transient SNAIL1 expression is necessary for metastatic competence in breast cancer. Cancer Res 2014; 74:6330-40. [PMID: 25164016 DOI: 10.1158/0008-5472.can-14-0923] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
SNAIL1 has been suggested to regulate breast cancer metastasis based on analyses of human breast tumor transcriptomes and experiments using cancer cell lines and xenografts. However, in vivo genetic experimental support for a role for SNAIL1 in breast cancer metastasis that develops in an immunocompetent tumor microenvironment has not been determined. To address this question, we created a genetic SNAIL1 model by coupling an endogenous SNAIL1 reporter with an inducible SNAIL1 transgene. Using multiple genetic models of breast cancer, we demonstrated that endogenous SNAIL1 expression was restricted to primary tumors that ultimately disseminate. SNAIL1 gene deletion either during the premalignant phase or after primary tumors have reached a palpable size blunted metastasis, indicating that late metastasis was the main driver of metastasis and that this was dependent on SNAIL1. Importantly, SNAIL1 expression during breast cancer metastasis was transient and forced transient, but not continuous. SNAIL1 expression in breast tumors was sufficient to increase metastasis.
Collapse
Affiliation(s)
- Hung D Tran
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Krishna Luitel
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Kim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kun Zhang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Gregory D Longmore
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. ICCE Institute, Washington University School of Medicine, St. Louis, Missouri. Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - David D Tran
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
24
|
An integrative view on the physiology of human early placental villi. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 114:33-48. [PMID: 24291663 DOI: 10.1016/j.pbiomolbio.2013.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/20/2013] [Indexed: 01/08/2023]
Abstract
The placenta is an indispensable organ for intrauterine protection, development and growth of the embryo and fetus. It provides tight contact between mother and conceptus, enabling the exchange of gas, nutrients and waste products. The human placenta is discoidal in shape, and bears a hemo-monochorial interface as well as villous materno-fetal interdigitations. Since Peter Medawar's astonishment to the paradoxical nature of the mother-fetus relationship in 1953, substantial knowledge in the domain of placental physiology has been gathered. In the present essay, an attempt has been made to build an integrated understanding of morphological dynamics, cell biology, and functional aspects of genomic and proteomic expression of human early placental villous trophoblast cells followed by a commentary on the future directions of research in this field.
Collapse
|
25
|
Tande DL, Ralph JL, Johnson LK, Scheett AJ, Hoverson BS, Anderson CM. First trimester dietary intake, biochemical measures, and subsequent gestational hypertension among nulliparous women. J Midwifery Womens Health 2013; 58:423-30. [PMID: 23895215 DOI: 10.1111/jmwh.12007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
INTRODUCTION The purpose of this study was to evaluate the relationships between first-trimester dietary factors and biochemical measures and subsequent risk of gestational hypertension. METHODS This pilot study used a prospective design utilizing a convenience sample of nulliparous women enrolled at their first prenatal visit. A total of 57 women completed the study. Participants were divided into 2 groups for data analysis: normotensive pregnancy and gestational hypertension. RESULTS Nearly one-quarter of study participants (22.8%) developed gestational hypertension, of whom 84.6% had significant proteinuria meeting the criteria for preeclampsia. There were no significant differences in micronutrient or macronutrient dietary intakes between groups. Serum iron and zinc levels were lower for the gestational hypertension group compared with the normotensive pregnancy group (P ≤ .01). Low serum zinc levels were related to a risk of developing gestational hypertension (adjusted odds ratio, 0.930; 95% confidence interval, 0.872-0.992). DISCUSSION Ensuring adequate intake of zinc and monitoring serum zinc levels in nulliparous pregnant women may help to prevent or contribute to early detection of gestational hypertension.
Collapse
Affiliation(s)
- Desiree L Tande
- College of Nursing and Professional Disciplines, University of North Dakota, 430 Oxford Street, Grand Forks, ND 58202-9025, USA
| | | | | | | | | | | |
Collapse
|
26
|
Fedorova LV, Sodhi K, Gatto-Weis C, Puri N, Hinds TD, Shapiro JI, Malhotra D. Peroxisome proliferator-activated receptor δ agonist, HPP593, prevents renal necrosis under chronic ischemia. PLoS One 2013; 8:e64436. [PMID: 23691217 PMCID: PMC3654981 DOI: 10.1371/journal.pone.0064436] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 04/15/2013] [Indexed: 12/15/2022] Open
Abstract
The Goldblatt’s 2 kidney 1 clip (2K1C) rat animal model of renovascular hypertension is characterized by ischemic nephropathy of the clipped kidney. 2K1C rats were treated with a specific peroxisome proliferator-activated receptor δ (PPARδ) agonist, HPP593. Clipped kidneys from untreated rats developed tubular and glomerular necrosis and massive interstitial, periglomerular and perivascular fibrosis. HPP593 kidneys did not exhibit any histochemical features of necrosis; fibrotic lesions were present only in perivascular areas. Necrosis in the untreated clipped kidneys was associated with an increased oxidative stress, up regulation and mitochondrial translocation of the pro-death protein BNIP3 specifically in tubules. In the kidneys of HPP593-treated rats oxidative stress was attenuated and BNIP3 protein decreased notably in the mitochondrial fraction when compared to untreated animals. In untreated clipped kidneys, mitochondria were dysfunctional as revealed by perturbations in the levels of MCAD, COXIV, TFAM, and Parkin proteins and AMPK activation, while in HPP593-treated rats these proteins remained at the physiological levels. Nuclear amounts of oxidative stress-responsive proteins, NRF1 and NRF2 were below physiological levels in treated kidneys. Mitochondrial biogenesis and autophagy were inhibited similarly in both treated and untreated 2K1C kidneys as indicated by a decrease in PGC1-α and deficiency of the autophagy-essential proteins LC3-II and ATG5. However, HPP593 treatment resulted in increased accumulation of p62 protein, an autophagic substrate and an enhancer of NRF2 activity. Therefore, inhibition of BNIP3 activation by the preservation of mitochondrial function and control of oxidative stress by PPARδ is the most likely mechanism to account for the prevention of necrotic death in the kidney under conditions of persistent ischemia.
Collapse
Affiliation(s)
- Larisa V Fedorova
- Department of Medicine, The University of Toledo School of Medicine, Toledo, Ohio, United States of America.
| | | | | | | | | | | | | |
Collapse
|
27
|
Kurlak L, Mistry H, Kaptein E, Visser T, Broughton Pipkin F. Thyroid hormones and their placental deiodination in normal and pre-eclamptic pregnancy. Placenta 2013; 34:395-400. [DOI: 10.1016/j.placenta.2013.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 02/20/2013] [Accepted: 02/24/2013] [Indexed: 12/14/2022]
|