1
|
Wu Z, Zhang L, Jia Y, Bi B, Fang L, Cheng JC. GDF-11 downregulates placental human chorionic gonadotropin expression by activating SMAD2/3 signaling. Cell Commun Signal 2023; 21:179. [PMID: 37480123 PMCID: PMC10362589 DOI: 10.1186/s12964-023-01201-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/17/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND The production of human chorionic gonadotropin (hCG) by the placental trophoblast cells is essential for maintaining a normal pregnancy. Aberrant hCG levels are associated with reproductive disorders. The protein of hCG is a dimer consisting of an α subunit and a β subunit. The β subunit is encoded by the CGB gene and is unique to hCG. Growth differentiation factor-11 (GDF-11), a member of the transforming growth factor-β (TGF-β) superfamily, is expressed in the human placenta and can stimulate trophoblast cell invasion. However, whether the expression of CGB and the production of hCG are regulated by GDF-11 remains undetermined. METHODS Two human choriocarcinoma cell lines, BeWo and JEG-3, and primary cultures of human cytotrophoblast (CTB) cells were used as experimental models. The effects of GDF-11 on CGB expression and hCG production, as well as the underlying mechanisms, were explored by a series of in vitro experiments. RESULTS Our results show that treatment of GDF-11 downregulates the expression of CGB and the production of hCG in both BeWo and JEG-3 cells as well as in primary CTB cells. Using a pharmacological inhibitor and siRNA-mediated approach, we reveal that both ALK4 and ALK5 are required for the GDF-11-induced downregulation of CGB expression. In addition, treatment of GDF-11 activates SMAD2/3 but not SMAD1/5/8 signaling pathways. Moreover, both SMAD2 and SMAD3 are involved in the GDF-11-downregulated CGB expression. ELISA results show that the GDF-11-suppressed hCG production requires the ALK4/5-mediated activation of SMAD2/3 signaling pathways. CONCLUSIONS This study not only discovers the biological function of GDF-11 in the human placenta but also provides important insights into the regulation of the expression of hCG. Video Abstract.
Collapse
Affiliation(s)
- Ze Wu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China
| | - Lingling Zhang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China
| | - Yuanyuan Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, China
| | - Beibei Bi
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, 40, Daxue Road, Zhengzhou, 450052, Henan, 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, 450052, Henan, 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, 450052, Henan, China.
| |
Collapse
|
2
|
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: 1.3] [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
|
3
|
Peng X, Zhang Z, Mo Y, Liu J, Wang S, Liu H. Bioinformatics Analysis of Choriocarcinoma-Related MicroRNA-Transcription Factor-Target Gene Regulatory Networks and Validation of Key miRNAs. Onco Targets Ther 2021; 14:3903-3919. [PMID: 34234459 PMCID: PMC8254590 DOI: 10.2147/ott.s311291] [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: 03/16/2021] [Accepted: 06/15/2021] [Indexed: 11/23/2022] Open
Abstract
Objective The aim of the current research was to construct a miRNA-transcription factor (TF)-target gene regulatory network in order to investigate the mechanism underlying choriocarcinoma and to verify the network through the overexpression or silencing of hub miRNAs in vitro. Materials and Methods A mRNA expression dataset and two miRNA expression datasets were analysed to identify differentially expressed genes (DEGs) and miRNAs (DEMs) between normal cells and choriocarcinoma cells. The top 400 upregulated and downregulated DEGs were identified as candidate DEGs, which were then mapped to construct protein–protein interaction (PPI) networks and select hub genes. Moreover, the DGIdb database was utilized to select candidate drugs for hub genes. Moreover, DEM target genes were predicted through the miRWalk2.0 database and overlaid with candidate DEGs to identify the differentially expressed target genes (DETGs). Furthermore, we established miRNA-TF-target gene regulatory networks and performed functional enrichment analysis of hub DEMs. Finally, we transfected mimics or inhibitors of hub DEMs into choriocarcinoma cells and assessed cell proliferation and migration to verify the vital role of hub DEMs in choriocarcinoma. Results A total of 140 DEMs and 400 candidate DEGs were screened from choriocarcinoma cells and normal cells. A PPI network of 400 candidate DEGs was established. Twenty-nine hub genes and 99 associated small molecules were identified to provide potential target drugs for choriocarcinoma treatment. We obtained 70 DETGs of DEMs derived from the intersection between predicted miRNA target genes and candidate DEGs. Subsequently, 3 hub DEMs were selected, and miRNA-TF-target gene regulatory networks containing 4 TFs, 3 TFs and 3 TFs for each network were constructed. The RT-PCR results confirmed that miR-29b-3p was highly expressed and that miR-519c-3p and miR-520a-5p were expressed at low levels in choriocarcinoma cells. The overexpression or silencing results suggested that 3 dysregulated hub DEMs jointly accelerated the proliferation and migration of choriocarcinoma. Conclusion Association of miRNA-TF-target gene regulatory networks may help us explore the underlying mechanism and provide potential targets for the diagnosis and treatment of choriocarcinoma.
Collapse
Affiliation(s)
- Xiaotong Peng
- Department of Gynaecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Zhirong Zhang
- Department of Gynaecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yanqun Mo
- Department of Gynaecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Junliang Liu
- Department of Gynaecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Shuo Wang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Huining Liu
- Department of Gynaecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| |
Collapse
|
4
|
Bone morphogenetic protein 2 induces the activation of WNT/β-catenin signaling and human trophoblast invasion through up-regulating BAMBI. Cell Signal 2020; 67:109489. [DOI: 10.1016/j.cellsig.2019.109489] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/28/2022]
|
5
|
The role of Galectin-1 in HIV associated preeclampsia. Eur J Obstet Gynecol Reprod Biol 2020; 246:138-144. [PMID: 32018196 DOI: 10.1016/j.ejogrb.2020.01.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE In this study, the role of Gal1, a regulatory protein involved in receptor binding and gene transcription within trophoblast cells, in the pathophysiology of HIV associated preeclampsia was determined by immunolocalizing its expression in the placenta of a South African cohort. STUDY DESIGN this is an analytical study carried out at the Optics and Imaging Center, Neslon R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa. A hundred and twenty HIV negative or positive, Black African primigrad or multigravid women with pre-eclamptic and normotensive pregnancies were involved in the study. Post-delivery, full thickness of centrally located placental tissue obtained was fixed for immunohistochemistry. The expression of Gal1 was immunolocalized using immunohistochemical assay kit and further quantified with using AxioVision Image analysis software package. Student t-test was used to compare the levels of the analytes while One-way ANOVA was used for comparison across the groups. RESULTS Gal1 immunoreactivity was observed within the Hofbauer cells, cytotrophoblast, syncytial knots and in the endothelial cells lining blood vessels in both exchange and conducting villi of both normotensive and preeclamptic pregnancies regardless of HIV status. There was a down regulation in Gal1 immunoreactivity in both the exchange and conducting villi of preeclamptic compared to normotensive pregnancies. However, there was no significant effect of HIV infection on Gal1 immunostaining in both villi types. CONCLUSION The down regulation of Gal1 in preeclampsia may be due to the inhibition of the MAPK pathway. Since Gal1 influences differentiation and migration, the defective trophoblast invasion in preeclampsia may emanate from its decreased immunoexpression. This highlights the role of Gal1 in angiogenesis and placentation.
Collapse
|
6
|
Farrell A, Alahari S, Ermini L, Tagliaferro A, Litvack M, Post M, Caniggia I. Faulty oxygen sensing disrupts angiomotin function in trophoblast cell migration and predisposes to preeclampsia. JCI Insight 2019; 4:127009. [PMID: 30996134 DOI: 10.1172/jci.insight.127009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Human placenta development and a successful pregnancy is incumbent upon precise oxygen-dependent control of trophoblast migration/invasion. Persistent low oxygen leading to failed trophoblast invasion promotes inadequate spiral artery remodeling, a characteristic of preeclampsia. Angiomotin (AMOT) is a multifaceted scaffolding protein involved in cell polarity and migration, yet its upstream regulation and significance in the human placenta remain unknown. Herein, we show that AMOT is primarily expressed in migratory extravillous trophoblast cells (EVTs) of the intermediate and distal anchoring column. Its expression increases after 10 weeks of gestation when oxygen tension rises and EVT migration/invasion peaks. Time-lapse imaging confirmed that the AMOT 80-kDa isoform promotes migration of trophoblastic JEG3 and HTR-8/SVneo cells. In preeclampsia, however, AMOT expression is decreased and its localization to migratory fetomaternal interface EVTs is disrupted. We demonstrate that Jumonji C domain-containing protein 6 (JMJD6), an oxygen sensor, positively regulates AMOT via oxygen-dependent lysyl hydroxylation. Furthermore, in vitro and ex vivo studies show that transforming growth factor-β (TGF-β) regulates AMOT expression, its interaction with polarity protein PAR6, and its subcellular redistribution from tight junctions to cytoskeleton. Our data reveal an oxygen- and TGF-β-driven migratory function for AMOT in the human placenta, and implicate its deficiency in impaired trophoblast migration that plagues preeclampsia.
Collapse
Affiliation(s)
- Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrea Tagliaferro
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michael Litvack
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Post
- Institute of Medical Sciences, and.,Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Shih J, Lin H, Hsiao A, Su Y, Tsai S, Chien C, Kung H. Unveiling the role of microRNA‐7 in linking TGF‐β‐Smad‐mediated epithelial‐mesenchymal transition with negative regulation of trophoblast invasion. FASEB J 2019; 33:6281-6295. [DOI: 10.1096/fj.201801898rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jin‐Chung Shih
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
- Graduate Institute of Medical Genomics and ProteomicsCollege of MedicineNational Taiwan University Taipei Taiwan
| | - Hua‐Heng Lin
- Department of Obstetrics and GynecologyCollege of MedicineNational Taiwan University Hospital Taipei Taiwan
| | - An‐Che Hsiao
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
| | - Yi‐Ting Su
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
| | - Shawn Tsai
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
| | - Chung‐Liang Chien
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
| | - Hsiu‐Ni Kung
- Graduate Institute of Anatomy and Cell BiologyCollege of MedicineNational Taiwan University Taipei Taiwan
| |
Collapse
|
8
|
Interactions of NK Cells and Trophoblast Cells. Methodological Aspects. Bull Exp Biol Med 2018; 165:548-553. [PMID: 30121921 DOI: 10.1007/s10517-018-4212-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Indexed: 12/28/2022]
Abstract
NK cells present in different organs differ by their functional characteristics, in particular, proliferative activity. For studying tissue-resident NK cells, tissue-specific microenvironment should be reproduced. In case of decidual NK cells, this microenvironment is created by trophoblast cells. We developed a method for evaluation of proliferative activity of peripheral blood NK cells in the presence of trophoblast cells. Proliferative activity of peripheral blood NK cells was evaluated by the expression of protein Ki-67 after culturing with JEG-3 trophoblast cells. This method allows evaluating the functional state of NK cells in microenvironment specific for the decidua.
Collapse
|
9
|
Malik A, Pal R, Gupta SK. Interdependence of JAK-STAT and MAPK signaling pathways during EGF-mediated HTR-8/SVneo cell invasion. PLoS One 2017; 12:e0178269. [PMID: 28542650 PMCID: PMC5444796 DOI: 10.1371/journal.pone.0178269] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/10/2017] [Indexed: 12/03/2022] Open
Abstract
Invasion of trophoblast cells is spatio-temporally regulated by various cytokines and growth factors. In pregnancy, complications like preeclampsia, shallow invasion of trophoblast cells and low amounts of epidermal growth factor (EGF) have been reported. In the present study, regulatory mechanisms associated with EGF-mediated invasion in HTR-8/SVneo trophoblastic cells have been delineated. Treatment of HTR-8/SVneo cells with EGF (10 ng/ml) led to eight fold increase (p < 0.05) in invasion. Increased invasion of HTR-8/SVneo cells by EGF was associated with an increase in phosphorylation of ERK½. In addition, significant phosphorylation of STAT1 (ser 727) and STAT3 (both tyr 705 and ser 727 residues) was also observed, accompanied by a decrease in total STAT1. Inhibition of ERK½ phosphorylation by U0126 (10 μM) led to a significant decrease in EGF-mediated invasion with simultaneous decrease in the phosphorylated forms of STAT3 and STAT1. Decrease in total STAT1 was also reversed on inhibition of ERK½. Interestingly, inhibition of STAT3 by siRNA led to a significant decrease in EGF-mediated invasion of HTR-8/SVneo cells and phosphorylation of STAT1, but it did not have any effect on the activation of ERK½. On the other hand, inhibition of STAT1 by siRNA, also led to a significant decrease in the EGF-mediated invasion of HTR-8/SVneo cells, showed concomitant decrease in ERK½ phosphorylation and STAT3 phosphorylation at ser 727 residue. These results suggest cross-communication between ERK½ and JAK-STAT pathways during EGF-mediated increase in invasion of trophoblast cells; phosphorylation at ser 727 residue of both STAT3 and STAT1 appears to be critical.
Collapse
Affiliation(s)
- Ankita Malik
- Reproductive Cell Biology Laboratory, National Institute of Immunology, New Delhi, Delhi, India
| | - Rahul Pal
- Immunoendocrinology Laboratory, National Institute of Immunology, New Delhi, Delhi, India
| | - Satish Kumar Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, New Delhi, Delhi, India
- * E-mail:
| |
Collapse
|
10
|
Namgung S, Yoon JJ, Yoon CS, Han BH, Choi ES, Oh H, Kim YC, Lee YJ, Kang DG, Lee HS. Prunella vulgaris Attenuates Diabetic Renal Injury by Suppressing Glomerular Fibrosis and Inflammation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:475-495. [PMID: 28359196 DOI: 10.1142/s0192415x1750029x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetic nephropathy is both the most common complication and the leading cause of mortality associated with diabetes. Prunella vulgaris, a well-known traditional medicinal plant, is used for the cure of abscess, scrofula, hypertension and urinary diseases. This study confirmed whether an aqueous extract of Prunella vulgaris (APV) suppresses renal inflammation and fibrosis. In human mesangial cell (HMC), pretreatment of APV attenuated 25[Formula: see text]mM HG-induced suppressed TGF-[Formula: see text] and Smad-2/4 expression; it increased the expression level of Smad-7. Connective tissue growth factor (CTGF) and collagen IV, fibrosis biomarkers, were significantly decreased by APV. APV suppressed inflammatory factors such as intracellular cell adhesion molecule-1 (ICAM-1) and monocyte chemoattractant protein-1 (MCP-1). APV inhibited activation and translocation of nuclear factor kappa-B (NF-[Formula: see text]B) in HG-stimulated HMCs. Moreover, APV significantly improved HG-induced ROS in a dose-dependent manner. In diabetic rat models, APV significantly decreased blood glucose, blood urea nitrogen (BUN) and ameliorated plasma creatinine (PCr). APV reduced the PAS positivity staining intensity and basement membrane thickening in glomeruli of diabetic rats. Fibrosis related proteins such as collagen IV and TGF-[Formula: see text]1 were also inhibited by APV. These results suggest that APV has a significant protective effect against diabetic renal dysfunction including inflammation and fibrosis through disruption of the TGF-[Formula: see text]/Smad signaling. Therefore, APV may be useful in potential therapies that target glomerulonephritis and glomerulosclerosis, which lead to diabetic nephropathy.
Collapse
Affiliation(s)
- Seung Namgung
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jung Joo Yoon
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Chi-Su Yoon
- † Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,‡ College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Byung Hyuk Han
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Eun Sik Choi
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hyuncheol Oh
- † Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,‡ College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Youn-Chul Kim
- † Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,‡ College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yun Jung Lee
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dae Gill Kang
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ho Sub Lee
- * College of Oriental Medicine and Professional, Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea.,† Hanbang Body-fluid Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
11
|
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: 22.6] [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
|
12
|
Lala PK, Nandi P. Mechanisms of trophoblast migration, endometrial angiogenesis in preeclampsia: The role of decorin. Cell Adh Migr 2016; 10:111-25. [PMID: 26745663 DOI: 10.1080/19336918.2015.1106669] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The objective of the present review is to synthesize the information on the cellular and molecular players responsible for maintaining a homeostatic balance between a naturally invasive human placenta and the maternal uterus in pregnancy; to review the roles of decorin (DCN) as a molecular player in this homeostasis; to list the common maladies associated with a break-down in this homeostasis, resulting from a hypo-invasive or hyper-invasive placenta, and their underlying mechanisms. We show that both the fetal components of the placenta, represented primarily by the extravillous trophoblast, and the maternal component represented primarily by the decidual tissue and the endometrial arterioles, participate actively in this balance. We discuss the process of uterine angiogenesis in the context of uterine arterial changes during normal pregnancy and preeclampsia. We compare and contrast trophoblast growth and invasion with the processes involved in tumorigenesis with special emphasis on the roles of DCN and raise important questions that remain to be addressed. Decorin (DCN) is a small leucine-rich proteoglycan produced by stromal cells, including dermal fibroblasts, chondrocytes, chorionic villus mesenchymal cells and decidual cells of the pregnant endometrium. It contains a 40 kDa protein core having 10 leucine-rich repeats covalently linked with a glycosaminoglycan chain. Biological functions of DCN include: collagen assembly, myogenesis, tissue repair and regulation of cell adhesion and migration by binding to ECM molecules or antagonising multiple tyrosine kinase receptors (TKR) including EGFR, IGF-IR, HGFR and VEGFR-2. DCN restrains angiogenesis by binding to thrombospondin-1, TGFβ, VEGFR-2 and possibly IGF-IR. DCN can halt tumor growth by antagonising oncogenic TKRs and restraining angiogenesis. DCN actions at the fetal-maternal interface include restraint of trophoblast migration, invasion and uterine angiogenesis. We demonstrate that DCN overexpression in the decidua is associated with preeclampsia (PE); this may have a causal role in PE by compromising endovascular differentiation of the trophoblast and uterine angiogenesis, resulting in poor arterial remodeling. Elevated DCN level in the maternal blood is suggested as a potential biomarker in PE.
Collapse
Affiliation(s)
- Peeyush K Lala
- a Department of Anatomy and Cell Biology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada.,b Department of Oncology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada.,c Chidren's Health Research Institute, Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada
| | - Pinki Nandi
- a Department of Anatomy and Cell Biology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada
| |
Collapse
|
13
|
Nandi P, Siddiqui MF, Lala PK. Restraint of Trophoblast Invasion of the Uterus by Decorin: Role in Pre-eclampsia. Am J Reprod Immunol 2015; 75:351-60. [DOI: 10.1111/aji.12449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/16/2015] [Indexed: 11/28/2022] Open
Affiliation(s)
- Pinki Nandi
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| | - Mohammad Fyyaz Siddiqui
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| | - Peeyush K Lala
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
- Department of Oncology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
- Children's Health Research Institute; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| |
Collapse
|
14
|
Abstract
The hydatidiform mole (HM) is a placental pathology of androgenetic origin. Placental villi have an abnormal hyperproliferation event and hydropic degeneration. Three situations can be envisaged at its origin: 1. The destruction/expulsion of the female pronucleus at the time of fertilization by 1 or 2 spermatozoa with the former being followed by an endoreplication of the male pronucleus leading to a complete hydatidiform mole (CHM) 2. A triploid zygote (fertilization by 2 spermatozoa) leading to a partial hydatidiform mole (PHM) but can also lead to haploid and diploid clones. The diploid clone may produce a normal fetus while the haploid clone after endoreplication generates a CHM 3. A nutritional defect during the differentiation of the oocytes or the deterioration of the limited oxygen pressure during the first trimester of gestation may lead to the formation of a HM. In countries with poor medical health care system, moles (mainly the CHM) can become invasive or, in rare cases, lead to gestational choriocarcinomas.
Collapse
Affiliation(s)
- Jean-Jacques Candelier
- a Unité 1197 INSERM, Stem cell-niches Interactions: Physiology , Tumors and Tissular Repair, Hôpital Paul Brousse, Bâtiment Lavoisier , Villejuif , France.,b University of Paris-Saclay , Saint-Aubin , France
| |
Collapse
|
15
|
Babwah AV. Uterine and placental KISS1 regulate pregnancy: what we know and the challenges that lie ahead. Reproduction 2015; 150:R121-8. [PMID: 26183891 DOI: 10.1530/rep-15-0252] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/16/2015] [Indexed: 01/11/2023]
Abstract
Hypothalamic KISS1 and its derivatives (kisspeptins) are now well recognized as potent stimulators of GnRH secretion and thereby major regulators of the neuroendocrine-reproductive axis. Recent studies in the mouse strongly suggest that independent of the hypothalamus and pituitary, peripherally derived KISS1 also regulates fertility, and disruption of local KISS1 signaling in the ovary and uterus is sufficient to trigger infertility. With this increasing recognition that peripherally derived KISS1 regulates fertility, the first goal of this review is to critically discuss the data that have led to this conclusion, focusing on uterine- and placental-derived KISS1. Given that a significant amount of this data was generated in animals such as the mouse and rat, a second goal of this review is to identify and discuss the limitations of the animal data in the context of better understanding KISS1 as a regulator of human pregnancy. The growing evidence suggests that in both man and mouse, KISS1 plays an important role in regulating very early pregnancy events such as embryo implantation. However, as pregnancy advances, although it seems that KISS1 continues to play important roles in regulating human pregnancy, it might not do so in the mouse. This surprising functional dichotomy between human females and mice appears also to exist between women and a large number of animal species, including lower primates. These findings are of tremendous significance and will greatly shape how KISS1 will be developed as a therapeutic agent in augmenting the reproductive potential of both women and important livestock species.
Collapse
Affiliation(s)
- Andy V Babwah
- The Children's Health Research InstituteLawson Health Research InstituteDepartments of Obstetrics and Gynaecology and Physiology and PharmacologyThe University of Western Ontario, 800 Commissioners Road East, London, Ontario, Canada N6C 2V5 The Children's Health Research InstituteLawson Health Research InstituteDepartments of Obstetrics and Gynaecology and Physiology and PharmacologyThe University of Western Ontario, 800 Commissioners Road East, London, Ontario, Canada N6C 2V5 The Children's Health Research InstituteLawson Health Research InstituteDepartments of Obstetrics and Gynaecology and Physiology and PharmacologyThe University of Western Ontario, 800 Commissioners Road East, London, Ontario, Canada N6C 2V5
| |
Collapse
|
16
|
Cheng JC, Chang HM, Leung PCK. Transforming growth factor-β1 inhibits trophoblast cell invasion by inducing Snail-mediated down-regulation of vascular endothelial-cadherin protein. J Biol Chem 2013; 288:33181-92. [PMID: 24106276 DOI: 10.1074/jbc.m113.488866] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human trophoblast cells express transforming growth factor-β (TGF-β) and TGF-β receptors. It has been shown that TGF-β1 treatment decreases the invasiveness of trophoblast cells. However, the molecular mechanisms underlying TGF-β1-decreased trophoblast invasion are still not fully understood. In the current study, we demonstrated that treatment of HTR-8/SVneo human trophoblast cells with TGF-β1 decreased cell invasion and down-regulated the expression of vascular endothelial cadherin (VE-cadherin). In addition, the inhibitory effect of TGF-β1 on VE-cadherin was confirmed in primary cultures of human trophoblast cells. Moreover, knockdown of VE-cadherin using siRNA decreased the invasiveness of HTR-8/SVneo cells and primary cultures of trophoblast cells. Treatment with TGF-β1 induced the activation of Smad-dependent signaling pathways and the expression of Snail and Slug. Knockdown of Smads attenuated TGF-β1-induced up-regulation of Snail and Slug and down-regulation of VE-cadherin. Interestingly, depletion of Snail, but not Slug, attenuated TGF-β1-induced down-regulation of VE-cadherin. Furthermore, overexpression of Snail suppressed VE-cadherin expression. Chromatin immunoprecipitation analyses showed the direct binding of Snail to the VE-cadherin promoter. These results provide evidence that Snail mediates TGF-β1-induced down-regulation of VE-cadherin, which subsequently contributed to TGF-β1-decreased trophoblast cell invasion.
Collapse
Affiliation(s)
- Jung-Chien Cheng
- From the Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V6H 3V5, Canada
| | | | | |
Collapse
|
17
|
Zhao H, Jiang Y, Cao Q, Hou Y, Wang C. Role of Integrin Switch and Transforming Growth Factor Beta 3 in Hypoxia-Induced Invasion Inhibition of Human Extravillous Trophoblast Cells1. Biol Reprod 2012; 87:47. [DOI: 10.1095/biolreprod.112.099937] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
18
|
Suman P, Godbole G, Thakur R, Morales-Prieto DM, Modi DN, Markert UR, Gupta SK. AP-1 transcription factors, mucin-type molecules and MMPs regulate the IL-11 mediated invasiveness of JEG-3 and HTR-8/SVneo trophoblastic cells. PLoS One 2012; 7:e29745. [PMID: 22235337 PMCID: PMC3250480 DOI: 10.1371/journal.pone.0029745] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/02/2011] [Indexed: 12/18/2022] Open
Abstract
This study examines the IL-11 mediated activation of downstream signaling and expression of effector molecules to resolve the controversies associated with the IL-11 mediated regulation of the invasiveness of two commonly used trophoblastic cell models viz. JEG-3 and HTR-8/SVneo cells. It has been reported that IL-11 increases the invasiveness of JEG-3 cells while, reduces the invasiveness of HTR-8/SVneo cells. Invasion assay performed simultaneously for both the cell lines confirmed the above findings. In addition, HTR-8/SVneo cells showed a higher basal invasiveness than JEG-3 cells. Western blot showed the IL-11 mediated activation of STAT3(tyr705) and STAT1(tyr701) in both the cell lines. However, IL-11 activated the ERK1/2 phosphorylation in JEG-3 cells but, inhibited it in HTR-8/SVneo cells. Within 10 min of IL-11 treatment, p-STAT3(tyr705) was localized inside the nucleus of both the cell lines but, there was enhanced co-localization of protein inhibitor of activated STAT1/3 (PIAS1/3) and p-STAT3(tyr705) in HTR-8/SVneo cells and not in JEG-3 cells. This could be reason for the poor responsiveness of STAT3 responsive genes like mucin 1 (MUC1) in HTR-8/SVneo cells and not in JEG-3 cells. Further, microarray analysis of the IL-11 treated cells revealed differential responsiveness of JEG-3 as compared to HTR-8/SVneo cells. Several family of genes like activator protein-1 (AP-1) transcription factors (Jun and Fos), mucin-type molecules, MMP23B etc showed enhanced expression in IL-11 treated JEG-3 cells while, there was no response or decrease in their expression in IL-11 treated HTR-8/SVneo cells. Expression of these molecules was confirmed by quantitative RT-PCR. In addition, HTR-8/SVneo cells also showed a significant decrease in the expression of MMP2, MMP3 and MMP9 upon IL-11 treatment. Hence, IL-11 mediated differential activation of signaling and expression of effector molecules is responsible for the differential invasive response of JEG-3 and HTR-8/SVneo cells.
Collapse
Affiliation(s)
- Pankaj Suman
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Geeta Godbole
- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, India
| | - Ravi Thakur
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Diana M. Morales-Prieto
- Placenta Laboratory, Department of Obstetrics, Faculty of Medicine, Friedrich-Schiller University, Jena, Germany
| | - Deepak N. Modi
- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, India
| | - Udo R. Markert
- Placenta Laboratory, Department of Obstetrics, Faculty of Medicine, Friedrich-Schiller University, Jena, Germany
| | - Satish K. Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
- * E-mail:
| |
Collapse
|
19
|
Schiro MM, Stauber SE, Peterson TL, Krueger C, Darnell SJ, Satyshur KA, Drinkwater NR, Newton MA, Hoffmann FM. Mutations in protein-binding hot-spots on the hub protein Smad3 differentially affect its protein interactions and Smad3-regulated gene expression. PLoS One 2011; 6:e25021. [PMID: 21949838 PMCID: PMC3176292 DOI: 10.1371/journal.pone.0025021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/22/2011] [Indexed: 02/06/2023] Open
Abstract
Background Hub proteins are connected through binding interactions to many other proteins. Smad3, a mediator of signal transduction induced by transforming growth factor beta (TGF-β), serves as a hub protein for over 50 protein-protein interactions. Different cellular responses mediated by Smad3 are the product of cell-type and context dependent Smad3-nucleated protein complexes acting in concert. Our hypothesis is that perturbation of this spectrum of protein complexes by mutation of single protein-binding hot-spots on Smad3 will have distinct consequences on Smad3-mediated responses. Methodology/Principal Findings We mutated 28 amino acids on the surface of the Smad3 MH2 domain and identified 22 Smad3 variants with reduced binding to subsets of 17 Smad3-binding proteins including Smad4, SARA, Ski, Smurf2 and SIP1. Mutations defective in binding to Smad4, e.g., D408H, or defective in nucleocytoplasmic shuttling, e.g., W406A, were compromised in modulating the expression levels of a Smad3-dependent reporter gene or six endogenous Smad3-responsive genes: Mmp9, IL11, Tnfaip6, Fermt1, Olfm2 and Wnt11. However, the Smad3 mutants Y226A, Y297A, W326A, K341A, and E267A had distinct differences on TGF-β signaling. For example, K341A and Y226A both reduced the Smad3-mediated activation of the reporter gene by ∼50% but K341A only reduced the TGF-β inducibilty of Olfm2 in contrast to Y226A which reduced the TGF-β inducibility of all six endogenous genes as severely as the W406A mutation. E267A had increased protein binding but reduced TGF-β inducibility because it caused higher basal levels of expression. Y297A had increased TGF-β inducibility because it caused lower Smad3-induced basal levels of gene expression. Conclusions/Significance Mutations in protein binding hot-spots on Smad3 reduced the binding to different subsets of interacting proteins and caused a range of quantitative changes in the expression of genes induced by Smad3. This approach should be useful for unraveling which Smad3 protein complexes are critical for specific biological responses.
Collapse
Affiliation(s)
- Michelle M. Schiro
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sara E. Stauber
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Tami L. Peterson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Chateen Krueger
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Steven J. Darnell
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kenneth A. Satyshur
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Norman R. Drinkwater
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Michael A. Newton
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - F. Michael Hoffmann
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
20
|
Lu J, Aggarwal R, Kanji S, Das M, Joseph M, Pompili V, Das H. Human ovarian tumor cells escape γδ T cell recognition partly by down regulating surface expression of MICA and limiting cell cycle related molecules. PLoS One 2011; 6:e23348. [PMID: 21935360 PMCID: PMC3173356 DOI: 10.1371/journal.pone.0023348] [Citation(s) in RCA: 23] [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: 05/13/2011] [Accepted: 07/12/2011] [Indexed: 11/29/2022] Open
Abstract
Background Mechanisms of human Vγ2Vδ2 T cell-mediated tumor immunity have yet to be fully elucidated. Methods and Findings At least some tumor cell recognition is mediated by NKG2D-MICA interactions. Herein, by using MTT assay and PI-BrdU co-staining and Western-blot, we show that these Vγ2Vδ2 T cells can limit the proliferation of ovarian tumor cells by down regulation of apoptosis and cell cycle related molecules in tumor cells. Cell-to-cell contact is critical. γδ T cell-resistant, but not susceptible ovarian tumor cells escape γδ T cell-mediated immune recognition by up-regulating pErk1/2, thereby decreasing surface MICA levels. Erk1/2 inhibitor pretreatment or incubation prevents this MICA decrease, while up-regulating key cell cycle related molecules such as CDK2, CDK4 and Cyclin D1, as well as apoptosis related molecules making resistant tumor cells now vulnerable to γδ T cell-mediated lysis. Conclusion These findings demonstrate novel effects of γδT cells on ovarian tumor cells.
Collapse
Affiliation(s)
- Jingwei Lu
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Reeva Aggarwal
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Suman Kanji
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Manjusri Das
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew Joseph
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Vincent Pompili
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Hiranmoy Das
- Cardiovascular Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
- Innate Immunity, Comprehensive Cancer Center, Richard J. Solove Research Institute, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
21
|
The role of placental homeobox genes in human fetal growth restriction. J Pregnancy 2011; 2011:548171. [PMID: 21547091 PMCID: PMC3087155 DOI: 10.1155/2011/548171] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 02/17/2011] [Indexed: 12/04/2022] Open
Abstract
Fetal growth restriction (FGR) is an adverse pregnancy outcome associated with significant perinatal and paediatric morbidity and mortality, and an increased risk of chronic disease later in adult life. One of the key causes of adverse pregnancy outcome is fetal growth restriction (FGR). While a number of maternal, fetal, and environmental factors are known causes of FGR, the majority of FGR cases remain idiopathic. These idiopathic FGR pregnancies are frequently associated with placental insufficiency, possibly as a result of placental maldevelopment. Understanding the molecular mechanisms of abnormal placental development in idiopathic FGR is, therefore, of increasing importance. Here, we review our understanding of transcriptional control of normal placental development and abnormal placental development associated with human idiopathic FGR. We also assess the potential for understanding transcriptional control as a means for revealing new molecular targets for the detection, diagnosis, and clinical management of idiopathic FGR.
Collapse
|
22
|
Papadimitriou E, Kardassis D, Moustakas A, Stournaras C. TGFβ-induced Early Activation of the Small GTPase RhoA is Smad2/3-independent and Involves Src and the Guanine Nucleotide Exchange Factor Vav2. Cell Physiol Biochem 2011; 28:229-38. [DOI: 10.1159/000331734] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2011] [Indexed: 12/12/2022] Open
|
23
|
Rajaraman G, Murthi P, Brennecke SP, Kalionis B. Homeobox Gene HLX Is a Regulator of HGF/c-met-Mediated Migration of Human Trophoblast-Derived Cell Lines1. Biol Reprod 2010; 83:676-83. [DOI: 10.1095/biolreprod.109.078634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
24
|
Matsui S, Takahashi C, Tsujimoto Y, Matsushima K. Stimulatory Effects of Low-concentration Reactive Oxygen Species on Calcification Ability of Human Dental Pulp Cells. J Endod 2009; 35:67-72. [DOI: 10.1016/j.joen.2008.08.034] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2008] [Revised: 08/18/2008] [Accepted: 08/24/2008] [Indexed: 12/28/2022]
|
25
|
Gordon KJ, Kirkbride KC, How T, Blobe GC. Bone morphogenetic proteins induce pancreatic cancer cell invasiveness through a Smad1-dependent mechanism that involves matrix metalloproteinase-2. Carcinogenesis 2008; 30:238-48. [PMID: 19056927 DOI: 10.1093/carcin/bgn274] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) have an emerging role in human cancers. Here we demonstrate that the BMP-signaling pathway is intact and functional in human pancreatic cancer cells, with several BMP signaling components and transcriptional targets upregulated in human pancreatic cancer specimens compared with normal pancreatic tissue. Functionally, multiple BMP family members, including BMP-2, BMP-4 and BMP-7, induce an epithelial to mesenchymal transition (EMT) in the human pancreatic cancer cell line Panc-1, as demonstrated by morphological alterations and loss of E-cadherin expression. BMP-mediated EMT results in an increase in invasiveness of Panc-1 cells, in part through increased expression and activity of matrix metalloproteinase (MMP)-2, a known mediator of pancreatic cancer cell invasiveness. Accompanying EMT, BMP reduces expression of the transforming growth factor (TGF)-beta superfamily receptor, transforming growth factor-beta type III receptor (TbetaRIII), for which we have previously demonstrated loss of expression during pancreatic cancer progression. Maintaining TbetaRIII expression inhibits BMP-mediated invasion and suppresses Smad1 activation. Further, Smad1 is required for BMP-induced invasiveness and partially responsible for BMP-mediated increases in MMP-2 activity. These data suggest that BMP signaling, through Smad1 induction and upregulation of MMP-2, is an important mediator of pancreatic cancer invasiveness and a potential therapeutic target for treating this deadly disease.
Collapse
Affiliation(s)
- Kelly J Gordon
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
26
|
Iacob D, Cai J, Tsonis M, Babwah A, Chakraborty C, Bhattacharjee RN, Lala PK. Decorin-mediated inhibition of proliferation and migration of the human trophoblast via different tyrosine kinase receptors. Endocrinology 2008; 149:6187-97. [PMID: 18703624 DOI: 10.1210/en.2008-0780] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Decorin (DCN), a decidua-derived TGFbeta-binding proteoglycan, negatively regulates proliferation, migration, and invasiveness of human extravillous trophoblast (EVT) cells in a TGFbeta-independent manner. The present study examined underlying mechanisms, in particular possible roles of epidermal growth factor receptor (EGFR), IGF receptor (IGFR)-I, and vascular endothelial growth factor receptor (VEGFR)-2. EVT cell sprouting from first-trimester chorionic villus explants in the presence or absence of TGFbeta-neutralizing antibody was inhibited with DCN, suggesting its negative regulatory role in situ. Inhibition of migration of the human EVT cell line HTR-8/SVneo in transwells undercoated with fibronectin was stronger when cells were briefly preincubated with DCN at 4 C (known to retard dissociation of receptor-ligand complex) than at 37 C, suggesting possible DCN action by cell membrane binding. Pretreatment of cells with an IGFR-I blocking agent, but not two EGFR blocking agents or a VEGFR blocking agent, significantly abrogated migration inhibitory effects of DCN, suggesting the involvement of IGFR-I but not EGFR or VEGFR in migration inhibition by DCN. On the other hand, pretreatment with either of the EGFR blocking agents, or the VEGFR blocking agent but not the IGFR-I blocking agent, blocked proliferation inhibitory effects of DCN, indicating the roles of EGFR and VEGFR, but not IGFR-I in antiproliferative action of DCN. EVT cells expressed EGFR, IGFR-I, and VEGFR-2. IGFR-I and VEGF-R2 were phosphorylated in the presence of their natural ligands as well as DCN, and these events were blocked by pretreatment with respective receptor blocking agents indicating DCN-mediated activation of these receptors. In conclusion, DCN effects on EVT cells are mediated selectively by multiple tyrosine kinase receptors.
Collapse
Affiliation(s)
- D Iacob
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | | | | | | | |
Collapse
|
27
|
Baek HJ, Lim SC, Kitisin K, Jogunoori W, Tang Y, Marshall MB, Mishra B, Kim TH, Cho KH, Kim SS, Mishra L. Hepatocellular cancer arises from loss of transforming growth factor beta signaling adaptor protein embryonic liver fodrin through abnormal angiogenesis. Hepatology 2008; 48:1128-37. [PMID: 18704924 PMCID: PMC2747753 DOI: 10.1002/hep.22460] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
UNLABELLED We have previously demonstrated that 40%-70% of elf(+/-) mice spontaneously develop hepatocellular cancer (HCC) within 15 months, revealing the importance of the transforming growth factor-beta (TGF-beta) signaling pathway in suppressing tumorigenesis in the liver. The current study was carried out to investigate mechanisms by which embryonic liver fodrin (ELF), a crucial Smad3/4 adaptor, suppresses liver tumor formation. Histological analysis of hyperplastic liver tissues from elf(+/-) mice revealed abundant newly formed vascular structures, suggesting aberrant angiogenesis with loss of ELF function. In addition, elf(+/-) mice displayed an expansion of endothelial progenitor cells. Ectopic ELF expression in fetal bovine heart endothelial (FBHE) cells resulted in cell cycle arrest and apoptosis. Further analysis of developing yolk sacs of elf(-/-) mice revealed a failure of normal vasculature and significantly decreased endothelial cell differentiation with embryonic lethality. Immunohistochemical analysis of hepatocellular cancer (HCC) from the elf(+/-) mice revealed an abnormal angiogenic profile, suggesting the role of ELF as an angiogenic regulator in suppressing HCC. Lastly, acute small interfering RNA (siRNA) inhibition of ELF raised retinoblastoma protein (pRb) levels nearly fourfold in HepG2 cells (a hepatocellular carcinoma cell line) as well as in cow pulmonary artery endothelial (CPAE) cells, respectively. CONCLUSION Taken together these results, ELF, a TGF-beta adaptor and signaling molecule, functions as a critical adaptor protein in TGF-beta modulation of angiogenesis as well as cell cycle progression. Loss of ELF in the liver leads the cancer formation by deregulated hepatocyte proliferation and stimulation of angiogenesis in early cancers. Our studies propose that ELF is potentially a powerful target for mimetics enhancing the TGF-beta pathway tumor suppression of HCC.
Collapse
Affiliation(s)
- Hye Jung Baek
- Radiation Medicine Branch, National Cancer Center, Goyang, Korea
| | - Sung Chul Lim
- Department of Pathology, Research Center for Resistant Cells, College of Medicine, Chosun University, Gwangju, Korea
| | - Krit Kitisin
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
| | - Wilma Jogunoori
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
| | - Yi Tang
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
| | - M. Blair Marshall
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
| | - Bibhuti Mishra
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
| | - Tae Hyun Kim
- Radiation Medicine Branch, National Cancer Center, Goyang, Korea
| | - Kwan Ho Cho
- Radiation Medicine Branch, National Cancer Center, Goyang, Korea
| | - Sang Soo Kim
- Radiation Medicine Branch, National Cancer Center, Goyang, Korea
| | - Lopa Mishra
- Department of Surgery, Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Lombardi Cancer Center, Georgetown University, Washington, DC
- Department of Veterans Affairs, Washington, DC
| |
Collapse
|
28
|
Ripamonti U, Ramoshebi LN, Teare J, Renton L, Ferretti C. The induction of endochondral bone formation by transforming growth factor-beta(3): experimental studies in the non-human primate Papio ursinus. J Cell Mol Med 2008; 12:1029-48. [PMID: 18494943 PMCID: PMC4401141 DOI: 10.1111/j.1582-4934.2008.00126.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Transforming growth factor-β3 (TGF-β3), a multi-functional growth modulator of embryonic development, tissue repair and morphogenesis, immunoregulation, fibrosis, angiogenesis and carcinogenesis, is the third mammalian isoform of the TGF-β subfamily of proteins. The pleiotropism of the signalling proteins of the TGF-β superfamily, including the TGF-β proteins per se, are highlighted by the apparent redundancy of soluble molecular signals initiating de novo endochondral bone induction in the primate only. In the heterotopic bioassay for bone induction in the subcutaneous site of rodents, the TGF-β3 isoform does not initiate endochondral bone formation. Strikingly and in marked contrast to the rodent bioassay, recombinant human (h)TGF-β3, when implanted in the rectus abdominis muscle of adult non-human primates Papio ursinus at doses of 5, 25 and 125 μg per 100 mg of insoluble collagenous matrix as carrier, induces rapid endochondral bone formation resulting in large corticalized ossicles by day 30 and 90. In the same animals, the delivery of identical or higher doses of theTGF-β3 protein results in minimal repair of calvarial defects on day 30 with limited bone regeneration across the pericranial aspect of the defects on day 90. Partial restoration of the bone induction cascade by the hTGF-β3 protein is obtained by mixing the hTGF-β3 device with minced fragments of autogenous rectus abdominis muscle thus adding responding stem cells for further bone induction by the hTGF-β3 protein. The observed limited bone induction in hTGF-β3/treated and untreated calvarial defects in Papio ursinus and therefore by extension to Homo sapiens, is due to the influence of Smad-6 and Smad-7 down-stream antagonists of the TGF-β signalling pathway. RT-PCR, Western and Northern blot analyses of tissue specimens generated by the TGF-β3 isoform demonstrate robust expression of Smad-6 and Smad-7 in orthotopic calvarial sites with limited expression in heterotopic rectus abdominis sites. Smad-6 and -7 overexpression in hTGF-β3/treated and untreated calvarial defects may be due to the vascular endothelial tissue of the arachnoids expressing signalling proteins modulating the expression of the inhibitory Smads in pre-osteoblastic and osteoblastic calvarial cell lines controlling the induction of bone in the primate calvarium.
Collapse
Affiliation(s)
- Ugo Ripamonti
- Bone Research Unit, Medical Research Council/University of the Witwatersrand, Johannesburg, South Africa.
| | | | | | | | | |
Collapse
|
29
|
Vardouli L, Vasilaki E, Papadimitriou E, Kardassis D, Stournaras C. A novel mechanism of TGFβ-induced actin reorganization mediated by Smad proteins and Rho GTPases. FEBS J 2008; 275:4074-87. [DOI: 10.1111/j.1742-4658.2008.06549.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Matsui S, Takeuchi H, Tsujimoto Y, Matsushima K. Effects of Smads and BMPs induced by Ga-Al-As laser irradiation on calcification ability of human dental pulp cells. J Oral Sci 2008; 50:75-81. [DOI: 10.2334/josnusd.50.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
31
|
Abstract
When inundated with numerous specimens of products of conception as the consequence of miscarriage, it is all too easy for histopathologists to forget that the biology of trophoblast and the events of early placental implantation continue to fascinate because of the inherently invasive properties of the non-villous (extravillous) trophoblast. However, unlike the invasion of a malignant tumour, the invasion of trophoblast is controlled. The failure of adequate conversion of maternal uteroplacental arteries is a major pathogenetic phenomenon of important disorders of pregnancy including pre-eclampsia. However, it is in the field of gestational trophoblastic disease that diagnostic acumen is most called for. There are several problematic areas that give rise to diagnostic error; e.g., the diagnosis of early complete mole as partial mole, the over-diagnosis of hydatidiform mole in tubal pregnancy and the diagnosis of placental site non-villous trophoblast as placental site trophoblastic tumour or choriocarcinoma, particularly if associated with atypia, as frequently observed in complete mole. The chorionic villi of early diploid complete mole show characteristic features of villous profile, stromal mucin and stromal nuclear debris. The distinction between complete mole and triploid partial mole can be facilitated by ploidy analysis and immunohistochemistry for the product of the paternally imprinted, maternally expressed gene, p57kip2. Persistent trophoblastic disease (PTD) is a clinical not a histopathological diagnosis and the role of the histopathologist once a diagnosis of PTD has been made is limited. Invasive mole and choriocarcinoma are encompassed by PTD. Tumours of the non-villous trophoblast are placental site trophoblastic tumour and the more recently recognised epithelioid trophoblastic tumour. The role of immunohistochemistry in the elucidation of trophoblastic lesions is discussed pragmatically.
Collapse
Affiliation(s)
- Michael Wells
- Academic Unit of Pathology, University of Sheffield Medical School, United Kingdom.
| |
Collapse
|
32
|
Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem 2007; 101:9-33. [PMID: 17340614 DOI: 10.1002/jcb.21255] [Citation(s) in RCA: 286] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is an important growth inhibitor of epithelial cells, and insensitivity to this cytokine results in uncontrolled cell proliferation and can contribute to tumorigenesis. Smad2 and Smad3 are direct mediators of TGF-beta signaling, however little is known about the selective activation of Smad2 versus Smad3. The Smad2 and Smad3 knockout mouse phenotypes and studies comparing Smad2 and Smad3 activation of TGF-beta target genes, suggest that Smad2 and Smad3 have distinct roles in TGF-beta signaling. The observation that TGF-beta inhibits proliferation of Smad3-null mammary gland epithelial cells, whereas Smad3 deficient fibroblasts are only partially growth inhibited, suggests that Smad3 has a different role in epithelial cells and fibroblasts. Herein, the current understanding of Smad2 and Smad3-mediated TGF-beta signaling and their relative roles are discussed, in addition to potential mechanisms for the selective activation of Smad2 versus Smad3. Since alterations in the TGF-beta signaling pathway play an important role in promoting tumorigenesis and cancer progression, methods for therapeutic targeting of the TGF-beta signaling pathway are being pursued. Determining how Smad2 or Smad3 differentially regulate the TGF-beta response may translate into developing more effective strategies for cancer therapy.
Collapse
Affiliation(s)
- Kimberly A Brown
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
33
|
Wang X, Fu S, Freedman RS, Liu J, Kavanagh JJ. Immunobiology of gestational trophoblastic diseases. Int J Gynecol Cancer 2006; 16:1500-15. [PMID: 16884358 DOI: 10.1111/j.1525-1438.2006.00539.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Gestational trophoblastic diseases (GTDs) comprise a group of interrelated diseases characterized by development after gestation, widespread metastases, and high curability with chemotherapy. The good prognosis of GTDs is considered partly a result of the host immune response to paternal antigens expressed on trophoblastic cells. In this study, we review current understanding of the immunobiology of GTDs. First of all, we describe the microenvironment between trophoblastic cells and subpopulation of immune cells. Second, immunogenetics, immune microenvironment around abnormal trophoblast, and mechanism of GTDs escaping from maternal immune system surveillance were also discussed. Third, we propose the possible immunotherapy for persistent GTDs, particularly the vaccine designed on human chorionic gonadotrophin, which is generally accepted as a tumor marker for GTDs diagnosis. Due to the low incidence of GTDs and high response to chemotherapy, there have been few literatures about immunobiologic characteristics of GTDs compared with the other gynecologic malignancies, such as ovarian cancer, but the immunologic behavior of GTDs should be explored for further understanding of the etiology of these diseases and to help designing immunotherapeutic strategies for persistent GTDs.
Collapse
Affiliation(s)
- X Wang
- Department of Gynecologic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | | | | | | | | |
Collapse
|
34
|
Zhao MR, Qiu W, Li YX, Zhang ZB, Li D, Wang YL. Dual effect of transforming growth factor β1 on cell adhesion and invasion in human placenta trophoblast cells. Reproduction 2006; 132:333-41. [PMID: 16885541 DOI: 10.1530/rep.1.01112] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transforming growth factor β (TGFβ) has been shown to be a multifunctional cytokine required for embryonic development and regulation of trophoblast cell behaviors. In the present study, a non-transformed cell-line representative of normal human trophoblast (NPC) was used to examine the effect of TGFβ1 on trophoblast cell adhesion and invasion.In vitroassay showed that TGFβ1 could significantly promote intercellular adhesion, while inhibiting cell invasion across the collagen I-coated filter. Reverse transcription (RT)-PCR and gelatin zymography demonstrated that TGFβ1 evidently repressed the mRNA expression and proenzyme production of matrix metalloproteinase (MMP)-9, but exerted no effect on mRNA expression and secretion of MMP-2. On the other hand, both the mRNA and protein expression of epithelial-cadherin and β-catenin were obviously upregulated by TGFβ1 in dose-dependent fashion, as revealed by RT-PCR and western-blot analysis. What is more, one of the critical TGFβ signaling molecules – Smad2 was notably phosphorylated in TGFβ1-treated NPC cells. The data indicates that cell invasion and adhesion are coordinated processes in human trophoblasts and that there exists paracrine regulation on adhesion molecules and invasion-associated enzymes in human placenta.
Collapse
Affiliation(s)
- Mei-rong Zhao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 25 Bei Si Huan Xi Road, Beijing 100080, People's Republic of China
| | | | | | | | | | | |
Collapse
|
35
|
Okabe T, Matsushima K. Regulation of ALP Activity by TNF-α on Human Dental Pulp. J Endod 2006; 32:516-20. [PMID: 16728240 DOI: 10.1016/j.joen.2005.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Revised: 12/11/2005] [Accepted: 12/13/2005] [Indexed: 10/24/2022]
Abstract
In this study, we examined the effects of TNF-alpha on Bone morphogenetic protein (BMP-2), Smads (which play intracellular signaling of BMPs) expression and alkaline phosphatase (ALP) activity of human dental pulp (HDP) cells to clarify the mechanism of tertiary dentin formation. The quantity of RT-PCR product for BMP-2 from the HDP cells stimulated by TNF-alpha is increased. However, ALP activity was not increased on the cells incubated with TNF-alpha. On the other hand, ALP activity was significantly increased on HDP cells treated with Ammonium Pyrrolidinedithiocarbamate (PDTC, NF- kappaB inhibitor) groups and combined supplementation of TNF-alpha and PDTC groups. Furthermore, we examined the effect of TNF-alpha and PDTC on Smad7 expression using RT-PCR and western blot analysis. Smad7 expression in HDP cells was increased by TNF-alpha, but decreased by PDTC treatment. These results suggest that NF- kappaB and Smad7 play an important role in the down regulation of ALP activity by TNF-alpha on HDP cells.
Collapse
Affiliation(s)
- Tatsu Okabe
- Department of Endodontics, Nihon University School of Dentistry at Matsudo, Matsudo, Chiba, Japan.
| | | |
Collapse
|
36
|
Ishimatsu S, Itakura A, Okada M, Kotani T, Iwase A, Kajiyama H, Ino K, Kikkawa F. Angiotensin II Augmented Migration and Invasion of Choriocarcinoma Cells Involves PI3K Activation Through the AT1 Receptor. Placenta 2006; 27:587-91. [PMID: 16122787 DOI: 10.1016/j.placenta.2005.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Revised: 06/27/2005] [Accepted: 07/05/2005] [Indexed: 11/20/2022]
Abstract
While angiotensin II (Ang II) has been shown to inhibit migration of extravillous trophoblasts via plasminogen activator inhibitor-1 (PAI-1) activation, it has remained unclear whether it stimulates or inhibits malignant behavior of choriocarcinoma cells. Since we previously found an involvement of the renin-angiotensin system (RAS) in the proliferative potential in choriocarcinoma cells (BeWo), mediated via the Ang II type 1 receptor (AT1R), in the present study we investigated the effects of Ang II on choriocarcinoma cell migration/invasion in vitro using Transwell cell culture chambers. Ang II (10(-8)M) promoted migration and invasion by a choriocarcinoma cell line and augmented random cell mobility on checkerboard analysis. Immunoblotting showed Ang II to activate the phosphorylation of FAK and Akt in BeWo cells. Furthermore Ang II effects on cell migration were abolished by a selective AT1R antagonist and a phosphatidylinositol 3-kinase (PI3K) inhibitor. The present results suggest that Ang II-induced migration and invasion of choriocarcinoma cells probably involves PI3K following binding to the AT1R.
Collapse
Affiliation(s)
- S Ishimatsu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Xu G, Zhou H, Wang Q, Auersperg N, Peng C. Activin receptor-like kinase 7 induces apoptosis through up-regulation of Bax and down-regulation of Xiap in normal and malignant ovarian epithelial cell lines. Mol Cancer Res 2006; 4:235-46. [PMID: 16603637 DOI: 10.1158/1541-7786.mcr-05-0174] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transforming growth factor-beta superfamily has been implicated in tumorigenesis. We have recently shown that Nodal, a member of transforming growth factor-beta superfamily, and its receptor, activin receptor-like kinase 7 (ALK7), inhibit proliferation and induce apoptosis in human epithelial ovarian cancer cell lines. In this study, we further investigated the cellular mechanisms underlying the apoptotic action of ALK7 using an immortalized ovarian surface epithelial cell line, IOSE397, and an epithelial ovarian cancer cell line, OV2008. Infection of these cells with an adenoviral construct carrying constitutively active ALK7 (Ad-ALK7-ca) potently induced cell death; all cells died after 3 and 5 days of Ad-ALK7-ca infection in IOSE397 and OV2008 cells, respectively. ALK7-ca induced the expression of proapoptotic factor Bax but suppressed the expression of antiapoptotic factors Bcl-2, Bcl-XL, and Xiap. Silencing of Bax by small interfering RNA in IOSE397 cells significantly reduced ALK7-ca-induced apoptosis as measured by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay but partially blocked ALK7-ca-induced caspase-3 activation and did not affect the down-regulation of Xiap by ALK7-ca. Dominant-negative Smad2, Smad3, and Smad4 blocked ALK7-ca-regulated Xiap and Bax expression and caspase-3 activation. Thus, ALK7-induced apoptosis is at least in part through two Smad-dependent pathways, Bax/Bcl-2 and Xiap.
Collapse
Affiliation(s)
- Guoxiong Xu
- Department of Biology, York University, 4700 Keel Street, Toronto, Ontario, Canada M3J 1P3
| | | | | | | | | |
Collapse
|
38
|
Tower CL, Chappell SL, Morgan K, Kalsheker N, Baker PN, Morgan LJ. Transforming growth factor beta1 regulates angiotensin II type I receptor gene expression in the extravillous trophoblast cell line SGHPL-4. Mol Hum Reprod 2005; 11:847-52. [PMID: 16339777 DOI: 10.1093/molehr/gah242] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The angiotensin II type 1 (AT1) receptor, transforming growth factor beta1 (TGFbeta1) and Oncostatin M (OSM) control key pathways that may be important during placentation. Although interactions between them exist in other tissues, trophoblast cells have not been investigated. Extravillous trophoblast cells, SGHPL-4, were stimulated with 10 ng/ml TGFbeta1 +/- 100 ng/ml OSM for 24 h. Real-time PCR showed that AT1 expression increased 2.76-fold [95% confidence interval (CI) = 1-6.74, P = 0.05] in response to TGFbeta1 and 4.21-fold (95% CI = 1.33-11.76, P = 0.03) with TGFbeta1 + OSM. Luciferase reporter gene constructs containing three haplotypes of the 59 flanking region of the AT1 receptor gene were transfected into SGHPL-4 and HepG2 cells and stimulated with 0.1, 1 and 10 ng/ml TGFbeta1 and 50 ng/ml OSM. Responses were dose and cell dependent. Luciferase activity increased in HepG2 cells in response to TGFbeta1 alone or together with OSM (P < 0.001); transcriptional activation differed between AT1 receptor gene haplotypes. In SGHPL-4 cells, luciferase activity was reduced on exposure to low concentrations of TGFbeta1 or high concentrations of TGFbeta1 combined with OSM (P = 0.003); the response was unaffected by haplotype. Interaction between AT1 and TGFbeta1 is a novel observation in trophoblast and suggests new avenues for the study of placentation.
Collapse
Affiliation(s)
- C L Tower
- Division of Clinical Chemistry, Institute of Genetics, School of Molecular Medical Sciences, University Hospital, Nottingham, UK
| | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Jeffery L Miller
- Molecular Medicine Branch, NIDDK, NIH, Bethesda, Maryland 20892, USA.
| |
Collapse
|
40
|
Pollheimer J, Knöfler M. Signalling pathways regulating the invasive differentiation of human trophoblasts: a review. Placenta 2005; 26 Suppl A:S21-30. [PMID: 15837062 DOI: 10.1016/j.placenta.2004.11.013] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2004] [Indexed: 01/22/2023]
Abstract
The invasive differentiation pathway of trophoblasts is an indispensable physiological process of early human placental development. Formation of anchoring villi, proliferation of cell columns and invasion of extravillous cytotrophoblasts into maternal decidual stroma and vessels induce vascular changes ensuring an adequate blood supply to the growing fetus. Extravillous trophoblast differentiation is regulated by numerous growth factors as well as by extracellular matrix proteins and adhesion molecules expressed at the fetal-maternal interface. These regulatory molecules control cell invasion by modulating activities of matrix-degrading protease systems and ECM adhesion. The differentiation process involves numerous signalling cascades/proteins such as the GTPases RhoA, the protein kinases ROCK, ERK1, ERK2, FAK, PI3K, Akt/protein kinase B and mTOR as well as TGF-beta-dependent SMAD factors. While an increasing number of signalling pathways regulating trophoblast differentiation are being unravelled, downstream effectors such as executing transcription factors remain largely elusive. Here, we summarise our current knowledge on signal transduction cascades regulating invasive trophoblast differentiation. We will focus on cell model systems which are used to study the particular differentiation process and discuss signalling pathways which regulate trophoblast proliferation and motility.
Collapse
Affiliation(s)
- J Pollheimer
- Department of Obstetrics and Gynecology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | |
Collapse
|
41
|
Báez A, Cantor A, Fonseca S, Marcos-Martinez M, Mathews LA, Muro-Cacho CA, Muñoz-Antonia T. Differences in Smad4 expression in human papillomavirus type 16-positive and human papillomavirus type 16-negative head and neck squamous cell carcinoma. Clin Cancer Res 2005; 11:3191-7. [PMID: 15867212 DOI: 10.1158/1078-0432.ccr-04-1299] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The SMADs are a group of interrelated proteins that mediate transforming growth factor beta (TGF-beta) signaling. Upon TGF-beta binding the TGF-beta type I receptor phosphorylates Smad2 and Smad3, which then complex with Smad4 and translocate to the nucleus, with subsequent activation of target genes. Disruption of TGF-beta signaling is thought to contribute to the development of head and neck squamous cell carcinomas (HNSCC). Alterations in the function of the DPC4/Smad4 tumor suppressor gene have been found to inactivate TGF-beta signaling in several tumor types. For example, DPC4/Smad4 is lost or mutated in colorectal, pancreatic, and esophageal cancers. In addition, DPC4/Smad4 transcriptional activity and TGF-beta ability to inhibit DNA synthesis is blocked by the E7 protein of the human papillomavirus type 16 (HPV16) in cervical carcinoma cell lines. HPV16 infection is a risk factor for the development of a subset of HNSCC. This study was undertaken to investigate a potential correlation between expression of components of the TGF-beta signaling pathway and HPV16 status in HNSCC tumors. We examined the expression of TGF-beta signaling proteins Smad2, Smad2-P, and Smad4 by immunohistochemistry in 27 HPV16-negative and 16 HPV16-positive HNSCCs. We compared the expression patterns and assessed their relationship to HPV16 status. No significant differences were detected between HPV16-positive and HPV16-negative tumors in the expression of Smad2 and Smad2-P. Smad4 expression, however, was decreased in 56% of the HPV16-positive tumors and in 39% of HPV16-negative tumors. This difference was statistically significant (P = 0.01) suggesting that loss of Smad4 expression may be involved in HPV16-induced carcinogenesis of HNSCC.
Collapse
Affiliation(s)
- Adriana Báez
- Department of Otolaryngology-Head and Neck Surgery, University of Puerto Rico School of Medicine, San Juan, Puerto Rico.
| | | | | | | | | | | | | |
Collapse
|
42
|
Nicola C, Timoshenko AV, Dixon SJ, Lala PK, Chakraborty C. EP1 receptor-mediated migration of the first trimester human extravillous trophoblast: the role of intracellular calcium and calpain. J Clin Endocrinol Metab 2005; 90:4736-46. [PMID: 15886234 DOI: 10.1210/jc.2005-0413] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
CONTEXT The root cause of preeclampsia in the human lies in the placenta, where a subpopulation of cytotrophoblast cells called extravillous trophoblasts (EVT), known to be involved in the invasion of the uterine endometrium and utero-placental arteries, become less invasive, resulting in poor perfusion of maternal blood into placenta. OBJECTIVES Because EVT migrate into the prostaglandin (PG) E2-rich decidua, we tested the roles of PGE2 and PGE2-mediated signaling in EVT migration, using our well-characterized EVT line HTR-8/Svneo as well as first trimester villus explants in culture. DESIGN mRNA expression of different PGE2 receptors (EPs) in HTR-8/Svneo cells was studied using RT-PCR. To characterize the functional significance of EP receptors in EVT, different EP receptor agonists and antagonists were used in our migration assay systems and in the measurements of intracellular concentration of Ca2+ ([Ca2+]i) and calpain activity. RESULTS Exogenous PGE2 stimulated EVT migration both in vitro and in the villus explant cultures. Although EVT expressed mRNA for all EP receptors (EP 1-4), a functional predominance of EP1 and EP4 was demonstrated in migration assays using specific EP agonists and antagonists. EP1-receptor-mediated signaling events such as activation of phospholipase C and elevation of cytosolic free [Ca2+]i were confirmed by the following findings: 1) exogenous PGE2 or an EP1 agonist, but not an EP4 agonist, increased [Ca2+]i, which could be blocked with an EP1 antagonist as well as BAPTA and thapsigargin; 2) phospholipase C inhibitor U73122, BAPTA, and thapsigargin inhibited PGE2-mediated migratory response of EVT; and 3) PGE2-mediated EVT migration was shown to be dependent on a class of Ca2+-dependent proteases called calpains, known to be involved in cell detachment from substratum during migratory responses. The presence of PGE2 stimulated calpain activity, whereas two calpain inhibitors, calpastatin and N-Ac-Leu-Leu-methioninal (ALLM), blocked EVT migration. CONCLUSION PGE2 stimulates EVT migration by signaling through EP1 receptors, increasing [Ca2+]i, and activating calpain.
Collapse
Affiliation(s)
- Catalin Nicola
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Canada N6A 5C1
| | | | | | | | | |
Collapse
|
43
|
Prokova V, Mavridou S, Papakosta P, Kardassis D. Characterization of a novel transcriptionally active domain in the transforming growth factor beta-regulated Smad3 protein. Nucleic Acids Res 2005; 33:3708-21. [PMID: 15994459 PMCID: PMC1169234 DOI: 10.1093/nar/gki679] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/20/2005] [Accepted: 06/12/2005] [Indexed: 01/09/2023] Open
Abstract
Transforming growth factor beta (TGFbeta) regulates transcriptional responses via activation of cytoplasmic effector proteins termed Smads. Following their phosphorylation by the type I TGFbeta receptor, Smads form oligomers and translocate to the nucleus where they activate the transcription of TGFbeta target genes in cooperation with nuclear cofactors and coactivators. In the present study, we have undertaken a deletion analysis of human Smad3 protein in order to characterize domains that are essential for transcriptional activation in mammalian cells. With this analysis, we showed that Smad3 contains two domains with transcriptional activation function: the MH2 domain and a second middle domain that includes the linker region and the first two beta strands of the MH2 domain. Using a protein-protein interaction assay based on biotinylation in vivo, we were able to show that a Smad3 protein bearing an internal deletion in the middle transactivation domain is characterized by normal oligomerization and receptor activation properties. However, this mutant has reduced transactivation capacity on synthetic or natural promoters and is unable to interact physically and functionally with the histone acetyltransferase p/CAF. The loss of interaction with p/CAF or other coactivators could account, at least in part, for the reduced transactivation capacity of this Smad3 mutant. Our data support an essential role of the previously uncharacterized middle region of Smad3 for nuclear functions, such as transcriptional activation and interaction with coactivators.
Collapse
Affiliation(s)
- Vassiliki Prokova
- Department of Basic Sciences, University of Crete Medical SchoolHeraklion 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-HellasHeraklion 71003, Greece
| | - Sofia Mavridou
- Department of Basic Sciences, University of Crete Medical SchoolHeraklion 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-HellasHeraklion 71003, Greece
| | - Paraskevi Papakosta
- Department of Basic Sciences, University of Crete Medical SchoolHeraklion 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-HellasHeraklion 71003, Greece
| | - Dimitris Kardassis
- Department of Basic Sciences, University of Crete Medical SchoolHeraklion 71003, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-HellasHeraklion 71003, Greece
| |
Collapse
|
44
|
Munir S, Xu G, Wu Y, Yang B, Lala PK, Peng C. Nodal and ALK7 inhibit proliferation and induce apoptosis in human trophoblast cells. J Biol Chem 2004; 279:31277-86. [PMID: 15150278 DOI: 10.1074/jbc.m400641200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nodal, a member of the transforming growth factor-beta superfamily, is known to play critical roles in early vertebrate development, but its functions in extraembryonic tissues are unclear. ALK7 is a type I receptor for Nodal. Recently, we demonstrated that Nodal mRNA and several ALK7 transcripts are expressed in human placenta throughout pregnancy (Roberts, H. J., Hu, S., Qiu, Q., Leung, P. C. K., Cannigia, I., Gruslin, A., Tsang, B., and Peng, C. (2003) Biol. Reprod. 68, 1719-1726). In this study, we determined the role of Nodal and ALK7 in trophoblast cell proliferation and apoptosis. Overexpression of Nodal in normal trophoblast cells (HTR8/SVneo) and several choriocarcinoma cell lines resulted in a significant decrease in the number of metabolically active cells. The effect of Nodal could be mimicked by constitutively active ALK7 (ALK7-ca), but was blocked by kinase-deficient ALK7. The growth inhibitory effect of Nodal was also blocked by dominant-negative Smad2/3. Overexpression of Nodal and ALK7-ca induced apoptosis in trophoblast cells as determined by Hoechst staining, flow cytometry, and caspase-3 Western blotting. In addition, Nodal and ALK7-ca decreased the number of proliferating cells as measured by bromodeoxyuridine assays. Furthermore, overexpression of Nodal or ALK7-ca increased p27 expression, but reduced the levels of Cdk2 and cyclin D(1). Taken together, this study demonstrates for the first time that Nodal, acting through ALK7 and Smad2/3, inhibits proliferation and induces apoptosis in human trophoblast cells. Our findings also suggest that the Nodal-ALK7 pathway inhibits cell proliferation by inducing G(1) cell cycle arrest and that this effect is mediated in part by the p27-cyclin E/Cdk2 pathway.
Collapse
Affiliation(s)
- Sadia Munir
- Department of Biology, York University, Toronto, Ontario M3J 1P3, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Brown KA, Aakre ME, Gorska AE, Price JO, Eltom SE, Pietenpol JA, Moses HL. Induction by transforming growth factor-beta1 of epithelial to mesenchymal transition is a rare event in vitro. Breast Cancer Res 2004; 6:R215-31. [PMID: 15084245 PMCID: PMC400675 DOI: 10.1186/bcr778] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2003] [Revised: 02/10/2004] [Accepted: 02/20/2004] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Transforming growth factor (TGF)-beta1 is proposed to inhibit the growth of epithelial cells in early tumorigenesis, and to promote tumor cell motility and invasion in the later stages of carcinogenesis through the induction of an epithelial to mesenchymal transition (EMT). EMT is a multistep process that is characterized by changes in cell morphology and dissociation of cell-cell contacts. Although there is growing interest in TGF-beta1-mediated EMT, the phenotype is limited to only a few murine cell lines and mouse models. METHODS To identify alternative cell systems in which to study TGF-beta1-induced EMT, 18 human and mouse established cell lines and cultures of two human primary epithelial cell types were screened for TGF-beta1-induced EMT by analysis of cell morphology, and localization of zonula occludens-1, E-cadherin, and F-actin. Sensitivity to TGF-beta1 was also determined by [3H]thymidine incorporation, flow cytometry, phosphorylation of Smad2, and total levels of Smad2 and Smad3 in these cell lines and in six additional cancer cell lines. RESULTS TGF-beta1 inhibited the growth of most nontransformed cells screened, but many of the cancer cell lines were insensitive to the growth inhibitory effects of TGF-beta1. In contrast, TGF-beta1 induced Smad2 phosphorylation in the majority of cell lines, including cell lines resistant to TGF-beta1-mediated cell cycle arrest. Of the cell lines screened only two underwent TGF-beta1-induced EMT. CONCLUSION The results presented herein show that, although many cancer cell lines have lost sensitivity to the growth inhibitory effect of TGF-beta1, most show evidence of TGF-beta1 signal transduction, but only a few cell lines undergo TGF-beta1-mediated EMT.
Collapse
Affiliation(s)
- Kimberly A Brown
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mary E Aakre
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Agnieska E Gorska
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - James O Price
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Veterans Affairs Medical Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sakina E Eltom
- Department of Pharmacology, Meharry Medical College, Nashville, Tennessee, USA
| | - Jennifer A Pietenpol
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Harold L Moses
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
46
|
Henrich D, Hahn P, Wahl M, Wilhelm K, Dernbach E, Dimmeler S, Marzi I. Serum Derived from Multiple Trauma Patients Promotes the Differentiation of Endothelial Progenitor Cells In Vitro: Possible Role of Transforming Growth Factor-??1 and Vascular Endothelial Growth Factor165. Shock 2004; 21:13-6. [PMID: 14676678 DOI: 10.1097/01.shk.0000101669.49265.50] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ischemia in various organs and tissues takes place during and as a direct result of multiple trauma (MT). Bone marrow-derived endothelial progenitor cells (EPCs) are involved in neovascularization after ischemic incidences. Here, we report that serum derived from patients with MT stimulates differentiation of EPCs in vitro from peripheral blood mononuclear cells (PBMCs). EPCs were identified by DiL-Acetyl-LDL-uptake with concomitant UEA-I-lectin binding. A significant increase in EPC numbers was noted when PBMCs were cultivated for 72 h with the serum of MT patients (n = 25) obtained at 5 days. Furthermore, serum from MT patients enhanced the functional acting of EPCs to form prevascular structures in matrigel. Reverse transcription polymerase chain reaction analysis revealed gene expression of transforming growth factor (TGF)-beta1- and vascular endothelial growth factor (VEGF) receptors 1 and 2. Reverse transcription polymerase chain reaction analysis was based on further cultivated cell preparations, which contained at least 80% EPCs. Moreover, the addition of recombinant VEGF or low concentrations of TGF-beta increased EPC differentiation. In addition, neutralization of TGF-beta1 and of VEGF165 in MT serum using specific antibodies resulted in a significant decrease in EPC differentiation. Our data indicate that TGF-beta1 and VEGF165 play a pivotal role for EPC differentiation induced by serum of polytrauma patients.
Collapse
Affiliation(s)
- Dirk Henrich
- Department of Trauma Surgery, Johann-Wolfgang-Goethe University, Frankfurt/Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
47
|
Kretschmer A, Moepert K, Dames S, Sternberger M, Kaufmann J, Klippel A. Differential regulation of TGF-beta signaling through Smad2, Smad3 and Smad4. Oncogene 2003; 22:6748-63. [PMID: 14555988 DOI: 10.1038/sj.onc.1206791] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Smad transcription factors mediate the growth inhibitory effect of transforming growth factor-beta (TGF-beta) in many cell types. Mutational inactivation of Smads has been correlated with loss of responsiveness to TGF-beta-mediated signal transduction. In this study, we compare the contribution of individual Smads to TGF-beta-induced growth inhibition and endogenous gene expression in isogenic cellular backgrounds. Smad2, Smad3 and Smad4 expression were selectively inhibited in differentiation-competent cells by using improved antisense molecules. We found that TGF-beta mediates its inhibitory effect on HaCaT keratinocyte cell growth predominantly through Smad3. Inhibition of Smad3 expression was sufficient to interfere with TGF-beta-induced cell cycle arrest and to induce or suppress endogenous cell cycle regulators. Inhibition of Smad4 expression exhibited a partial effect, whereas inhibition of Smad2 expression had no effect. By gene expression profiling, we identified TGF-beta-dependent genes that are differentially regulated by Smad2 and Smad3 under regular growth conditions on a genome-wide scale. We show that Smad2, Smad3 and Smad4 contribute to the regulation of TGF-beta responses to varying extents, and demonstrate, in addition, that these Smads exhibit distinct roles in different cell types.
Collapse
Affiliation(s)
- Anny Kretschmer
- atugen AG, Robert-Roessle-Strasse 10, Otto Warburg Hause (No 80), Berlin 13125, Germany
| | | | | | | | | | | |
Collapse
|
48
|
Lala PK, Chakraborty C. Factors regulating trophoblast migration and invasiveness: possible derangements contributing to pre-eclampsia and fetal injury. Placenta 2003; 24:575-87. [PMID: 12828917 DOI: 10.1016/s0143-4004(03)00063-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Impaired trophoblast invasiveness and spiral arterial remodelling, which results in poor placental perfusion during early pregnancy, is believed to cause fetal injury and growth retardation, and also endothelial cell activation/dysfunction in a susceptible mother, leading to clinical manifestations of pre-eclampsia. This article briefly reviews the regulatory roles of certain locally active factors in trophoblast migration and invasiveness. This background is then used to discuss and debate whether derangements or dysfunction of some of these factors can manifest as early serum markers predictive of the disease, as opposed to the intermediate and late stage markers which may reflect manifestations and consequences of the disease. Of particular significance are the observed derangements in uPA/uPAR/PAI system, IGFBP-1, HGF, HB-EGF and TGFbeta, factors which are known to regulate trophoblast migration and invasiveness in situ. An emphasis is placed on the need for longitudinal studies in order to identify predictive serum markers which may help strategies for prevention or amelioration of fetal injury and pre-eclampsia.
Collapse
Affiliation(s)
- P K Lala
- Department of Anatomy and Cell Biology, The University of Western Ontario, Ontario, N6A 5C1, London, Canada.
| | | |
Collapse
|
49
|
Govinden R, Bhoola KD. Genealogy, expression, and cellular function of transforming growth factor-beta. Pharmacol Ther 2003; 98:257-65. [PMID: 12725873 DOI: 10.1016/s0163-7258(03)00035-4] [Citation(s) in RCA: 172] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transforming growth factor-beta (TGF-beta) gene superfamily expresses a large set of structurally and functionally related polypeptides. Three TGF-beta isoforms are regulated by specific genes and have been identified in mammals (TGF-beta1, -beta2, and -beta3). All three-protein isoforms are observed abundantly during development and display overlapping and distinct spatial and temporal patterns of expressions. Each isoform plays a distinct role, the nature of which depends on the cell type, its state of differentiation, and growth conditions, and on the other growth factors present. TGF-beta regulates many of the processes common to both tissue repair and disease, including angiogenesis, chemotoxins, fibroblast proliferation and the controlled synthesis, and degradation of matrix proteins, such as collagen and fibronectin. This review will examine the genealogy and mode of actions of TGF-beta on the cell types involved in inflammation and repair, as well as in carcinoma.
Collapse
Affiliation(s)
- R Govinden
- HIV Prevention Research Unit, Medical Research Council, Durban, South Africa
| | | |
Collapse
|
50
|
Chen VTS, Peng C, Leung PCK. Activin-A up-regulates type I activin receptor mRNA levels in human immortalized extravillous trophoblast cells. Reprod Biol Endocrinol 2003; 1:29. [PMID: 12702211 PMCID: PMC153493 DOI: 10.1186/1477-7827-1-29] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2003] [Accepted: 03/24/2003] [Indexed: 11/10/2022] Open
Abstract
Activin is known to play an important regulatory role in reproduction, including pregnancy. To further examine the role and signaling mechanism of activin in regulating placental function, the steady-state level of activin type I receptor (ActRI) mRNA in immortalized extravillous trophoblasts (IEVT) cells was measured using competitive PCR (cPCR). An internal standard of ActRI cDNA for cPCR was constructed for the quantification of ActRI mRNA levels in IEVT cells. ActRI mRNA levels were increased in a dose-dependent manner by activin-A with the maximal effect observed at the dose of 10 ng/ml. Time course studies revealed that activin-A had maximal effects on ActRI mRNA levels at 6 hours after treatment. The effects of activin-A on ActRI mRNA levels was blocked by follistatin, an activin binding protein, in a dose-dependent manner. In addition, inhibin-A inhibited basal, as well as activin-A-induced ActRI mRNA levels. These findings provide evidence, for the first time, that activin-A modulates ActRI mRNA levels in human trophoblast cells.
Collapse
Affiliation(s)
- Victor T S Chen
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada.
| | | | | |
Collapse
|