1
|
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H, Imanaka S. An integral role of mitochondrial function in the pathophysiology of preeclampsia. Mol Biol Rep 2024; 51:330. [PMID: 38393449 DOI: 10.1007/s11033-024-09285-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/24/2024] [Indexed: 02/25/2024]
Abstract
Preeclampsia (PE) is associated with high maternal and perinatal morbidity and mortality. The development of effective treatment strategies remains a major challenge due to the limited understanding of the pathogenesis. In this review, we summarize the current understanding of PE research, focusing on the molecular basis of mitochondrial function in normal and PE placentas, and discuss perspectives on future research directions. Mitochondria integrate numerous physiological processes such as energy production, cellular redox homeostasis, mitochondrial dynamics, and mitophagy, a selective autophagic clearance of damaged or dysfunctional mitochondria. Normal placental mitochondria have evolved innovative survival strategies to cope with uncertain environments (e.g., hypoxia and nutrient starvation). Cytotrophoblasts, extravillous trophoblast cells, and syncytiotrophoblasts all have distinct mitochondrial morphology and function. Recent advances in molecular studies on the spatial and temporal changes in normal mitochondrial function are providing valuable insight into PE pathogenesis. In PE placentas, hypoxia-mediated mitochondrial fission may induce activation of mitophagy machinery, leading to increased mitochondrial fragmentation and placental tissue damage over time. Repair mechanisms in mitochondrial function restore placental function, but disruption of compensatory mechanisms can induce apoptotic death of trophoblast cells. Additionally, molecular markers associated with repair or compensatory mechanisms that may influence the development and progression of PE are beginning to be identified. However, contradictory results have been obtained regarding some of the molecules that control mitochondrial biogenesis, dynamics, and mitophagy in PE placentas. In conclusion, understanding how the mitochondrial morphology and function influence cell fate decisions of trophoblast cells is an important issue in normal as well as pathological placentation biology. Research focusing on mitochondrial function will become increasingly important for elucidating the pathogenesis and effective treatment strategies of PE.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara, 634-0813, Japan.
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan.
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, 2-897-5 Shichijyonishi-machi, Nara, 630-8581, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Medicine, Kei Oushin Clinic, 5-2-6, Naruo-cho, Nishinomiya, 663-8184, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, 3-3-17 Kitatomigaoka-cho, Nara, 634- 0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, 871-1 Shijo-cho, Kashihara, 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-cho, Kashihara, 634-8522, Japan
| |
Collapse
|
2
|
Jaremek A, Shaha S, Jeyarajah MJ, Jaju Bhattad G, Chowdhury D, Riddell M, Renaud SJ. Genome-Wide Analysis of Hypoxia-Inducible Factor Binding Reveals Targets Implicated in Impaired Human Placental Syncytiotrophoblast Formation under Low Oxygen. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:846-865. [PMID: 37028593 DOI: 10.1016/j.ajpath.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/08/2023]
Abstract
Preeclampsia (PE) is a common and serious complication of pregnancy with no cure except premature delivery. The root cause of PE is improper development of the placenta-the temporary organ supporting fetal growth and development. Continuous formation of the multinucleated syncytiotrophoblast (STB) layer via differentiation and fusion of cytotrophoblasts (CTBs) is vital for healthy placentation and is impaired in preeclamptic pregnancies. In PE, there is reduced/intermittent placental perfusion, likely resulting in a persistently low O2 environment. Low O2 inhibits differentiation and fusion of CTBs into STB and may thus contribute to PE pathogenesis; however, the underlying mechanisms are unknown. Because low O2 activates a transcription factor complex in cells known as the hypoxia-inducible factor (HIF), the objective of this study was to investigate whether HIF signaling inhibits STB formation by regulating genes required for this process. Culture of primary CTBs, the CTB-like cell line BeWo, and human trophoblast stem cells under low O2 reduced cell fusion and differentiation into STB. Knockdown of aryl hydrocarbon receptor nuclear translocator (a key component of the HIF complex) in BeWo cells restored syncytialization and expression of STB-associated genes under different O2 levels. Chromatin immunoprecipitation sequencing facilitated the identification of global aryl hydrocarbon receptor nuclear translocator/HIF binding sites, including several near genes implicated in STB development, such as ERVH48-1 and BHLHE40, providing new insights into mechanisms underlying pregnancy diseases linked to poor placental O2 supply.
Collapse
Affiliation(s)
- Adam Jaremek
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sumaiyah Shaha
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Diba Chowdhury
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Meghan Riddell
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
3
|
Ezoe K, Fujiwara N, Miki T, Kato K. Post-warming culture of human vitrified blastocysts with prolactin improves trophoblast outgrowth. Reprod Biol Endocrinol 2023; 21:6. [PMID: 36653830 PMCID: PMC9847091 DOI: 10.1186/s12958-023-01062-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human embryos express the prolactin (PRL) receptor at the morula and blastocyst stages. Treatment with PRL from cleavage to the blastocyst stage improves blastocyst outgrowth on fibronectin-coated dishes. However, whether post-warming PRL treatment of blastocysts cultured without PRL could improve outgrowth competence remains unknown. Furthermore, the optimal time for post-warming PRL treatment remains to be ascertained. This study investigated the effects of PRL treatment during recovery culture on human blastocyst outgrowth and its related genes. METHODS In total, 374 discarded vitrified blastocysts were randomly allocated to two groups, to be cultured with (n = 208) or without PRL (control; n = 166) for 120 min for recovery, and then plated on fibronectin-coated dishes. The expression level of PRL-interacting genes, blastocyst adhesion rate, outgrowth area, distance of trophoblast migration, and outgrowth degeneration were examined. RESULTS The mRNA expression of ezrin, radixin, and moesin, which regulate cell adhesion and invasion by controlling actin reorganization during epithelial-to-mesenchymal transition (EMT), was stimulated by PRL treatment for 120 min. The expression of EMT-related genes, transforming growth factor β1, snail1, and twist1 was also promoted following treatment with PRL for 120 min. PRL-treated blastocysts also exhibited augmented expression of cadherin 2 and transcriptional repression of cadherin 1. Higher mRNA expression of integrin-based focal adhesion-related genes, ITGA5 and ITGB1, was observed after treatment with PRL for 120 min than in the non- and shorter-treatment groups. PRL treatment for 120 min did not alter the rate of blastocyst adhesion to fibronectin-coated dishes 96 h after the outgrowth culture assay. However, multiple linear regression analysis revealed that the outgrowth area was significantly increased in PRL-treated blastocysts. The migration distance of trophoblast cells was significantly increased and degeneration rate was significantly decreased after PRL treatment. Furthermore, a more beneficial effect of PRL treatment on blastocyst outgrowth was observed when the blastocysts were vitrified on day 5 than when they were vitrified on day 6. CONCLUSIONS Post-warming culture of human vitrified blastocysts with PRL for 120 min promoted trophoblast outgrowth in vitrified human blastocysts. Furthermore, PRL treatment may reduce outgrowth degeneration by increasing resistance to apoptosis during trophoblast migration.
Collapse
Affiliation(s)
- Kenji Ezoe
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Nanoha Fujiwara
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Tetsuya Miki
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Keiichi Kato
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
4
|
Zhu Y, Liu X, Xu Y, Lin Y. Hyperglycemia disturbs trophoblast functions and subsequently leads to failure of uterine spiral artery remodeling. Front Endocrinol (Lausanne) 2023; 14:1060253. [PMID: 37091848 PMCID: PMC10113679 DOI: 10.3389/fendo.2023.1060253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Uterine spiral artery remodeling is necessary for fetal growth and development as well as pregnancy outcomes. During remodeling, trophoblasts invade the arteries, replace the endothelium and disrupt the vascular smooth muscle, and are strictly regulated by the local microenvironment. Elevated glucose levels at the fetal-maternal interface are associated with disorganized placental villi and poor placental blood flow. Hyperglycemia disturbs trophoblast proliferation and invasion via inhibiting the epithelial-mesenchymal transition, altering the protein expression of related proteases (MMP9, MMP2, and uPA) and angiogenic factors (VEGF, PIGF). Besides, hyperglycemia influences the cellular crosstalk between immune cells, trophoblast, and vascular cells, leading to the failure of spiral artery remodeling. This review provides insight into molecular mechanisms and signaling pathways of hyperglycemia that influence trophoblast functions and uterine spiral artery remodeling.
Collapse
Affiliation(s)
- Yueyue Zhu
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yi Lin,
| |
Collapse
|
5
|
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
|
6
|
Amruta N, Bix G. ATN-161 Ameliorates Ischemia/Reperfusion-induced Oxidative Stress, Fibro-inflammation, Mitochondrial damage, and Apoptosis-mediated Tight Junction Disruption in bEnd.3 Cells. Inflammation 2021; 44:2377-2394. [PMID: 34420157 PMCID: PMC8380192 DOI: 10.1007/s10753-021-01509-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/25/2021] [Accepted: 06/27/2021] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated the significance of endothelial cell-expressed α5β1 integrin in ischemic stroke, having shown that α5β1 integrin endothelial cell-selective knockout mice are significantly resistance to ischemic stroke injury via preservation of the tight junction protein claudin-5 and subsequent stabilization of the blood–brain barrier (BBB). In addition, inhibition of α5β1 by the small peptide noncompetitive integrin α5 inhibitor, ATN-161, is beneficial in a mouse model of ischemic stroke through reduction of infarct volume, edema, stabilization of the BBB, and reduced inflammation and immune cell infiltration into the brain. In continuation with our previous findings, we have further evaluated the mechanistic role of ATN-161 in vitro and found that oxygen and glucose deprivation and reperfusion (OGD/R)-induced inflammation, oxidative stress, apoptosis, mitochondrial depolarization, and fibrosis attenuate tight junction integrity via induction of α5, NLRP3, p-FAK, and p-AKT signaling in mouse brain endothelial cells. ATN-161 treatment (10 µM) effectively inhibited OGD/R-induced extracellular matrix (ECM) deposition by reducing integrin α5, MMP-9, and fibronectin expression, as well as reducing oxidative stress by reducing mitochondrial superoxide radicals, intracellular ROS, inflammation by reducing NLRP3 inflammasome, tight junction loss by reducing claudin-5 and ZO-1 expression levels, mitochondrial damage by inhibiting mitochondrial depolarization, and apoptosis via regulation of p-FAK and p-AKT levels. Taken together, our results further support therapeutically targeting α5 integrin with ATN-161, a safe, well-tolerated, and clinically validated peptide, in ischemic stroke.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA. .,Department of Neurology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA. .,Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
| |
Collapse
|
7
|
Ezoe K, Miki T, Ohata K, Fujiwara N, Yabuuchi A, Kobayashi T, Kato K. Prolactin receptor expression and its role in trophoblast outgrowth in human embryos. Reprod Biomed Online 2021; 42:699-707. [PMID: 33608185 DOI: 10.1016/j.rbmo.2021.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/27/2020] [Accepted: 01/10/2021] [Indexed: 11/26/2022]
Abstract
RESEARCH QUESTION What is the gene expression pattern of prolactin receptor (PRLR) in human pre-implantation embryos and what are its functions during the embryonic development and adhesion process? DESIGN A total of 405 discarded human vitrified oocytes and embryos donated for research by consenting couples were used in this study. The oocytes and embryos were used to analyse PRLR expression and to evaluate the influence of prolactin (PRL) supplementation in the embryo culture medium on embryo developmental competence and viability. The rates of blastocyst development and adhesion, outgrowth area, cytoskeletal reorganization and nascent adhesion formation were compared between groups. RESULTS PRLR expression increased significantly after embryo compaction (P < 0.0001) and blastulation (P < 0.0001). Supplementation of the embryo culture medium with PRL did not improve the developmental rate and morphological grade. In contrast, blastocyst outgrowth was significantly increased in embryos cultured with PRL (P = 0.0004). Phosphorylation of JAK2, downstream of the prolactin receptor family, was markedly higher in the PRL-treated embryos than in embryos cultured without PRL. Furthermore, the expression of mRNAs encoding ezrin-radixin-moesin proteins and epithelial-mesenchymal transition-related genes was stimulated by the activation of PRL-JAK2 signalling. The PRL-treated embryos had higher mRNA expression of integrins than non-treated embryos, and transcriptional repression of cadherin 1 was observed after PRL treatment. More nascent adherent cells expressed focal adhesion kinase and paxillin in PRL-treated embryos than in non-treated embryos. CONCLUSIONS Human embryos express PRLR at the morula and blastocyst stages, and PRLR signalling stimulates blastocyst adhesion by promoting integrin-based focal adhesions and cytoskeletal organization during trophoblast outgrowth.
Collapse
Affiliation(s)
- Kenji Ezoe
- Kato Ladies Clinic, Shinjuku-ku Tokyo 160-0023, Japan
| | - Tetsuya Miki
- Kato Ladies Clinic, Shinjuku-ku Tokyo 160-0023, Japan
| | - Kazuki Ohata
- Kato Ladies Clinic, Shinjuku-ku Tokyo 160-0023, Japan
| | | | | | | | - Keiichi Kato
- Kato Ladies Clinic, Shinjuku-ku Tokyo 160-0023, Japan.
| |
Collapse
|
8
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Marinello WP, Mohseni ZS, Cunningham SJ, Crute C, Huang R, Zhang JJ, Feng L. Perfluorobutane sulfonate exposure disrupted human placental cytotrophoblast cell proliferation and invasion involving in dysregulating preeclampsia related genes. FASEB J 2020; 34:14182-14199. [PMID: 32901980 DOI: 10.1096/fj.202000716rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
We reported that maternal PFBS, an emerging pollutant, exposure is positively associated with preeclampsia which can result from aberrant trophoblasts invasion and subsequent placental ischemia. In this study, we investigated the effects of PFBS on trophoblasts proliferation/invasion and signaling pathways. We exposed a human trophoblast line, HTR8/SVneo, to PFBS. Cell viability, proliferation, and cell cycle were evaluated by the MTS assay, Ki-67 staining, and flow cytometry, respectively. We assessed cell migration and invasion with live-cell imaging-based migration assay and matrigel invasion assay, respectively. Signaling pathways were examined by Western blot, RNA-seq, and qPCR. PFBS exposure interrupted cell proliferation and invasion in a dose-dependent manner. PFBS (100 μM) did not cause cell death but instead significant cell proliferation without cell cycle disruption. PFBS (10 and 100 μM) decreased cell migration and invasion, while PFBS (0.1 μM) significantly increased cell invasion but not migration. Further, RNA-seq analysis identified dysregulated HIF-1α target genes that are relevant to cell proliferation/invasion and preeclampsia, while Western Blot data showed the activation of HIF-1α, but not Notch, ERK1/2, (PI3K)AKT, and P38 pathways. PBFS exposure altered trophoblast cell proliferation/invasion which might be mediated by preeclampsia-related genes, suggesting a possible association between prenatal PFBS exposure and adverse placentation.
Collapse
Affiliation(s)
- William P Marinello
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Zahra S Mohseni
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Sarah J Cunningham
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Christine Crute
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Jun J Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Interaction of Pregnancy-Specific Glycoprotein 1 With Integrin Α5β1 Is a Modulator of Extravillous Trophoblast Functions. Cells 2019; 8:cells8111369. [PMID: 31683744 PMCID: PMC6912793 DOI: 10.3390/cells8111369] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 01/23/2023] Open
Abstract
Human pregnancy-specific glycoproteins (PSGs) serve immunomodulatory and pro-angiogenic functions during pregnancy and are mainly expressed by syncytiotrophoblast cells. While PSG mRNA expression in extravillous trophoblasts (EVTs) was reported, the proteins were not previously detected. By immunohistochemistry and immunoblotting, we show that PSGs are expressed by invasive EVTs and co-localize with integrin 5. In addition, we determined that native and recombinant PSG1, the most highly expressed member of the family, binds to 51 and induces the formation of focal adhesion structures resulting in adhesion of primary EVTs and EVT-like cell lines under 21% oxygen and 1% oxygen conditions. Furthermore, we found that PSG1 can simultaneously bind to heparan sulfate in the extracellular matrix and to 51 on the cell membrane. Wound healing assays and single-cell movement tracking showed that immobilized PSG1 enhances EVT migration. Although PSG1 did not affect EVT invasion in the in vitro assays employed, we found that the serum PSG1 concentration is lower in African-American women diagnosed with early-onset and late-onset preeclampsia, a pregnancy pathology characterized by shallow trophoblast invasion, than in their respective healthy controls only when the fetus was a male; therefore, the reduced expression of this molecule should be considered in the context of preeclampsia as a potential therapy.
Collapse
|
11
|
Tseng AM, Mahnke AH, Wells AB, Salem NA, Allan AM, Roberts VH, Newman N, Walter NA, Kroenke CD, Grant KA, Akison LK, Moritz KM, Chambers CD, Miranda RC. Maternal circulating miRNAs that predict infant FASD outcomes influence placental maturation. Life Sci Alliance 2019; 2:2/2/e201800252. [PMID: 30833415 PMCID: PMC6399548 DOI: 10.26508/lsa.201800252] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023] Open
Abstract
Maternal gestational circulating microRNAs, predictive of adverse infant outcomes, including growth deficits, following prenatal alcohol exposure, contribute to placental pathology by impairing the EMT pathway in trophoblasts. Prenatal alcohol exposure (PAE), like other pregnancy complications, can result in placental insufficiency and fetal growth restriction, although the linking causal mechanisms are unclear. We previously identified 11 gestationally elevated maternal circulating miRNAs (HEamiRNAs) that predicted infant growth deficits following PAE. Here, we investigated whether these HEamiRNAs contribute to the pathology of PAE, by inhibiting trophoblast epithelial–mesenchymal transition (EMT), a pathway critical for placental development. We now report for the first time that PAE inhibits expression of placental pro-EMT pathway members in both rodents and primates, and that HEamiRNAs collectively, but not individually, mediate placental EMT inhibition. HEamiRNAs collectively, but not individually, also inhibited cell proliferation and the EMT pathway in cultured trophoblasts, while inducing cell stress, and following trophoblast syncytialization, aberrant endocrine maturation. Moreover, a single intravascular administration of the pooled murine-expressed HEamiRNAs, to pregnant mice, decreased placental and fetal growth and inhibited the expression of pro-EMT transcripts in the placenta. Our data suggest that HEamiRNAs collectively interfere with placental development, contributing to the pathology of PAE, and perhaps also, to other causes of fetal growth restriction.
Collapse
Affiliation(s)
- Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Alan B Wells
- Clinical and Translational Research Institute, University of California San Diego, San Diego, CA, USA.,Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Nihal A Salem
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Andrea M Allan
- Department of Neurosciences, University of New Mexico, Albuquerque, NM, USA
| | - Victoria Hj Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Natali Newman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Nicole Ar Walter
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Christopher D Kroenke
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Lisa K Akison
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Karen M Moritz
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Christina D Chambers
- Clinical and Translational Research Institute, University of California San Diego, San Diego, CA, USA .,Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX, USA
| | | |
Collapse
|
12
|
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
|
13
|
Zhang J, Mo HQ, Tian FJ, Zeng WH, Liu XR, Ma XL, Li X, Qin S, Fan CF, Lin Y. EIF5A1 promotes trophoblast migration and invasion via ARAF-mediated activation of the integrin/ERK signaling pathway. Cell Death Dis 2018; 9:926. [PMID: 30206208 PMCID: PMC6134074 DOI: 10.1038/s41419-018-0971-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/03/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
Abstract
Trophoblast dysfunction is one mechanism implicated in the etiology of recurrent miscarriage (RM). Regulation of trophoblast function, however, is complex and the mechanisms contributing to dysregulation remain to be elucidated. Herein, we found EIF5A1 expression levels to be significantly decreased in cytotrophoblasts in RM villous tissues compared with healthy controls. Using the HTR-8/SVneo cell line as a model system, we found that overexpression of EIF5A1 promotes trophoblast proliferation, migration and invasion in vitro. Knockdown of EIF5A1 or inhibiting its hypusination with N1-guanyl-1,7-diaminoheptane (GC7) suppresses these activities. Similarly, mutating EIF5A1 to EIF5A1K50A to prevent hypusination abolishes its effects on proliferation, migration and invasion. Furthermore, upregulation of EIF5A1 increases the outgrowth of trophoblasts in a villous explant culture model, whereas knockdown has the opposite effect. Suppression of EIF5A1 hypusination also inhibits the outgrowth of trophoblasts in explants. Mechanistically, ARAF mediates the regulation of trophoblast migration and invasion by EIF5A1. Hypusinated EIF5A1 regulates the integrin/ERK signaling pathway via controlling the translation of ARAF. ARAF level is also downregulated in trophoblasts of RM villous tissues and expression of ARAF is positively correlated with EIF5A1. Together, our results suggest that EIF5A1 may be a regulator of trophoblast function at the maternal-fetal interface and low levels of EIF5A1 and ARAF may be associated with RM.
Collapse
Affiliation(s)
- Jing Zhang
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Hui-Qin Mo
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Fu-Ju Tian
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Wei-Hong Zeng
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Xiao-Rui Liu
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Xiao-Ling Ma
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Xiao Li
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Shi Qin
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Cui-Fang Fan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Yi Lin
- International Peace Maternity & Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China.
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China.
| |
Collapse
|
14
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
15
|
Na JY, Seok J, Park S, Kim JS, Kim GJ. Effects of selenium on the survival and invasion of trophoblasts. Clin Exp Reprod Med 2018; 45:10-16. [PMID: 29662820 PMCID: PMC5897242 DOI: 10.5653/cerm.2018.45.1.10] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 11/29/2017] [Accepted: 01/23/2018] [Indexed: 01/29/2023] Open
Abstract
Objective Placental oxidative stress is known to be a factor that contributes to pregnancy failure. The aim of this study was to determine whether selenium could induce antioxidant gene expression and regulate invasive activity and mitochondrial activity in trophoblasts, which are a major cell type of the placenta. Methods To understand the effects of selenium on trophoblast cells exposed to hypoxia, the viability and invasive activity of trophoblasts were analyzed. The expression of antioxidant enzymes was assessed by reverse-transcription polymerase chain reaction. In addition, the effects of selenium treatment on mitochondrial activity were evaluated in terms of adenosine triphosphate production, mitochondrial membrane potential, and reactive oxygen species levels. Results Selenium showed positive effects on the viability and migration activity of trophoblast cells when exposed to hypoxia. Interestingly, the increased heme oxygenase 1 expression under hypoxic conditions was decreased by selenium treatment, whereas superoxide dismutase expression was increased in trophoblast cells by selenium treatment for 72 hours, regardless of hypoxia. Selenium-treated trophoblast cells showed increased mitochondrial membrane potential and decreased reactive oxygen species levels under hypoxic conditions for 72 hours. Conclusion These results will be used as basic data for understanding the mechanism of how trophoblast cells respond to oxidative stress and how selenium promotes the upregulation of related genes and improves the survival rate and invasive ability of trophoblasts through regulating mitochondrial activity. These results suggest that selenium may be used in reproductive medicine for purposes including infertility treatment.
Collapse
Affiliation(s)
- Jee Yoon Na
- Cheongshim International Academy, Gapyeong, Korea
| | - Jin Seok
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | - Sohae Park
- Department of Biomedical Science, CHA University, Seongnam, Korea
| | | | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, Korea
| |
Collapse
|
16
|
Kong DB, Chen F, Sima N. Focal adhesion kinases crucially regulate TGFβ-induced migration and invasion of bladder cancer cells via Src kinase and E-cadherin. Onco Targets Ther 2017; 10:1783-1792. [PMID: 28367061 PMCID: PMC5370070 DOI: 10.2147/ott.s122463] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor protein-tyrosine kinase that is triggered off by special extracellular signals such as some growth factors and integrins. FAK is found in cell-matrix attachment sites and implicated in cell migration, invasion, movement, gene expression, survival and apoptosis. In this study, we aimed to investigate whether FAK plays a role in invasion and migration of bladder cancer cells. Using an FAK-specific small interfering RNA (siRNA) and an FAK inhibitor PF-228, we found that inhibition of FAK tyrosine phosphorylation or knockdown of FAK suppressed invasion and migration of bladder cancer cells. Src is an important mediator of FAK-regulated migratory and invasive activity. Tyrosine phosphorylation of Src and FAK is mutually dependent and plays a key role in transforming growth factor beta (TGFβ)-induced invasion and migration. E-cadherin acts downstream of FAK and is a critical negative regulator in FAK-regulated invasion and migration of bladder cancer cells. These findings imply that FAK is involved in oncogenic signaling of invasion and migration, which can be a novel therapeutic target to treat patients with bladder cancer.
Collapse
Affiliation(s)
- De-Bo Kong
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang
| | - Feng Chen
- Department of Surgery, The Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, Jiangxi
| | - Ni Sima
- Women's Reproductive Health Key Laboratory of Zhejiang Province, Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
17
|
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
|
18
|
Hypoxia Inducible Factor-1α Regulates the Migration of Bone Marrow Mesenchymal Stem Cells via Integrin α 4. Stem Cells Int 2016; 2016:7932185. [PMID: 26880989 PMCID: PMC4736322 DOI: 10.1155/2016/7932185] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/06/2015] [Accepted: 10/12/2015] [Indexed: 12/13/2022] Open
Abstract
Although hypoxic environments have been known to regulate the migratory ability of bone marrow-derived mesenchymal stem cells (BM-MSCs), which is a critical factor for maximizing the therapeutic effect, the underlying mechanisms remain unclear. Therefore, we aimed to confirm the effect of hypoxia-inducible factor-1α (HIF-1α) on the migration of BM-MSCs and to analyze the interaction between HIF-1α and integrin-mediated signals. Hypoxia-activated HIF-1α significantly increased BM-MSC migration. The expression of integrin α4 was decreased in BM-MSCs by increased HIF-1α under hypoxia, whereas the expression of Rho-associated kinase 1 (ROCK1) and Rac1/2/3 was increased. After downregulation of HIF-1α by YC-1, which is an inhibitor of HIF-1α, BM-MSC migration was decreased via upregulation of integrin α4 and downregulation of ROCK1 and Rac1/2/3. Knockdown of integrin α4 by integrin α4 siRNA (siITGA4) treatment increased BM-MSC migration by upregulation of ROCK1, Rac1/2/3, and matrix metalloproteinase-2 regardless of oxygen tension. Moreover, siITGA4 treatment increased HIF-1α expression and augmented the translocation of HIF-1α into the nucleus under hypoxia. Taken together, the alternative expression of HIF-1α induced by microenvironment factors, such as hypoxia and integrin α4, may regulate the migration of BM-MSCs. These findings may provide insights to the underlying mechanisms of BM-MSC migration for successful stem cell-based therapy.
Collapse
|
19
|
Differentiation of first trimester cytotrophoblast to extravillous trophoblast involves an epithelial-mesenchymal transition. Placenta 2015; 36:1412-8. [PMID: 26545962 DOI: 10.1016/j.placenta.2015.10.013] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 12/30/2022]
Abstract
The transformation of cytotrophoblast (CTB) to extravillous trophoblast (EVT) is an essential process for placental implantation. EVT generated at the tips of the anchoring villi migrate away from the placenta and invade the endometrium and maternal spiral arteries, where they modulate maternal immune responses and remodel the arteries into high-volume conduits to facilitate uteroplacental blood flow. The process of EVT differentiation has several factors in common with the epithelial-to-mesenchymal transition (EMT) observed in embryonic development, wound healing and cancer metastasis. We hypothesized that the generation of invasive EVT from CTB was a form of EMT. We isolated paired CTB and EVT from first trimester placentae, and compared their gene expression using a PCR array comprising probes for genes involved in EMT. Out of 84 genes, 24 were down-regulated in EVT compared to CTB, including epithelial markers such as E-cadherin (-11-fold) and occludin (-75-fold). Another 30 genes were up-regulated in EVT compared to CTB including mesenchymal markers such as vimentin (235-fold) and fibronectin (107-fold) as well as the matrix metalloproteinases, MMP2 and MMP9 (357-fold, 129-fold). These alterations also included major increases in the ZEB2 (zinc finger E-box binding homeobox 2, 198-fold) and TCF4 (transcription factor 4, 18-fold) transcription factors, suggesting possible stimulatory mechanisms. There was substantial up-regulation of the genes encoding TGFβ1 and TGFβ2 (48-fold, 115-fold), which may contribute to the maintenance of the mesenchymal-like phenotype. We conclude that transformation of CTB to EVT is consistent with an EMT, although the differences with other types of EMT suggest this may be a unique form.
Collapse
|
20
|
Liu L, Wang Y, Shen C, He J, Liu X, Ding Y, Gao R, Chen X. Benzo(a)pyrene inhibits migration and invasion of extravillous trophoblast HTR-8/SVneo cells via activation of the ERK and JNK pathway. J Appl Toxicol 2015; 36:946-55. [PMID: 26359795 DOI: 10.1002/jat.3227] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 08/02/2015] [Accepted: 08/02/2015] [Indexed: 12/31/2022]
Abstract
Benzo(a)pyrene (BaP) is a persistent organic pollutant (POP) that is a serious threat to human health. Numerous studies have shown that BaP causes adverse effects in pregnancy, but the mechanism remains unclear. The moderate invasion of trophoblast cells into the endometrium is an important factor during successful embryo implantation. The aim of this study was to investigate the effect and mechanism of BaP on the invasion and migration of trophoblast cells. HTR-8/SVneo cells were treated with different concentrations (1, 5, 10, 25, 50 and 100 μM) of BaP. The invasion and migration of HTR-8/SVneo cells were observed after BaP treatment. The protein levels related to migration and invasion was detected by Western blot. The results confirmed that BaP inhibits the migration and invasion of extravillous trophoblast HTR-8/SVneo cells. Further investigations indicated that the protein levels of MMP-2, MMP-9 and E-cadherin in HTR-8/SVneo cells were changed by BaP treatment. Moreover, the data demonstrated that BaP activated the MAPK signaling pathway. Pretreatment with specific inhibitors of MAPK rescued BaP-induced change in the migration and invasion of HTR-8/SVneo cells. Taken together, our results indicated that BaP inhibits invasion and the migration of HTR-8/SVneo cells, which might cause a failure in early pregnancy. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Liyuan Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Cha Shen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No.1 Yixueyuan Road, Yuzhong District, 400016, Chongqing, China
| |
Collapse
|
21
|
Ren Y, Hao P, Law SKA, Sze SK. Hypoxia-induced changes to integrin α 3 glycosylation facilitate invasion in epidermoid carcinoma cell line A431. Mol Cell Proteomics 2014; 13:3126-37. [PMID: 25078904 DOI: 10.1074/mcp.m114.038505] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is a critical microenvironmental factor that drives cancer progression through angiogenesis and metastasis. Glycoproteins, especially those on the plasma membrane, orchestrate this process; however, questions remain regarding hypoxia-perturbed protein glycosylation in cancer cells. We focused on the effects of hypoxia on the integrin family of glycoproteins, which are central to the cellular processes of attachment and migration and have been linked with cancer in humans. We employed electrostatic repulsion hydrophilic interaction chromatography coupled with iTRAQ labeling and LC-MS/MS to identify and quantify glycoproteins expressed in A431. The results revealed that independent of the protein-level change, N-glycosylation modifications of integrin α 3 (ITGA3) were inhibited by hypoxia, unlike in other integrin subunits. A combination of Western blot, flow cytometry, and cell staining assays showed that hypoxia-induced alterations to the glycosylation of ITGA3 prevented its efficient translocation to the plasma membrane. Mutagenesis studies demonstrated that simultaneous mutation of glycosites 6 and 7 of ITGA3 prevented its accumulation at the K562 cell surface, which blocked integrin α 3 and β 1 heterodimer formation and thus abolished ITGA3's interaction with extracellular ligands. By generating A431 cells stably expressing ITGA3 mutated at glycosites 6 and 7, we showed that lower levels of ITGA3 on the cell surface, as induced by hypoxia, conferred an increased invasive ability to cancer cells in vitro under hypoxic conditions. Taken together, these results revealed that ITGA3 translocation to the plasma membrane suppressed by hypoxia through inhibition of glycosylation facilitated cell invasion in A431.
Collapse
Affiliation(s)
- Yan Ren
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr., Singapore 637551, Singapore
| | - Piliang Hao
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr., Singapore 637551, Singapore
| | - S K Alex Law
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr., Singapore 637551, Singapore
| | - Siu Kwan Sze
- From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr., Singapore 637551, Singapore.
| |
Collapse
|
22
|
Mierke CT. The role of focal adhesion kinase in the regulation of cellular mechanical properties. Phys Biol 2013; 10:065005. [PMID: 24304934 DOI: 10.1088/1478-3975/10/6/065005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The regulation of mechanical properties is necessary for cell invasion into connective tissue or intra- and extravasation through the endothelium of blood or lymph vessels. Cell invasion is important for the regulation of many healthy processes such as immune response reactions and wound healing. In addition, cell invasion plays a role in disease-related processes such as tumor metastasis and autoimmune responses. Until now the role of focal adhesion kinase (FAK) in regulating mechanical properties of cells and its impact on cell invasion efficiency is still not well known. Thus, this review focuses on mechanical properties regulated by FAK in comparison to the mechano-regulating protein vinculin. Moreover, it points out the connection between cancer cell invasion and metastasis and FAK by showing that FAK regulates cellular mechanical properties required for cellular motility. Furthermore, it sheds light on the indirect interaction of FAK with vinculin by binding to paxillin, which then impairs the binding of paxillin to vinculin. In addition, this review emphasizes whether FAK fulfills regulatory functions similar to vinculin. In particular, it discusses the differences and the similarities between FAK and vinculin in regulating the biomechanical properties of cells. Finally, this paper highlights that both focal adhesion proteins, vinculin and FAK, synergize their functions to regulate the mechanical properties of cells such as stiffness and contractile forces. Subsequently, these mechanical properties determine cellular invasiveness into tissues and provide a source sink for future drug developments to inhibit excessive cell invasion and hence, metastases formation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Biological Physics Division, University of Leipzig, Linnéstr. 5, D-04103 Leipzig, Germany
| |
Collapse
|
23
|
An integrative view on the physiology of human early placental villi. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 114:33-48. [PMID: 24291663 DOI: 10.1016/j.pbiomolbio.2013.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 11/20/2013] [Indexed: 01/08/2023]
Abstract
The placenta is an indispensable organ for intrauterine protection, development and growth of the embryo and fetus. It provides tight contact between mother and conceptus, enabling the exchange of gas, nutrients and waste products. The human placenta is discoidal in shape, and bears a hemo-monochorial interface as well as villous materno-fetal interdigitations. Since Peter Medawar's astonishment to the paradoxical nature of the mother-fetus relationship in 1953, substantial knowledge in the domain of placental physiology has been gathered. In the present essay, an attempt has been made to build an integrated understanding of morphological dynamics, cell biology, and functional aspects of genomic and proteomic expression of human early placental villous trophoblast cells followed by a commentary on the future directions of research in this field.
Collapse
|
24
|
Zhou J, Xiao D, Hu Y, Wang Z, Paradis A, Mata-Greenwood E, Zhang L. Gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 signaling in pregnant rats. Hypertension 2013; 62:599-607. [PMID: 23817493 DOI: 10.1161/hypertensionaha.113.01449] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Preeclampsia is a life-threatening pregnancy disorder. However, its pathogenesis remains unclear. We tested the hypothesis that gestational hypoxia induces preeclampsia-like symptoms via heightened endothelin-1 (ET-1) signaling. Time-dated pregnant and nonpregnant rats were divided into normoxic and hypoxic (10.5% O2 from the gestational day 6-21) groups. Chronic hypoxia had no significant effect on blood pressure or proteinuria in nonpregnant rats but significantly increased blood pressure on day 12 (systolic blood pressure, 111.7 ± 6.1 versus 138.5 ± 3.5 mm Hg; P=0.004) and day 20 (systolic blood pressure, 103.4 ± 4.6 versus 125.1 ± 6.1 mm Hg; P=0.02) in pregnant rats and urine protein (μg/μL)/creatinine (nmol/μL) ratio on day 20 (0.10 ± 0.01 versus 0.20 ± 0.04; P=0.04), as compared with the normoxic control group. This was accompanied with asymmetrical fetal growth restriction. Hypoxia resulted in impaired trophoblast invasion and uteroplacental vascular remodeling. In addition, plasma ET-1 levels, as well as the abundance of prepro-ET-1 mRNA, ET-1 type A receptor and angiotensin II type 1 receptor protein in the kidney and placenta were significantly increased in the chronic hypoxic group, as compared with the control animals. Treatment with the ET-1 type A receptor antagonist, BQ123, during the course of hypoxia exposure significantly attenuated the hypoxia-induced hypertension and other preeclampsia-like features. The results demonstrate that chronic hypoxia during gestation induces preeclamptic symptoms in pregnant rats via heightened ET-1 and ET-1 type A receptor-mediated signaling, providing a molecular mechanism linking gestational hypoxia and increased risk of preeclampsia.
Collapse
Affiliation(s)
- Jianjun Zhou
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Ren Y, Hao P, Dutta B, Cheow ESH, Sim KH, Gan CS, Lim SK, Sze SK. Hypoxia modulates A431 cellular pathways association to tumor radioresistance and enhanced migration revealed by comprehensive proteomic and functional studies. Mol Cell Proteomics 2012. [PMID: 23204318 DOI: 10.1074/mcp.m112.018325] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumor hypoxia induces cancer cell angiogenesis, invasiveness, treatment resistance, and contributes to poor clinical outcome. However, the molecular mechanism by which tumor hypoxia exerts a coordinated effect on different molecular pathways to enhance tumor growth and survival and lead to poor clinical outcome is not fully understood. In this study, we attempt to elucidate the global protein expression and functional changes in A431 epithelial carcinoma cells induced by hypoxia and reoxygenation using iTRAQ quantitative proteomics and biochemical functional assays. Quantitative proteomics results showed that 4316 proteins were quantified with FDR<1%, in which over 1200 proteins were modulated >1.2 fold, and DNA repair, glycolysis, integrin, glycoprotein turnover, and STAT1 pathways were perturbed by hypoxia and reoxygenation-induced oxidative stress. For the first time, hypoxia was shown to up-regulate the nonhomologous end-joining pathway, which plays a central role in DNA repair of irradiated cells, thereby potentially contributing to the radioresistance of hypoxic A431 cells. The up-regulation of Ku70/Ku80 dimer, a key molecular complex in the nonhomologous end-joining pathway, was confirmed by Western blot and liquid chromatography/tandem mass spectrometry-MRM methods. Functional studies confirmed that up-regulation of glycolysis, integrin, glycoprotein synthesis, and down-regulation of STAT1 pathways during hypoxia enhanced metastastic activity of A431 cells. Migration of A431 cells was dramatically repressed by glycolysis inhibitor (2-Deoxy-d-glucose), glycoprotein synthesis inhibitor (1-Deoxynojirimycin Hydrochloride), and STAT1α overexpression that enhanced the integrin-mediated cell adhesion. These results revealed that hypoxia induced several biological processes involved in tumor migration and radioresistance and provided potential new targets for tumor therapy.
Collapse
Affiliation(s)
- Yan Ren
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Tang CL, Zhao HB, Li MQ, Du MR, Meng YH, Li DJ. Focal Adhesion Kinase Signaling is Necessary for the Cyclosporin A-Enhanced Migration and Invasion of Human Trophoblast Cells. Placenta 2012; 33:704-11. [DOI: 10.1016/j.placenta.2012.06.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
|
27
|
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
|
28
|
Zhang X, Yan G, Diao Z, Sun H, Hu Y. NUR77 inhibits the expression of TIMP2 and increases the migration and invasion of HTR-8/SVneo cells induced by CYR61. Placenta 2012; 33:561-7. [PMID: 22551568 DOI: 10.1016/j.placenta.2012.04.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 03/15/2012] [Accepted: 04/05/2012] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the function and mechanism of CYR61 on the migration and invasion of the trophoblast cell line, HTR-8/SVneo cells. STUDY DESIGN The mRNA and protein levels of NUR77 in the placentas of normal and preeclampsia (PE) women were evaluated using real-time PCR and Western blot, respectively. Paraffin-embedded tissues were processed for localization of NUR77 protein in placental villus by immunohistochemistry. HTR-8/SVneo cells were cultured in the presence of CYR61, Ad-NUR77 or a small interfering RNA for NUR77 (Ad-sinur77). The expression of NUR77 in the HTR-8/SVneo cells was detected and the effects of CYR61 on the migration and invasion of HTR-8/SVneo cells were assessed in wound-healing and transwell experiments, respectively. Gelatin zymography was used to measure the MMP2 release in HTR-8/SVneo cells. RESULTS NUR77 is significantly decreased in the placenta of women with PE compared with the levels during a normal pregnancy. CYR61 can significantly increase the expression of NUR77 in HTR-8/SVneo cells. CYR61, as well as NUR77, can promote HTR-8/SVneo cells migration and invasion, which can be blocked by Ad-sinur77. Both CYR61 and Ad-nur77 reduced the mRNA expression of TIMP2 in HTR-8/SVneo cells. CONCLUSIONS CYR61 may promote HTR-8/SVneo cells migration and invasion through the upregulation of NUR77, leading to the increase of MMP2 release and the downregulation of TIMP2 expression.
Collapse
Affiliation(s)
- X Zhang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | | | | | | | | |
Collapse
|
29
|
Wallace AE, Fraser R, Cartwright JE. Extravillous trophoblast and decidual natural killer cells: a remodelling partnership. Hum Reprod Update 2012; 18:458-71. [PMID: 22523109 DOI: 10.1093/humupd/dms015] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND During pregnancy, maternal uterine spiral arteries (SAs) are remodelled from minimal-flow, high-resistance vessels into larger diameter vessels with low resistance and high flow. Fetal extravillous trophoblasts (EVT) have important roles in this process. Decidual natural killer cells (dNK cells) are the major maternal immune component of the decidua and accumulate around SAs before trophoblast invasion. A role for dNK cells in vessel remodelling is beginning to be elucidated. This review examines the overlapping and dissimilar mechanisms used by EVT and dNK cells in this process and how this may mirror another example of tissue remodelling, namely cancer development. METHODS The published literature was searched using Pubmed focusing on EVT, dNK cells and SA remodelling. Additional papers discussing cancer development are also included. RESULTS Similarities exist between actions carried out by dNK cells and EVT. Both interact with vascular cells lining the SA, as well as with each other, to promote transformation of the SA. EVT differentiation has previously been likened to the epithelial-mesenchymal transition in cancer cells, and we discuss how dNK-EVT interactions at the maternal-fetal interface can also be compared with the roles of immune cells in cancer. CONCLUSIONS The combined role that dNK cells and EVT play in SA remodelling suggests that these interactions could be described as a partnership. The investigation of pregnancy as a multicellular system involving both fetal and maternal components, as well as comparisons to similar examples of tissue remodelling, will further identify the key mechanisms in SA remodelling that are required for a successful pregnancy.
Collapse
Affiliation(s)
- Alison E Wallace
- Reproductive and Cardiovascular Disease Research Group, Division of Biomedical Sciences, St George's University of London, London, UK.
| | | | | |
Collapse
|
30
|
Fedorova L, Gatto-Weis C, Smaili S, Khurshid N, Shapiro JI, Malhotra D, Horrigan T. Down-regulation of the transcription factor snail in the placentas of patients with preeclampsia and in a rat model of preeclampsia. Reprod Biol Endocrinol 2012; 10:15. [PMID: 22360878 PMCID: PMC3298516 DOI: 10.1186/1477-7827-10-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 02/23/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Placental malfunction in preeclampsia is believed to be a consequence of aberrant differentiation of trophoblast lineages and changes in utero-placental oxygenation. The transcription factor Snail, a master regulator molecule of epithelial-mesenchymal transition in embryonic development and in cancer, is shown to be involved in trophoblast differentiation as well. Moreover, Snail can be controlled by oxidative stress and hypoxia. Therefore, we examined the expression of Snail and its downstream target, e-cadherin, in human normal term, preterm and preeclamptic placentas, and in pregnant rats that developed preeclampsia-like symptoms in the response to a 20-fold increase in sodium intake. METHODS Western blotting analysis was used for comparative expression of Snail and e- cadherin in total protein extracts. Placental cells expressing Snail and e-cadherin were identified by immunohistochemical double-labeling technique. RESULTS The levels of Snail protein were decreased in human preeclamptic placentas by 30% (p < 0.01) compared to normal term, and in the rat model by 40% (p < 0.001) compared to control placentas. In preterm placentas, the levels of Snail expression varied, yet there was a strong trend toward statistical significance between preterm and preeclamptic placentas. In humans, e-cadherin protein level was 30% higher in preeclamptic (p < 0.05) placentas and similarly, but not significantly (p = 0.1), high in the preterm placentas compared to normal term. In the rat model of preeclampsia, e-cadherin was increased by 60% (p < 0.01). Immunohistochemical examination of human placentas demonstrated Snail-positive staining in the nuclei of the villous trophoblasts and mesenchymal cells and in the invasive trophoblasts of the decidua. In the rat placenta, the majority of Snail positive cells were spongiotrophoblasts of the junctional zone, while in the labyrinth, Snail-positive sinusoidal giant trophoblasts cells were found in some focal areas located close to the junctional zone. CONCLUSION We demonstrated that human preeclampsia and the salt-induced rat model of preeclampsia are associated with the reduced levels of Snail protein in placenta. Down-regulation of the transcription factor Snail in placental progenitor cell lineages, either by intrinsic defects and/or by extrinsic and maternal factors, may affect normal placenta development and function and thus contribute to the pathology of preeclampsia.
Collapse
Affiliation(s)
- Larisa Fedorova
- Department of Medicine, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Cara Gatto-Weis
- Department of Pathology, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Sleiman Smaili
- Department of Obstetrics and Gynecology, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Nauman Khurshid
- Department of Obstetrics and Gynecology, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Joseph I Shapiro
- Department of Medicine, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Deepak Malhotra
- Department of Medicine, University of Toledo School of Medicine, Toledo, OH 43614, USA
| | - Terrence Horrigan
- Department of Obstetrics and Gynecology, University of Toledo School of Medicine, Toledo, OH 43614, USA
| |
Collapse
|
31
|
Leduc K, Bourassa V, Asselin É, Leclerc P, Lafond J, Reyes-Moreno C. Leukemia Inhibitory Factor Regulates Differentiation of Trophoblastlike BeWo Cells Through the Activation of JAK/STAT and MAPK3/1 MAP Kinase-Signaling Pathways1. Biol Reprod 2012; 86:54. [DOI: 10.1095/biolreprod.111.094334] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
32
|
Na KH, Lee HJ, Choi JH, Eun JW, Nam SW, Yoon TK, Kim GJ. Dynamic alterations in integrin α4 expression by hypoxia are involved in trophoblast invasion during early implantation. J Cell Biochem 2012; 113:685-94. [DOI: 10.1002/jcb.23398] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Baranwal S, Wang Y, Rathinam R, Lee J, Jin L, McGoey R, Pylayeva Y, Giancotti F, Blobe GC, Alahari SK. Molecular characterization of the tumor-suppressive function of nischarin in breast cancer. J Natl Cancer Inst 2011; 103:1513-28. [PMID: 21917605 DOI: 10.1093/jnci/djr350] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Nischarin (encoded by NISCH), an α5 integrin-binding protein, has been identified as a regulator of breast cancer cell invasion. We hypothesized that it might be a tumor suppressor and were interested in its regulation. METHODS We examined nischarin expression in approximately 300 human breast cancer and normal tissues using quantitative polymerase chain reaction and immunohistochemistry. Loss of heterozygosity analysis was performed by examining three microsatellite markers located near the NISCH locus in normal and tumor tissues. We generated derivatives of MDA-MB-231 human metastatic breast cancer cells that overexpressed nischarin and measured tumor growth from these cells as xenografts in mice; metastasis by these cells after tail vein injection; and α5 integrin expression, Rac, and focal adhesion kinase (FAK) signaling using western blotting. We also generated clones of MCF-7 human breast cancer cells in which nischarin expression was silenced and measured tumor growth in mouse xenograft models (n = 5 for all mouse experiments). P values were from two-sided Student t tests in pairwise comparisons. RESULTS Normal human breast tissue samples had statistically significantly higher expression of nischarin mRNA compared with tumor tissue samples (mean level in normal breast = 50.7 [arbitrary units], in breast tumor = 16.49 [arbitrary units], difference = 34.21, 95% confidence interval [CI] = 11.63 to 56.79, P = .003), and loss of heterozygosity was associated with loss of nischarin expression. MDA-MB-231 cells in which nischarin was overexpressed had statistically significantly reduced tumor growth and metastasis compared with parental MDA-MB-231 cells (mean volume at day 40, control vs nischarin-expressing tumors, 1977 vs 42.27 mm(3), difference = 1935 mm(3), 95% CI = 395 to 3475 mm(3), P = .025). Moreover, MCF-7 tumor xenografts in which nischarin expression was silenced grew statistically significantly faster than parental cells (mean volume at day 63, tumors with scrambled short hairpin RNA [shRNA] vs with nischarin shRNA, 224 vs 1262 mm(3), difference = 1038 mm(3), 95% CI = 899.6 to 1176 mm(3), P < .001). Overexpression of nischarin was associated with decreased α5 integrin expression, FAK phosphorylation, and Rac activation. CONCLUSION Nischarin may be a novel tumor suppressor that limits breast cancer progression by regulating α5 integrin expression and subsequently α5 integrin-, FAK-, and Rac-mediated signaling.
Collapse
Affiliation(s)
- Somesh Baranwal
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, Louisiana State University School Of Medicine, 1901 Perdido St, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hypoxia impairs primordial germ cell migration in zebrafish (Danio rerio) embryos. PLoS One 2011; 6:e24540. [PMID: 21931746 PMCID: PMC3169607 DOI: 10.1371/journal.pone.0024540] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 08/12/2011] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND As a global environmental concern, hypoxia is known to be associated with many biological and physiological impairments in aquatic ecosystems. Previous studies have mainly focused on the effect of hypoxia in adult animals. However, the effect of hypoxia and the underlying mechanism of how hypoxia affects embryonic development of aquatic animals remain unclear. METHODOLOGY/PRINCIPAL FINDINGS In the current study, the effect of hypoxia on primordial germ cell (PGC) migration in zebrafish embryos was investigated. Hypoxic embryos showed PGC migration defect as indicated by the presence of mis-migrated ectopic PGCs. Insulin-like growth factor (IGF) signaling is required for embryonic germ line development. Using real-time PCR, we found that the mRNA expression levels of insulin-like growth factor binding protein (IGFBP-1), an inhibitor of IGF bioactivity, were significantly increased in hypoxic embryos. Morpholino knockdown of IGFBP-1 rescued the PGC migration defect phenotype in hypoxic embryos, suggesting the role of IGFBP-1 in inducing PGC mis-migration. CONCLUSIONS/SIGNIFICANCE This study provides novel evidence that hypoxia disrupts PGC migration during embryonic development in fish. IGF signaling is shown to be one of the possible mechanisms for the causal link between hypoxia and PGC migration. We propose that hypoxia causes PGC migration defect by inhibiting IGF signaling through the induction of IGFBP-1.
Collapse
|
35
|
Sun YY, Lu M, Xi XW, Qiao QQ, Chen LL, Xu XM, Feng YJ. Regulation of epithelial-mesenchymal transition by homeobox gene DLX4 in JEG-3 trophoblast cells: a role in preeclampsia. Reprod Sci 2011; 18:1138-45. [PMID: 21602546 DOI: 10.1177/1933719111408112] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The pathogenesis of preeclampsia is unclear but is thought to be related to shallow trophoblast invasion. An invasive phenotype is acquired by trophoblasts through the process of epithelial-mesenchymal transition (EMT). We proposed that EMT in trophoblasts is deregulated in preeclampsia. The homeobox gene DLX4 plays an important role in epithelial-mesenchymal interactions during embryonic and placental development. To elucidate the role of DLX4 in trophoblast EMT and preeclampsia, we investigated the expression of DLX4 in preeclampsia-affected placentas and the effect of DLX4 on EMT in trophoblast-derived JEG-3 cells. DLX4 expression was downregulated in preeclampsia-affected placentas and hypoxic JEG-3 cells. Knockdown of DLX4 by RNA interference (RNAi) inhibited the motility and invasion ability of JEG-3 cells, decreased the expression of E-cadherin, and upregulated the expression of the E-cadherin repressor Snail. Our findings suggest that decreased expression of DLX4 leads to the pathogenesis of preeclampsia by inhibiting EMT in trophoblasts and provides new insight into the pathophysiological mechanism of preeclampsia.
Collapse
Affiliation(s)
- Yun-Yan Sun
- Department of Obstetrics and Gynecology, Affiliated First Hospital of Shanghai Jiao Tong University, Shanghai, China.
| | | | | | | | | | | | | |
Collapse
|
36
|
Wagner PK, Otomo A, Christians JK. Regulation of pregnancy-associated plasma protein A2 (PAPPA2) in a human placental trophoblast cell line (BeWo). Reprod Biol Endocrinol 2011; 9:48. [PMID: 21496272 PMCID: PMC3096916 DOI: 10.1186/1477-7827-9-48] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 04/15/2011] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Pregnancy-associated plasma protein A2 (PAPPA2) is an insulin-like growth factor-binding protein (IGFBP) protease expressed at high levels in the placenta and upregulated in pregnancies complicated by preeclampsia and HELLP (Hemolytic anemia, Elevated Liver enzymes, and Low Platelet count) syndrome. However, it is unclear whether elevated PAPPA2 expression causes abnormal placental development, or whether upregulation compensates for placental pathology. In the present study, we investigate whether PAPPA2 expression is affected by hypoxia, oxidative stress, syncytialization factors or substances known to affect the expression of PAPPA2's paralogue, PAPPA. METHODS BeWo cells, a model of placental trophoblasts, were treated with one of the following: hypoxia (2% O2), oxidative stress (20 microM hydrogen peroxide), forskolin (10 microM and 100 microM), TGF-beta (10 and 50 ng/mL), TNF-alpha (100 ng/mL), IL-1beta (100 ng/mL) or PGE2 (1 microM). We used quantitative RT-PCR (qRT-PCR) to quantify the mRNA levels of PAPPA2, as well as those of PAPPA and ADAM12 since these proteases have similar substrates and are also highly expressed in the placenta. Where we observed significant effects on PAPPA2 mRNA levels, we tested for effects at the protein level using an in-cell Western assay. RESULTS Hypoxia, but not oxidative stress, caused a 47-fold increase in PAPPA2 mRNA expression, while TNF-alpha resulted in a 6-fold increase, and both of these effects were confirmed at the protein level. PGE2 resulted in a 14-fold upregulation of PAPPA2 mRNA but this was not reflected at the protein level. Forskolin, TGF-beta and IL-1beta had no significant effect on PAPPA2 mRNA expression. We observed no effects of any treatment on PAPPA or ADAM12 expression. CONCLUSION Our study demonstrates that factors previously known to be highly expressed in preeclamptic placentae (PGE2 and TNF-alpha), contribute to the upregulation of PAPPA2. Hypoxia, known to occur in preeclamptic placentae, also increased PAPPA2 expression. These results are consistent with the hypothesis that PAPPA2 is upregulated as a consequence of placental pathology, rather than elevated PAPPA2 levels being a cause of preeclampsia.
Collapse
Affiliation(s)
- Pamela K Wagner
- Simon Fraser University, Biological Sciences, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Aki Otomo
- Simon Fraser University, Biological Sciences, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Julian K Christians
- Simon Fraser University, Biological Sciences, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| |
Collapse
|
37
|
Abstract
The transcriptional inhibitor Snail is a critical regulator for epithelial-mesenchymal transition (EMT). Although low oxygen induces Snail transcription, thereby stimulating EMT, a direct role of hypoxia-inducible factor (HIF) in this process remains to be demonstrated. Here we show that hypoxia induces the expression of Snail via HIF. In silico analysis identified a potential hypoxia-response element (HRE) close to the minimal promoter of the human and mouse genome of the snail gene. Gel shift assays demonstrated that a specific hypoxia-inducible complex is formed with the putative HRE and that the complex contains HIF proteins. ChIP assays confirmed the interaction of HIF proteins with the putative HRE in vivo. Reporter gene analyses showed that the putative HRE responds to hypoxia in its natural position as well as in front of a heterologous promoter and that the HRE is directly activated by HIF-1α or HIF-2α. HIF knockdown with siRNA at 2% oxygen and overexpression of an oxygen-insensitive HIF (HIF-ΔODD) mutant at 21% oxygen showed that HIF regulates Snail activation and subsequent cell migration. Our findings identify snail as a HIF target gene and provide novel insights into the regulation of snail and hypoxia-induced EMT.
Collapse
Affiliation(s)
- Daochun Luo
- Physiology and Experimental Medicine, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8 Canada
| | | | | | | |
Collapse
|
38
|
He K, Zhao H, Wang Q, Pan Y. A comparative genome analysis of gene expression reveals different regulatory mechanisms between mouse and human embryo pre-implantation development. Reprod Biol Endocrinol 2010; 8:41. [PMID: 20459759 PMCID: PMC2876170 DOI: 10.1186/1477-7827-8-41] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 05/11/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Pre-implantation development is a crucial step in successful implantation and pregnancy in mammals. It has been studied in depth, but mostly in laboratory animal models. Less is known about the regulatory mechanism involved in the pre-implantation development in humans and about the comparative aspects. METHODS Here, we employed the microarray datasets from the public database library of GEO and applied comparative analysis of genome wide temporal gene expression data based on statistical analysis and functional annotation for both mouse and human, demonstrating the discordance between the regulatory mechanisms of both mouse and human pre-implantation development. RESULTS There were differences between mouse and human pre-implantation development both in the global gene expression pattern and in the expression changes of individual genes at each stage, including different major transient waves of transcription profiles and some stage-specific genes and significantly related pathways. There also appeared to be different functional changes from one stage to another between mouse and human. CONCLUSIONS The analysis presented here lead to interesting and complementary conclusions that the regulatory mechanism of human pre-implantation development is not completely the same as the mouse. Not as the fact that 1-cell to 2-cell stage is important for mouse pre-implantation development, the 4-cell stage and 8-cell stage are both essential for human. Unlike in mouse, of which most of pathways found were related to energy, RNA and protein metabolism, the identified pathways in human were mostly disease-related and associated with human pre-implantation embryonic development. All of these suggest that a further comparative analysis should be required for applying the result of mouse expression data to human research or therapy, particularly in pre-implantation developments. Our study provides several potential targets of genes and pathways for studying the regulatory mechanism of human pre-implantation development using mouse model.
Collapse
Affiliation(s)
- Kan He
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Lab for Veterinary Biotechnology, Shanghai, 200240, China
| | - Hongbo Zhao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Lab for Veterinary Biotechnology, Shanghai, 200240, China
| | - Qishan Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Lab for Veterinary Biotechnology, Shanghai, 200240, China
| | - Yuchun Pan
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Lab for Veterinary Biotechnology, Shanghai, 200240, China
| |
Collapse
|
39
|
Ryu MH, Park HM, Chung J, Lee CH, Park HR. Hypoxia-inducible factor-1alpha mediates oral squamous cell carcinoma invasion via upregulation of alpha5 integrin and fibronectin. Biochem Biophys Res Commun 2010; 393:11-5. [PMID: 20097172 DOI: 10.1016/j.bbrc.2010.01.060] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 01/20/2010] [Indexed: 12/27/2022]
Abstract
With progressive and rapid growth of malignant tumors, cancer cells in an ischemic condition are expected to develop an increased potential for local invasive growth. To address this hypothesis, we first examined the effect of hypoxia on the invasiveness of oral squamous cell carcinoma (OSCC) cells using the Matrigel invasion assay. We then investigated the effect of hypoxia on the protein and mRNA expression of alpha5 integrin and fibronectin, which are major factors involved in tumor cell invasion. We showed that (i) hypoxia increased the invasiveness of OSCC cells, (ii) alpha5 integrin and fibronectin protein and mRNA expression levels were increased in OSCC cells under hypoxic conditions, (iii) hypoxia stimulated autocrine secretion of fibronectin in OSCC cells, (iv) administration of siRNA(HIF-1alpha) caused a significant decrease in alpha5 integrin and fibronectin protein, confirming that HIF-1alpha plays a role in their induction, and (v) siRNA(HIF-1alpha) abrogated hypoxia-induced cell invasion. Collectively, these data suggest that hypoxia promotes OSCC cell invasion that is elicited by HIF-1alpha-dependent alpha5 integrin and fibronectin induction.
Collapse
Affiliation(s)
- Mi Heon Ryu
- Department of Oral Pathology, School of Dentistry, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan 626-870, South Korea
| | | | | | | | | |
Collapse
|
40
|
Lowin T, Straub RH, Neumann E, Bosserhoff A, Vogel C, Moissl C, Anders S, Müller-Ladner U, Schedel JÃ. Glucocorticoids increase α5 integrin expression and adhesion of synovial fibroblasts but inhibit ERK signaling, migration, and cartilage invasion. ACTA ACUST UNITED AC 2009; 60:3623-32. [DOI: 10.1002/art.24985] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|