1
|
Liu Y, Yang R, Zhang Y, Zhu Y, Bao W. ANGPTL4 functions as an oncogene through regulation of the ETV5/CDH5/AKT/MMP9 axis to promote angiogenesis in ovarian cancer. J Ovarian Res 2022; 15:131. [PMID: 36517864 PMCID: PMC9749186 DOI: 10.1186/s13048-022-01060-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/15/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Angiopoietin-like 4 (ANGPTL4) is highly expressed in a variety of neoplasms and promotes cancer progression. Nevertheless, the mechanism of ANGPTL4 in ovarian cancer (OC) metastasis remains unclear. This study aimeds to explore whether ANGPTL4 regulates OC progression and elucidate the underlying mechanism. METHODS ANGPTL4 expression in clinical patient tumor samples was determined by immunohistochemistry (IHC) and high-throughput sequencing. ANGPTL4 knockdown (KD) and the addition of exogeneous cANGPTL4 protein were used to investigate its function. An in vivo xenograft tumor experiment was performed by intraperitoneal injection of SKOV3 cells transfected with short hairpin RNAs (shRNAs) targeting ANGPTL4 in nude mice. Western blotting and qRT-PCR were used to detect the levels of ANGPTL4, CDH5, p-AKT, AKT, ETV5, MMP2 and MMP9 in SKOV3 and HO8910 cells transfected with sh-ANGPTL4 or shRNAs targeting ETV5. RESULTS Increased levels of ANGPTL4 were associated with poor prognosis and metastasis in OC and induced the angiogenesis and metastasis of OC cells both in vivo and in vitro. This tumorigenic effect was dependent on CDH5, and the expression levels of ANGPTL4 and CDH5 in human OC werepositively correlated. In addition, CDH5 activated p-AKT, and upregulated the expression of MMP2 and MMP9. We also found that the expression of ETV5 was upregulated by ANGPTL4, which could bind the promoter region of CDH5, leading to increased CDH5 expression. CONCLUSION Our data indicated that an increase in the ANGPTL4 level results in increased ETV5 expression in OC, leading to metastasis via activation of the CDH5/AKT/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Yinping Liu
- Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Gongyuandong Road, Qingpu District, 201700, Shanghai, P. R. China
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Rui Yang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China
| | - Yaping Zhu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China.
| | - Wei Bao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 200080, Shanghai, P. R. China.
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 85 Wujin Road, Hongkou, 201620, Shanghai, P.R. China.
| |
Collapse
|
2
|
Wang L, Yang Y, Ma H, Xie Y, Xu J, Near D, Wang H, Garbutt T, Li Y, Liu J, Qian L. Single-cell dual-omics reveals the transcriptomic and epigenomic diversity of cardiac non-myocytes. Cardiovasc Res 2022; 118:1548-1563. [PMID: 33839759 PMCID: PMC9074971 DOI: 10.1093/cvr/cvab134] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
AIMS The precise cellular identity and molecular features of non-myocytes (non-CMs) in a mammalian heart at a single-cell level remain elusive. Depiction of epigenetic landscape with transcriptomic signatures using the latest single-cell multi-omics has the potential to unravel the molecular programs underlying the cellular diversity of cardiac non-myocytes. Here, we characterized the molecular and cellular features of cardiac non-CM populations in the adult murine heart at the single-cell level. METHODS AND RESULTS Through single-cell dual omics analysis, we mapped the epigenetic landscapes, characterized the transcriptomic profiles and delineated the molecular signatures of cardiac non-CMs in the adult murine heart. Distinct cis-regulatory elements and trans-acting factors for the individual major non-CM cell types (endothelial cells, fibroblast, pericytes, and immune cells) were identified. In particular, unbiased sub-clustering and functional annotation of cardiac fibroblasts (FBs) revealed extensive FB heterogeneity and identified FB sub-types with functional states related to the cellular response to stimuli, cytoskeleton organization, and immune regulation, respectively. We further explored the function of marker genes Hsd11b1 and Gfpt2 that label major FB subpopulations and determined the distribution of Hsd11b1+ and Gfp2+ FBs in murine healthy and diseased hearts. CONCLUSIONS In summary, we characterized the non-CM cellular identity at the transcriptome and epigenome levels using single-cell omics approaches and discovered previously unrecognized cardiac fibroblast subpopulations with unique functional states.
Collapse
Affiliation(s)
- Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yuchen Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hong Ma
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yifang Xie
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jun Xu
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - David Near
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haofei Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tiffany Garbutt
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, 111 Mason Farm Rd, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
3
|
Han DS, Lee EO. Sp1 Plays a Key Role in Vasculogenic Mimicry of Human Prostate Cancer Cells. Int J Mol Sci 2022; 23:1321. [PMID: 35163245 PMCID: PMC8835864 DOI: 10.3390/ijms23031321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Sp1 transcription factor regulates genes involved in various phenomena of tumor progression. Vasculogenic mimicry (VM) is the alternative neovascularization by aggressive tumor cells. However, there is no evidence of the relationship between Sp1 and VM. This study investigated whether and how Sp1 plays a crucial role in the process of VM in human prostate cancer (PCa) cell lines, PC-3 and DU145. A cell viability assay and three-dimensional culture VM tube formation assay were performed. Protein and mRNA expression levels were detected by Western blot and reverse transcriptase-polymerase chain reaction, respectively. The nuclear twist expression was observed by immunofluorescence assay. A co-immunoprecipitation assay was performed. Mithramycin A (MiA) and Sp1 siRNA significantly decreased serum-induced VM, whereas Sp1 overexpression caused a significant induction of VM. Serum-upregulated vascular endothelial cadherin (VE-cadherin) protein and mRNA expression levels were decreased after MiA treatment or Sp1 silencing. The protein expression and the nuclear localization of twist were increased by serum, which was effectively inhibited after MiA treatment or Sp1 silencing. The interaction between Sp1 and twist was reduced by MiA. On the contrary, Sp1 overexpression enhanced VE-cadherin and twist expressions. Serum phosphorylated AKT and raised matrix metalloproteinase-2 (MMP-2) and laminin subunit 5 gamma-2 (LAMC2) expressions. MiA or Sp1 silencing impaired these effects. However, Sp1 overexpression upregulated phosphor-AKT, MMP-2 and LAMC2 expressions. Serum-upregulated Sp1 was significantly reduced by an AKT inhibitor, wortmannin. These results demonstrate that Sp1 mediates VM formation through interacting with the twist/VE-cadherin/AKT pathway in human PCa cells.
Collapse
Affiliation(s)
- Deok-Soo Han
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Eun-Ok Lee
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
4
|
Li L, Liu Q, Shang T, Song W, Xu D, Allen TD, Wang X, Jeong J, Lobe CG, Liu J. Aberrant Activation of Notch1 Signaling in Glomerular Endothelium Induces Albuminuria. Circ Res 2021; 128:602-618. [PMID: 33435713 DOI: 10.1161/circresaha.120.316970] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Glomerular capillaries are lined with a highly specialized fenestrated endothelium and contribute to the glomerular filtration barrier. The Notch signaling pathway is involved in regulation of glomerular filtration barrier, but its role in glomerular endothelium has not been investigated due to the embryonic lethality of animal models with genetic modification of Notch pathway components in the endothelium. OBJECTIVE To determine the effects of aberrant activation of the Notch signaling in glomerular endothelium and the underlying molecular mechanisms. METHODS AND RESULTS We established the ZEG-NICD1 (notch1 intracellular domain)/Tie2-tTA/Tet-O-Cre transgenic mouse model to constitutively activate Notch1 signaling in endothelial cells of adult mice. The triple transgenic mice developed severe albuminuria with significantly decreased VE-cadherin (vascular endothelial cadherin) expression in the glomerular endothelium. In vitro studies showed that either NICD1 (Notch1 intracellular domain) lentiviral infection or treatment with Notch ligand DLL4 (delta-like ligand 4) markedly reduced VE-cadherin expression and increased monolayer permeability of human renal glomerular endothelial cells. In addition, Notch1 activation or gene knockdown of VE-cadherin reduced the glomerular endothelial glycocalyx. Further investigation demonstrated that activated Notch1 suppression of VE-cadherin was through the transcription factors SNAI1 (snail family transcriptional repressor 1) and ERG (Ets related gene), which bind to the -373 E-box and the -134/-118 ETS (E26 transformation-specific) element of the VE-cadherin promoter, respectively. CONCLUSIONS Our results reveal novel regulatory mechanisms whereby endothelial Notch1 signaling dictates the level of VE-cadherin through the transcription factors SNAI1 and ERG, leading to dysfunction of glomerular filtration barrier and induction of albuminuria. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Liqun Li
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Qiang Liu
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tongyao Shang
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Wei Song
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - Dongmei Xu
- Department of Nephrology (D.X.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Thaddeus D Allen
- Molecular and Cellular Biology Division, Sunnybrook Health Science Centre (T.D.A., J.J., C.G.L.), University of Toronto, Ontario, Canada.,Department of Medical Biophysics (T.D.A., C.G.L.), University of Toronto, Ontario, Canada.,Tradewind BioScience, Daly City, California (T.D.A.)
| | - Xia Wang
- School of Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China (L.L., T.S., W.S., X.W.)
| | - James Jeong
- General Internal Medicine, Markham Stouffville Hospital, Toronto, Ontario, Canada (J.J.)
| | - Corrinne G Lobe
- Molecular and Cellular Biology Division, Sunnybrook Health Science Centre (T.D.A., J.J., C.G.L.), University of Toronto, Ontario, Canada.,Department of Medical Biophysics (T.D.A., C.G.L.), University of Toronto, Ontario, Canada
| | - Ju Liu
- Institute of Microvascular Medicine, Medical Research Center (L.L., Q.L., J.L.), Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
5
|
Dong F, Zhao X, Wang J, Huang X, Li X, Zhang L, Dong H, Liu F, Fan M. Dihydroartemisinin inhibits the expression of von Willebrand factor by downregulation of transcription factor ERG in endothelial cells. Fundam Clin Pharmacol 2020; 35:321-330. [PMID: 33107067 PMCID: PMC7983977 DOI: 10.1111/fcp.12622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/18/2022]
Abstract
Dihydroartemisinin (DHA), a semi‐synthetic derivative of artemisinin, has effective antitumor and anti‐inflammatory actions. von Willebrand factor (vWF), a large multifunctional glycoprotein, has a prominent function in hemostasis and is a key factor in thrombus formation. In addition, vWF has been regarded as a prospective biomarker for the diagnosis of endothelial dysfunction. In our experiment, we observed that 25 μM DHA specifically downregulated the expression of vWF mRNA and protein in human umbilical vein endothelial cells (HUVECs). Further investigations demonstrated that this DHA‐decreased vWF expression was mediated by the transcription factor ERG and not GATA3. Luciferase activity assay confirmed that DHA regulated the ERG binding with the −56 ETS‐binding motif on the human vWF promoter. Thus, the −56 ETS motif on the vWF promoter region regulates the expression of vWF gene which is induced by DHA. Taken together, we proved that DHA decreased the vWF transcription through the downregulation of ERG in HUVECs. As vWF plays a key role in vascular homeostasis, our findings suggest a new role of DHA in vascular diseases.
Collapse
Affiliation(s)
- Fengyun Dong
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, China
| | - Xinghai Zhao
- Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Jianning Wang
- Department of Urology, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xin Huang
- Department of Clinical Pharmacy, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Xiao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 16369 Jingshi Road, Jinan, Shandong, 250011, China
| | - Liang Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, 88 Wenhuadong Street, Jinan, Shandong, 250014, China
| | - Haixin Dong
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong, 272029, China
| | - Fuhong Liu
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China
| | - Mengge Fan
- Laboratory of Microvascular Medicine, Medical Research Center, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, 16766 Jingshi Road, Jinan, Shandong, 250014, China.,Graduate School, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, 250000, China
| |
Collapse
|
6
|
Roy B, Palaniyandi SS. Aldehyde dehydrogenase 2 inhibition potentiates 4-hydroxy-2-nonenal induced decrease in angiogenesis of coronary endothelial cells. Cell Biochem Funct 2020; 38:290-299. [PMID: 31943249 DOI: 10.1002/cbf.3468] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/15/2019] [Accepted: 11/11/2019] [Indexed: 11/12/2022]
Abstract
Coronary endothelial cell (EC) dysfunction including defective angiogenesis is reported in cardiac diseases. 4-Hydroxynonenal (4HNE) is a lipid peroxidation product, which is increased in cardiac diseases and implicated in cellular toxicity. Aldehyde dehydrogenase (ALDH) 2 is a mitochondrial enzyme that metabolizes 4HNE and reduces 4HNE-mediated cytotoxicity. Thus, we hypothesize that ALDH2 inhibition potentiates 4HNE-mediated decrease in coronary EC angiogenesis in vitro. To test our hypothesis, first, we treated the cultured mouse coronary EC (MCEC) lines with 4HNE (25, 50, and 75 μM) for 2 and 4 hours. Next, we pharmacologically inhibited ALDH2 by disulfiram (DSF) (2.5 μM) before challenging the cells with 4HNE. In this study, we found that 4HNE attenuated tube formation which indicates decreased angiogenesis. Next, we found that 4HNE has significantly downregulated the expressions of vascular endothelial growth factor receptor (VEGFR) 2 (P < .05 for mRNA and P = .005 for protein), Sirtuin 1 (SIRT 1) (P < 0.0005 for mRNA), and Ets-related gene (ERG) (P < 0.0001 for mRNA and P < 0.005 for protein) in MCECs compared with control. ALDH 2 inhibition by DSF potentiated 4HNE-induced decrease in angiogenesis (P < 0.05 vs 4HNE at 2 h and P < 0.0005 vs 4HNE at 4 h) by decreasing the expressions of VEGFR2 (P < 0.005 for both mRNA and protein), SIRT 1 (P < 0.05), and ERG (P < 0.005) relative to 4HNE alone. Thus, we conclude that ALDH2 acts as a proangiogenic signaling molecule by alleviating the antiangiogenic effects of 4HNE in MCECs.
Collapse
Affiliation(s)
- Bipradas Roy
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University, Detroit, Michigan
| | - Suresh Selvaraj Palaniyandi
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan.,Department of Physiology, Wayne State University, Detroit, Michigan
| |
Collapse
|
7
|
Dong H, Zhou Y, Wang Y, Zhou Q, Zhang Y, Gan X, Luo Y, Li R. The protective role of intermedin in promoting angiogenesis during renal fibrosis. Gene 2019; 688:34-43. [DOI: 10.1016/j.gene.2018.11.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/09/2018] [Accepted: 11/16/2018] [Indexed: 01/12/2023]
|
8
|
Nagai N, Ohguchi H, Nakaki R, Matsumura Y, Kanki Y, Sakai J, Aburatani H, Minami T. Downregulation of ERG and FLI1 expression in endothelial cells triggers endothelial-to-mesenchymal transition. PLoS Genet 2018; 14:e1007826. [PMID: 30500808 PMCID: PMC6291168 DOI: 10.1371/journal.pgen.1007826] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 12/12/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022] Open
Abstract
Endothelial cell (EC) plasticity in pathological settings has recently been recognized as a driver of disease progression. Endothelial-to-mesenchymal transition (EndMT), in which ECs acquire mesenchymal properties, has been described for a wide range of pathologies, including cancer. However, the mechanism regulating EndMT in the tumor microenvironment and the contribution of EndMT in tumor progression are not fully understood. Here, we found that combined knockdown of two ETS family transcription factors, ERG and FLI1, induces EndMT coupled with dynamic epigenetic changes in ECs. Genome-wide analyses revealed that ERG and FLI1 are critical transcriptional activators for EC-specific genes, among which microRNA-126 partially contributes to blocking the induction of EndMT. Moreover, we demonstrated that ERG and FLI1 expression is downregulated in ECs within tumors by soluble factors enriched in the tumor microenvironment. These data provide new insight into the mechanism of EndMT, functions of ERG and FLI1 in ECs, and EC behavior in pathological conditions. Differentiated cells possess unique characteristics to maintain vital activities. However, cells occasionally show abnormal behavior in pathological settings due to dysregulated gene expression. Endothelial-to-mesenchymal transition (EndMT) is a phenomenon in which endothelial cells lose their characteristics and acquire mesenchymal-like properties. Although EndMT is observed in various diseases including cancer, and augments fibrosis and vascular defects, the mechanism of EndMT induction is not fully understood. Here, we show that EndMT is triggered via reduced expression of ERG and FLI1, which have recently been recognized as pivotal transcription factors in endothelial cells (ECs). Mechanistically, ERG and FLI1 activate EC-specific genes and repress mesenchymal-like genes via epigenetic regulation to prevent EndMT. Furthermore, we demonstrate that microRNA-126, which is specifically expressed in ECs, is the key downstream target of ERG/FLI1 for regulating EndMT. Finally, we show that ERG and FLI1 expression is decreased in ECs within tumors, suggesting that EndMT is induced in the tumor microenvironment. Collectively, these findings indicate that loss of ERG and FLI1 leads to the aberrant behavior of ECs in pathological conditions.
Collapse
Affiliation(s)
- Nao Nagai
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto, Japan
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroto Ohguchi
- Division of Disease Epigenetics, IRDA, Kumamoto University, Kumamoto, Japan
| | - Ryo Nakaki
- Division of Genome Sciences, RCAST, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Matsumura
- Division of Metabolic Medicine, RCAST, The University of Tokyo, Tokyo, Japan
| | - Yasuharu Kanki
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Juro Sakai
- Division of Metabolic Medicine, RCAST, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Division of Genome Sciences, RCAST, The University of Tokyo, Tokyo, Japan
| | - Takashi Minami
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto, Japan
- * E-mail:
| |
Collapse
|
9
|
Fish JE, Cantu Gutierrez M, Dang LT, Khyzha N, Chen Z, Veitch S, Cheng HS, Khor M, Antounians L, Njock MS, Boudreau E, Herman AM, Rhyner AM, Ruiz OE, Eisenhoffer GT, Medina-Rivera A, Wilson MD, Wythe JD. Dynamic regulation of VEGF-inducible genes by an ERK/ERG/p300 transcriptional network. Development 2017; 144:2428-2444. [PMID: 28536097 DOI: 10.1242/dev.146050] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
The transcriptional pathways activated downstream of vascular endothelial growth factor (VEGF) signaling during angiogenesis remain incompletely characterized. By assessing the signals responsible for induction of the Notch ligand delta-like 4 (DLL4) in endothelial cells, we find that activation of the MAPK/ERK pathway mirrors the rapid and dynamic induction of DLL4 transcription and that this pathway is required for DLL4 expression. Furthermore, VEGF/ERK signaling induces phosphorylation and activation of the ETS transcription factor ERG, a prerequisite for DLL4 induction. Transcription of DLL4 coincides with dynamic ERG-dependent recruitment of the transcriptional co-activator p300. Genome-wide gene expression profiling identified a network of VEGF-responsive and ERG-dependent genes, and ERG chromatin immunoprecipitation (ChIP)-seq revealed the presence of conserved ERG-bound putative enhancer elements near these target genes. Functional experiments performed in vitro and in vivo confirm that this network of genes requires ERK, ERG and p300 activity. Finally, genome-editing and transgenic approaches demonstrate that a highly conserved ERG-bound enhancer located upstream of HLX (which encodes a transcription factor implicated in sprouting angiogenesis) is required for its VEGF-mediated induction. Collectively, these findings elucidate a novel transcriptional pathway contributing to VEGF-dependent angiogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Manuel Cantu Gutierrez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan T Dang
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Nadiya Khyzha
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Zhiqi Chen
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Shawn Veitch
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Henry S Cheng
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Melvin Khor
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Lina Antounians
- Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Makon-Sébastien Njock
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Emilie Boudreau
- Toronto General Hospital Research Institute, University Health Network, Toronto M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada.,Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada
| | - Alexander M Herman
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander M Rhyner
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Oscar E Ruiz
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George T Eisenhoffer
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Graduate School of Biomedical Sciences, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alejandra Medina-Rivera
- Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Querétaro 76230, México
| | - Michael D Wilson
- Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto M5S 3H2, Canada.,Genetics and Genome Biology, Hospital for Sick Children, Toronto M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Joshua D Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA .,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Shah AV, Birdsey GM, Randi AM. Regulation of endothelial homeostasis, vascular development and angiogenesis by the transcription factor ERG. Vascul Pharmacol 2016; 86:3-13. [PMID: 27208692 PMCID: PMC5404112 DOI: 10.1016/j.vph.2016.05.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/08/2016] [Accepted: 05/16/2016] [Indexed: 01/06/2023]
Abstract
Over the last few years, the ETS transcription factor ERG has emerged as a major regulator of endothelial function. Multiple studies have shown that ERG plays a crucial role in promoting angiogenesis and vascular stability during development and after birth. In the mature vasculature ERG also functions to maintain endothelial homeostasis, by transactivating genes involved in key endothelial functions, while repressing expression of pro-inflammatory genes. Its homeostatic role is lineage-specific, since ectopic expression of ERG in non-endothelial tissues such as prostate is detrimental and contributes to oncogenesis. This review summarises the main roles and pathways controlled by ERG in the vascular endothelium, its transcriptional targets and its functional partners and the emerging evidence on the pathways regulating ERG's activity and expression.
Collapse
Affiliation(s)
- Aarti V Shah
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Graeme M Birdsey
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anna M Randi
- Vascular Sciences, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
11
|
Characterization of the 5'-flanking region of the human DNA helicase B (HELB) gene and its response to trans-Resveratrol. Sci Rep 2016; 6:24510. [PMID: 27079536 PMCID: PMC4832242 DOI: 10.1038/srep24510] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/30/2016] [Indexed: 12/17/2022] Open
Abstract
Human DNA helicase B (HELB/HDHB) regulates DNA replication through association with human DNA polymerase α-primase. In the present study, an 866-base pair (bp) of the 5′-flanking region of the human HELB gene-containing Luciferase (Luc) reporter plasmid, pHDHB-Luc was transfected into various cell lines and Luc activity was analyzed. Deletion analyses revealed that a 121-bp containing the major transcription start site (TSS) was essential for the basal promoter activity in all tested cells. TF-SEARCH analysis indicated that GC-box/Sp1 and duplicated GGAA-motifs containing putative STAT-x and c-ETS binding sites are located close to the TSS. Furthermore, chromatin immunoprecipitation (ChIP) analysis showed that PU.1 and Sp1 bind to the 121-bp region. Reverse transcriptase-polymerase chain reaction (RT-PCR) and western blot analyses showed the HELB gene and protein expression was up-regulated by trans-Resveratrol (Rsv) treatment in HeLa S3 cells. Moreover, transfection experiment indicated that mutations on the GC-boxes and the duplicated GGAA-motif greatly reduced promoter activity and the response to Rsv in HeLa S3 cells. These results suggest that Rsv, which is a natural compound that has been found to elongate the lifespan of various organisms, regulates HELB promoter activity through co-operation of the GC-boxes and the duplicated GGAA-motif in the 121-bp.
Collapse
|
12
|
Development of Endothelial-Specific Single Inducible Lentiviral Vectors for Genetic Engineering of Endothelial Progenitor Cells. Sci Rep 2015; 5:17166. [PMID: 26612671 PMCID: PMC4661691 DOI: 10.1038/srep17166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/22/2015] [Indexed: 12/02/2022] Open
Abstract
Endothelial progenitor cells (EPC) are able to migrate to tumor vasculature. These cells, if genetically modified, can be used as vehicles to deliver toxic material to, or express anticancer proteins in tumor. To test this hypothesis, we developed several single, endothelial-specific, and doxycycline-inducible self-inactivating (SIN) lentiviral vectors. Two distinct expression cassettes were inserted into a SIN-vector: one controlled by an endothelial lineage-specific, murine vascular endothelial cadherin (mVEcad) promoter for the expression of a transactivator, rtTA2S-M2; and the other driven by an inducible promoter, TREalb, for a firefly luciferase reporter gene. We compared the expression levels of luciferase in different vector constructs, containing either the same or opposite orientation with respect to the vector sequence. The results showed that the vector with these two expression cassettes placed in opposite directions was optimal, characterized by a robust induction of the transgene expression (17.7- to 73-fold) in the presence of doxycycline in several endothelial cell lines, but without leakiness when uninduced. In conclusion, an endothelial lineage-specific single inducible SIN lentiviral vector has been developed. Such a lentiviral vector can be used to endow endothelial progenitor cells with anti-tumor properties.
Collapse
|
13
|
Birdsey GM, Shah AV, Dufton N, Reynolds LE, Osuna Almagro L, Yang Y, Aspalter IM, Khan ST, Mason JC, Dejana E, Göttgens B, Hodivala-Dilke K, Gerhardt H, Adams RH, Randi AM. The endothelial transcription factor ERG promotes vascular stability and growth through Wnt/β-catenin signaling. Dev Cell 2015; 32:82-96. [PMID: 25584796 PMCID: PMC4292982 DOI: 10.1016/j.devcel.2014.11.016] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/24/2014] [Accepted: 11/10/2014] [Indexed: 12/14/2022]
Abstract
Blood vessel stability is essential for embryonic development; in the adult, many diseases are associated with loss of vascular integrity. The ETS transcription factor ERG drives expression of VE-cadherin and controls junctional integrity. We show that constitutive endothelial deletion of ERG (Erg(cEC-KO)) in mice causes embryonic lethality with vascular defects. Inducible endothelial deletion of ERG (Erg(iEC-KO)) results in defective physiological and pathological angiogenesis in the postnatal retina and tumors, with decreased vascular stability. ERG controls the Wnt/β-catenin pathway by promoting β-catenin stability, through signals mediated by VE-cadherin and the Wnt receptor Frizzled-4. Wnt signaling is decreased in ERG-deficient endothelial cells; activation of Wnt signaling with lithium chloride, which stabilizes β-catenin levels, corrects vascular defects in Erg(cEC-KO) embryos. Finally, overexpression of ERG in vivo reduces permeability and increases stability of VEGF-induced blood vessels. These data demonstrate that ERG is an essential regulator of angiogenesis and vascular stability through Wnt signaling.
Collapse
Affiliation(s)
- Graeme M Birdsey
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Aarti V Shah
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Neil Dufton
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Louise E Reynolds
- Centre for Tumour Biology, Barts Cancer Institute - a CR-UK Centre of Excellence, John Vane Science Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Lourdes Osuna Almagro
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Youwen Yang
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Irene M Aspalter
- Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3PX, UK
| | - Samia T Khan
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Justin C Mason
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Elisabetta Dejana
- FIRC Institute of Molecular Oncology Foundation, IFOM, 20139 Milan, Italy
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
| | - Kairbaan Hodivala-Dilke
- Centre for Tumour Biology, Barts Cancer Institute - a CR-UK Centre of Excellence, John Vane Science Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Holger Gerhardt
- Vascular Biology Laboratory, London Research Institute, Cancer Research UK, London WC2A 3PX, UK
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Anna M Randi
- National Heart and Lung Institute (NHLI) Vascular Sciences, Hammersmith Hospital, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
14
|
Transcriptional activation of OCT4 by the ETS transcription factor PEA3 in NCCIT human embryonic carcinoma cells. FEBS Lett 2014; 588:3129-36. [PMID: 24983502 DOI: 10.1016/j.febslet.2014.06.052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022]
Abstract
We examined the molecular mechanism of OCT4 gene regulation by polyomavirus enhancer activator 3 (PEA3) in NCCIT cells. Endogenous PEA3 and OCT4 were significantly elevated in undifferentiated cells and reduced upon differentiation. PEA3 knockdown led to a reduction in OCT4 levels. OCT4 promoter activity was significantly up-regulated by dose-dependent PEA3 overexpression. Deletion and site-directed mutagenesis of the OCT4 promoter revealed a putative binding site within the conserved region 2 (CR2). PEA3 interacted with the binding element within CR2 in NCCIT cells. This study reveals the molecular details of the mechanism by which the oncogenic factor PEA3 regulates OCT4 gene expression as a transcriptional activator.
Collapse
|
15
|
Chen T, Wang J, Xue B, Kong Q, Liu Z, Yu B. Identification and characterization of a novel porcine endothelial cell-specific Tie1 promoter. Xenotransplantation 2013; 20:438-48. [PMID: 24112087 DOI: 10.1111/xen.12059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/14/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND The use of a transgenic pig for xenotransplantation and as a cardiovascular disease model has caught much attention in the past decades. The vascular endothelial cell is the primary modification target for the application of genetically modified pigs in this field. However, the powerful porcine endothelial cell-specific promoter is still so rare that the mouse and human promoters are commonly used. In the study, the porcine Tie1 (sTie1) promoter was identified and characterized as a potential endothelial cell-specific promoter to generate a cardiovascular disease model. METHODS Tie1 promoters with different lengths of 5'-regulatory regions were cloned, and major putative DNA-binding motifs were mutated by site-directed mutagenesis. All fragments were ligated into the luciferase reporter system and were transiently transfected into endothelial cells to identify luciferase activity using a dual luciferase reporter assay. RESULTS The luciferase activities of sTie1 promoters with different lengths of the 5'-regulatory region were tested. Results showed that the luciferase activity of the 1234-bp sTie1 fragment was the strongest compared with that of others (P < 0.001). Site-directed mutagenesis in transcription-factor-binding sites, including Ets, GATA, and AP2, verified their key roles in regulating transcription, especially sites Ets (-103), GATA (-211), and AP2 (-3). The activities of Tie1 promoters from pig, human, and mouse were significantly different in pig iliac endothelial cells (PIECs) (P < 0.001), and the sTie1 promoter showed the highest activity. Moreover, sTie1 promoter activity could be detected in porcine embryo fibroblasts and skeletal muscle cells. CONCLUSIONS The sTie1 promoter shows a highly conserved sequence compared with the Tie1 promoters in human and mouse, but it has a greater activity in the porcine endothelial cell line than that of human and mouse promoters. Thus, sTie1 will be a valuable tool for generating a pig cardiovascular disease model.
Collapse
Affiliation(s)
- Tao Chen
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, Heilongjiang, China; Cardiology Division, the Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | | | | | | | | | | |
Collapse
|
16
|
Cheng JC, Chang HM, Leung PCK. Transforming growth factor-β1 inhibits trophoblast cell invasion by inducing Snail-mediated down-regulation of vascular endothelial-cadherin protein. J Biol Chem 2013; 288:33181-92. [PMID: 24106276 DOI: 10.1074/jbc.m113.488866] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human trophoblast cells express transforming growth factor-β (TGF-β) and TGF-β receptors. It has been shown that TGF-β1 treatment decreases the invasiveness of trophoblast cells. However, the molecular mechanisms underlying TGF-β1-decreased trophoblast invasion are still not fully understood. In the current study, we demonstrated that treatment of HTR-8/SVneo human trophoblast cells with TGF-β1 decreased cell invasion and down-regulated the expression of vascular endothelial cadherin (VE-cadherin). In addition, the inhibitory effect of TGF-β1 on VE-cadherin was confirmed in primary cultures of human trophoblast cells. Moreover, knockdown of VE-cadherin using siRNA decreased the invasiveness of HTR-8/SVneo cells and primary cultures of trophoblast cells. Treatment with TGF-β1 induced the activation of Smad-dependent signaling pathways and the expression of Snail and Slug. Knockdown of Smads attenuated TGF-β1-induced up-regulation of Snail and Slug and down-regulation of VE-cadherin. Interestingly, depletion of Snail, but not Slug, attenuated TGF-β1-induced down-regulation of VE-cadherin. Furthermore, overexpression of Snail suppressed VE-cadherin expression. Chromatin immunoprecipitation analyses showed the direct binding of Snail to the VE-cadherin promoter. These results provide evidence that Snail mediates TGF-β1-induced down-regulation of VE-cadherin, which subsequently contributed to TGF-β1-decreased trophoblast cell invasion.
Collapse
Affiliation(s)
- Jung-Chien Cheng
- From the Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V6H 3V5, Canada
| | | | | |
Collapse
|
17
|
Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D, Ding BS, Schachterle W, Liu Y, Rosenwaks Z, Butler JM, Xiang J, Rafii A, Shido K, Rabbany SY, Elemento O, Rafii S. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell 2013; 26:204-19. [PMID: 23871589 DOI: 10.1016/j.devcel.2013.06.017] [Citation(s) in RCA: 474] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/01/2013] [Accepted: 06/18/2013] [Indexed: 02/08/2023]
Abstract
Microvascular endothelial cells (ECs) within different tissues are endowed with distinct but as yet unrecognized structural, phenotypic, and functional attributes. We devised EC purification, cultivation, profiling, and transplantation models that establish tissue-specific molecular libraries of ECs devoid of lymphatic ECs or parenchymal cells. These libraries identify attributes that confer ECs with their organotypic features. We show that clusters of transcription factors, angiocrine growth factors, adhesion molecules, and chemokines are expressed in unique combinations by ECs of each organ. Furthermore, ECs respond distinctly in tissue regeneration models, hepatectomy, and myeloablation. To test the data set, we developed a transplantation model that employs generic ECs differentiated from embryonic stem cells. Transplanted generic ECs engraft into regenerating tissues and acquire features of organotypic ECs. Collectively, we demonstrate the utility of informational databases of ECs toward uncovering the extravascular and intrinsic signals that define EC heterogeneity. These factors could be exploited therapeutically to engineer tissue-specific ECs for regeneration.
Collapse
Affiliation(s)
- Daniel J Nolan
- Department of Genetic Medicine, Howard Hughes Medical Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Neumann P, Alsaffar H, Gertzberg N, Johnson A. Inhibition of GSK3α/β promotes increased pulmonary endothelial permeability to albumin by reactive oxygen/nitrogen species. Pulm Pharmacol Ther 2013; 26:685-92. [PMID: 23770025 DOI: 10.1016/j.pupt.2013.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 04/26/2013] [Accepted: 06/02/2013] [Indexed: 12/31/2022]
Abstract
Glycogen synthase kinase 3α/β (GSK3α/β) is a serine/threonine kinase that participates in numerous processes in many cell types. Importantly, the role of GSK3α/β in homeostatic maintenance of the pulmonary endothelial cell barrier to protein is not known. We tested the hypothesis that GSK3α/β regulates endothelial barrier function by measuring the permeability to albumin of a rat pulmonary microvessel endothelial cell monolayer (PMECM) treated with and without the selective GSK3α/β inhibitor SB 216763 (1.0, 5.0 and 10 uM) for 1 h. The treatment with the inhibitor SB 216763 caused a dose dependent decrease in phospho-β-catenin-Ser(33/37) levels indicating effective suppression of GSK3α/β. SB216763 caused an increase in both permeability to albumin and DCFDA (6-Carboxy-2',7'-Dichlorodihydrofluorescein Diacetate, Di(Acetoxymethyl Ester)) oxidation that were prevented by co-treatment with the anti-oxidant tiron or the nitric oxide synthase inhibitor L-NAME (Nω-nitro-l-arginine-methyl ester). In separate studies PMECMs were treated with the Akt inhibitor triciribine (12.5 uM) for 1 h to unmask Akt dependent constitutive suppression of GSK3α/β. Triciribine decreased phospho-GSK3α/β-Ser(21)/9 (i.e., the product of Akt) which was associated with an increase in phospho-β-catenin-Ser(33/37) (i.e., the product of GSK3α/β) indicating constitutive activity of Akt for GSK3α/β-Ser(21/9). The data indicates GSK3α/β inhibition causes increased endothelial monolayer protein permeability which is mediated by reactive oxygen/nitrogen species.
Collapse
Affiliation(s)
- Paul Neumann
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | | | | | | |
Collapse
|
19
|
Liang J, Huang W, Yu X, Ashraf A, Wary KK, Xu M, Millard RW, Ashraf M, Wang Y. Suicide gene reveals the myocardial neovascularization role of mesenchymal stem cells overexpressing CXCR4 (MSC(CXCR4)). PLoS One 2012; 7:e46158. [PMID: 23029422 PMCID: PMC3460871 DOI: 10.1371/journal.pone.0046158] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Accepted: 08/28/2012] [Indexed: 01/06/2023] Open
Abstract
Background Our previous studies indicated that MSCCXCR4 improved cardiac function after myocardial infarction (MI). This study was aimed to investigate the specific role of MSCCXCR4 in neovascularization of infarcted myocardium using a suicide gene approach. Methods MSCs were transduced with either lentivirus-null vector/GFP (MSCNull as control) or vector encoding for overexpressing CXCR4/GFP. The MSC derived-endothelial cell (EC) differentiation was assessed by a tube formation assay, Dil-ac-LDL uptake, EC marker expression, and VE-cadherin promoter activity assay. Gene expression was analyzed by quantitative RT-PCR or Western blot. The suicide gene approach was under the control of VE-cadherin promoter. In vivo studies: Cell patches containing MSCNull or MSCCXCR4 were transduced with suicide gene and implanted into the myocardium of MI rat. Rats received either ganciclovir (GCV) or vehicle after cell implantation. After one month, the cardiac functional changes and neovascularization were assessed by echocardiography, histological analysis, and micro-CT imaging. Results The expression of VEGF-A and HIF-1α was significantly higher in MSCCXCR4 as compared to MSCNull under hypoxia. Additionally, MSCCXCR4 enhanced new vessel formation and EC differentiation, as well as STAT3 phosphorylation under hypoxia. STAT3 participated in the transcription of VE-cadherin in MSCCXCR4 under hypoxia, which was inhibited by WP1066 (a STAT3 inhibitor). In addition, GCV specifically induced death of ECs with suicide gene activation. In vivo studies: MSCCXCR4 implantation promoted cardiac functional restoration, reduced infarct size, improved cardiac remodeling, and enhanced neovascularization in ischemic heart tissue. New vessels derived from MSCCXCR4 were observed at the injured heart margins and communicated with native coronary arteries. However, the derived vessel networks were reduced by GCV, reversing improvement of cardiac function. Conclusion The transplanted MSCCXCR4 enhanced neovascularization after MI by boosting release of angiogenic factors and increasing the potential of endothelial differentiation.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Endothelial Cells/cytology
- Endothelial Cells/metabolism
- Gene Expression
- Genes, Transgenic, Suicide
- Genetic Vectors
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lentivirus/genetics
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/metabolism
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardium/metabolism
- Neovascularization, Physiologic
- Phosphorylation
- Rats
- Rats, Sprague-Dawley
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Transduction, Genetic
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology, College of Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Arderiu G, Peña E, Aledo R, Espinosa S, Badimon L. Ets-1 transcription is required in tissue factor driven microvessel formation and stabilization. Angiogenesis 2012; 15:657-69. [PMID: 22869003 DOI: 10.1007/s10456-012-9293-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 07/28/2012] [Indexed: 12/19/2022]
Abstract
Tissue factor (TF) has well-recognized roles as initiator of blood coagulation as well as an intracellular signaling receptor. TF signaling regulates gene transcription and protein translation. Recently, we have shown that TF-induced mature neovessel formation is ultimately driven by CCL2 expression. However, the signaling process induced by TF to promote microvessel formation remains to be determined. This study was designed with the objective to investigate the mechanisms involved in TF-induced neovessel formation. Here, we have identified that Ets-1 expression is a downstream effector of TF signaling. TF-siRNA induced a highly significant reduction in Ets-1 expression levels and in Ets-1/DNA binding while inducing abrogation of microvessel formation. Activation of Ets-1 rescued the effect of TF inhibition and restored microvessel formation confirming the critical role of Ets-1 in TF-induced angiogenesis. VE-cadherin expression, a key regulator of endothelial intercellular junctions, and an Ets-1 target molecule was dependent of TF-inhibition. We show that TF signals through ERK1/2 to activate Ets-1 and induce CCL2 gene expression by binding to its promoter region. We conclude that endothelial cell TF signals through ERK1/2 and Ets-1 to trigger microvessel formation.
Collapse
Affiliation(s)
- Gemma Arderiu
- Cardiovascular Research Center (CSIC-ICCC), Hospital de Sant Pau (UAB), IIB-Sant Pau. CiberOBN, Instituto de Salut Carlos III, C/ Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | | | | | | | | |
Collapse
|
21
|
Yuan L, Le Bras A, Sacharidou A, Itagaki K, Zhan Y, Kondo M, Carman CV, Davis GE, Aird WC, Oettgen P. ETS-related gene (ERG) controls endothelial cell permeability via transcriptional regulation of the claudin 5 (CLDN5) gene. J Biol Chem 2012; 287:6582-91. [PMID: 22235125 DOI: 10.1074/jbc.m111.300236] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
ETS-related gene (ERG) is a member of the ETS transcription factor family. Our previous studies have shown that ERG expression is highly enriched in endothelial cells (EC) both in vitro and in vivo. ERG expression is markedly repressed in response to inflammatory stimuli. It has been shown that ERG is a positive regulator of several EC-restricted genes including VE-cadherin, endoglin, and von Willebrand factor, and a negative regulator of other genes such as interleukin (IL)-8 and intercellular adhesion molecule (ICAM)-1. In this study we have identified a novel role for ERG in the regulation of EC barrier function. ERG knockdown results in marked increases in EC permeability. This is associated with a significant increase of stress fiber and gap formation in EC. Furthermore, we identify CLDN5 as a downstream target of ERG in EC. Thus, our results suggest that ERG plays a pivotal role in regulating EC barrier function and that this effect is mediated in part through its regulation of CLDN5 gene expression.
Collapse
Affiliation(s)
- Lei Yuan
- Division of Cardiology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Schachterle W, Rojas A, Xu SM, Black BL. ETS-dependent regulation of a distal Gata4 cardiac enhancer. Dev Biol 2011; 361:439-49. [PMID: 22056786 DOI: 10.1016/j.ydbio.2011.10.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 09/30/2011] [Accepted: 10/08/2011] [Indexed: 01/10/2023]
Abstract
The developing heart contains an inner tube of specialized endothelium known as endocardium, which performs multiple essential functions. In spite of the essential role of the endocardium in heart development and function, the transcriptional pathways that regulate its development remain largely undefined. GATA4 is a zinc finger transcription factor that is expressed in multiple cardiovascular lineages and is required for endocardial cushion development and embryonic viability, but the transcriptional pathways upstream of Gata4 in the endocardium and its derivatives in the endocardial cushions are unknown. Here, we describe a distal enhancer from the mouse Gata4 gene that is briefly active in multiple cardiac lineages early in cardiac development but restricts to the endocardium where it remains active through cardiogenesis. The activity of this Gata4 cardiac enhancer in transgenic embryos and in cultured aortic endothelial cells is dependent on four ETS sites. To identify which ETS transcription factors might be involved in Gata4 regulation via the ETS sites in the enhancer, we determined the expression profile of 24 distinct ETS factors in embryonic mouse hearts. Among multiple ETS transcripts present, ETS1, FLI1, ETV1, ETV5, ERG, and ETV6 were the most abundant in the early embryonic heart. We found that ETS1, FLI1, and ERG were strongly expressed in the heart at embryonic day 8.5 and that ETS1 and ERG bound to the endogenous Gata4 enhancer in cultured endothelial cells. Thus, these studies define the ETS expression profile in the early embryonic heart and identify an ETS-dependent enhancer from the Gata4 locus.
Collapse
Affiliation(s)
- William Schachterle
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158-2517, USA
| | | | | | | |
Collapse
|
23
|
Meadows SM, Myers CT, Krieg PA. Regulation of endothelial cell development by ETS transcription factors. Semin Cell Dev Biol 2011; 22:976-84. [PMID: 21945894 DOI: 10.1016/j.semcdb.2011.09.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 09/14/2011] [Indexed: 10/17/2022]
Abstract
The ETS family of transcription factors plays an essential role in controlling endothelial gene expression. Multiple members of the ETS family are expressed in the developing endothelium and evidence suggests that the proteins function, to some extent, redundantly. However, recent studies have demonstrated a crucial non-redundant role for ETV2, as a primary player in specification and differentiation of the endothelial lineage. Here, we review the contribution of ETS factors, and their partner proteins, to the regulation of embryonic vascular development.
Collapse
Affiliation(s)
- Stryder M Meadows
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, United States
| | | | | |
Collapse
|
24
|
The possible functions of duplicated ets (GGAA) motifs located near transcription start sites of various human genes. Cell Mol Life Sci 2011; 68:2039-51. [PMID: 21461879 PMCID: PMC3101357 DOI: 10.1007/s00018-011-0674-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/28/2011] [Accepted: 03/17/2011] [Indexed: 12/19/2022]
Abstract
Transcription is one of the most fundamental nuclear functions and is an enzyme complex-mediated reaction that converts DNA sequences into mRNA. Analyzing DNA sequences of 5′-flanking regions of several human genes that respond to 12-O-tetradecanoyl-phorbol-13-acetate (TPA) in HL-60 cells, we have identified that the ets (GGAA) motifs are duplicated, overlapped, or clustered within a 500-bp distance from the most 5′-upstream region of the cDNA. Multiple protein factors including Ets family proteins are known to recognize and bind to the GGAA containing sequences. In addition, it has been reported that the ets motifs play important roles in regulation of various promoters. Here, we propose a molecular mechanism, defined by the presence of duplication and multiplication of the GGAA motifs, that is responsible for the initiation of transcription of several genes and for the recruitment of binding proteins to the transcription start site (TSS) of TATA-less promoters.
Collapse
|
25
|
Cai J, Wu L, Qi X, Shaw L, Li Calzi S, Caballero S, Jiang WG, Vinores SA, Antonetti D, Ahmed A, Grant MB, Boulton ME. Placenta growth factor-1 exerts time-dependent stabilization of adherens junctions following VEGF-induced vascular permeability. PLoS One 2011; 6:e18076. [PMID: 21464949 PMCID: PMC3064593 DOI: 10.1371/journal.pone.0018076] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 02/24/2011] [Indexed: 01/13/2023] Open
Abstract
Increased vascular permeability is an early event characteristic of tissue ischemia and angiogenesis. Although VEGF family members are potent promoters of endothelial permeability the role of placental growth factor (PlGF) is hotly debated. Here we investigated PlGF isoforms 1 and 2 and present in vitro and in vivo evidence that PlGF-1, but not PlGF-2, can inhibit VEGF-induced permeability but only during a critical window post-VEGF exposure. PlGF-1 promotes VE-cadherin expression via the trans-activating Sp1 and Sp3 interaction with the VE-cadherin promoter and subsequently stabilizes transendothelial junctions, but only after activation of endothelial cells by VEGF. PlGF-1 regulates vascular permeability associated with the rapid localization of VE-cadherin to the plasma membrane and dephosphorylation of tyrosine residues that precedes changes observed in claudin 5 tyrosine phosphorylation and membrane localization. The critical window during which PlGF-1 exerts its effect on VEGF-induced permeability highlights the importance of the translational significance of this work in that PLGF-1 likely serves as an endogenous anti-permeability factor whose effectiveness is limited to a precise time point following vascular injury. Clinical approaches that would pattern nature's approach would thus limit treatments to precise intervals following injury and bring attention to use of agents only during therapeutic windows.
Collapse
Affiliation(s)
- Jun Cai
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Lin Wu
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Xiaoping Qi
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
| | - Lynn Shaw
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Sergio Li Calzi
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Sergio Caballero
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Wen G. Jiang
- Department of Surgery, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Stanley A. Vinores
- Ophthalmology, Johns Hopkins University, Wilmer Eye Institute, Baltimore, Maryland, United States of America
| | - David Antonetti
- Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Asif Ahmed
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Maria B. Grant
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Michael E. Boulton
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
26
|
Li Z, Xie J, Li W, Tang A, Li X, Jiang Z, Han Y, Ye J, Jing J, Gui Y, Cai Z. Identification and characterization of human PCDH10 gene promoter. Gene 2011; 475:49-56. [PMID: 21237250 DOI: 10.1016/j.gene.2011.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 12/30/2010] [Accepted: 01/03/2011] [Indexed: 01/16/2023]
Abstract
Recent studies have suggested roles for PCDH10 as a novel tumor suppressor gene. In our previous work, we located the core promoter of PCDH10 to a 462-bp segment of 5'-flanking region characterized by a high GC content. Here we further identified and characterized the promoter for PCDH10. Transient transfection of PC3 and LNCaP cells with a series of deleted promoter constructs indicated that the minimal promoter region was between nucleotides -144 and -99. This segment contained a CAAT box, a GT box, and a putative transcription factor binding site for AP-4. Mutational analysis identified that the CAAT box and GT box are necessary for promoter activity. Ectopic expression of NF-Ys increased reporter gene activity, whereas expression of a dominant-negative NF-YA decreased reporter gene activity. Co-transfection of Sp1/Sp3 expression plasmids enhanced reporter gene activity in a dose-dependent manner. Mithramycin A, an inhibitor of Sp-DNA interaction, reduced PCDH10 promoter activity. Electrophoretic mobility shift assays and chromatin immunoprecipitation demonstrated binding of transcription factors Sp1/Sp3 to the promoter region in vitro and in vivo. Our data show that Sp1/Sp3 and CBF/NF-Y transcription factors play a crucial role in the basal expression of the human PCDH10 gene.
Collapse
Affiliation(s)
- Zesong Li
- Guangdong Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, 1120 Lianhua Road, Shenzhen, Guangdong, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Vascular bed-specific regulation of the von Willebrand factor promoter in the heart and skeletal muscle. Blood 2010; 117:342-51. [PMID: 20980682 DOI: 10.1182/blood-2010-06-287987] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A region of the human von Willebrand factor (VWF) gene between -2812 and the end of the first intron (termed vWF2) was previously shown to direct expression in the endothelium of capillaries and a subset of larger blood vessels in the heart and skeletal muscle. Here, our goal was to delineate the DNA sequences responsible for this effect. A series of constructs containing deletions or mutations of vWF2 coupled to LacZ were targeted to the Hprt locus of mice, and the resulting animals were analyzed for reporter gene expression. The findings demonstrate that DNA sequences between -843 and -620 are necessary for expression in capillary but not large vessel endothelium in heart and skeletal muscle. Further, expression of VWF in capillaries and larger vessels of both tissues required the presence of a native or heterologous intron. In vitro assays implicated a role for ERG-binding ETS motif at -56 in mediating basal expression of VWF. In Hprt-targeted mice, mutation of the ETS consensus motif resulted in loss of LacZ expression in the endothelium of the heart and skeletal muscle. Together, these data indicate that distinct DNA modules regulate vascular bed-specific expression of VWF.
Collapse
|
28
|
Mohamed AA, Tan SH, Mikhalkevich N, Ponniah S, Vasioukhin V, Bieberich CJ, Sesterhenn IA, Dobi A, Srivastava S, Sreenath TL. Ets family protein, erg expression in developing and adult mouse tissues by a highly specific monoclonal antibody. J Cancer 2010; 1:197-208. [PMID: 21060730 PMCID: PMC2974237 DOI: 10.7150/jca.1.197] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 10/24/2010] [Indexed: 12/03/2022] Open
Abstract
Oncogenic activation of the ETS Related Gene (ERG) in humans was originally identified in subsets of Ewing sarcomas, myeloid leukemias and, recently, in the majority of prostate cancers. Expression of human ERG protein and consequently its functions in normal and disease states needs to be better understood in light of its suggested role in cell differentiation and proliferation. Here, we analyzed temporal and spatial expression of the Erg (mouse protein) by immunohistochemical analysis during mouse embryonic and adult organogenesis using a highly specific ERG monoclonal antibody (ERG MAb). This study establishes widespread immunolocalization of Erg protein in endothelial cells and restricted expression in precartilage and hematopoietic tissues. Intriguingly, Erg is not expressed in any epithelial tissue including prostate epithelium, or in infiltrating lymphocytes that are occasionally seen in the prostate environment, a common site of tumors with ERG rearrangements and unscheduled ERG expression. These findings will further aid in investigations of Erg functions in normal and disease conditions.
Collapse
Affiliation(s)
- Ahmed A Mohamed
- 1. Center For Prostate Disease Research, Department of Surgery, United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda MD 20814, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Salanga MC, Meadows SM, Myers CT, Krieg PA. ETS family protein ETV2 is required for initiation of the endothelial lineage but not the hematopoietic lineage in the Xenopus embryo. Dev Dyn 2010; 239:1178-87. [PMID: 20235229 DOI: 10.1002/dvdy.22277] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Transcription factors of the ETS family are important regulators of endothelial and hematopoietic development. We have characterized the Xenopus orthologue of the ETS transcription factor, ETV2. Expression analysis shows that etv2 is highly expressed in hematopoietic and endothelial precursor cells in the Xenopus embryo. In gain-of-function experiments, ETV2 is sufficient to activate ectopic expression of vascular endothelial markers. In addition, ETV2 activated expression of hematopoietic genes representing the myeloid but not the erythroid lineage. Loss-of-function studies indicate that ETV2 is required for expression of all endothelial markers examined. However, knockdown of ETV2 has no detectable effects on expression of either myeloid or erythroid markers. This contrasts with studies in mouse and zebrafish where ETV2 is required for development of the myeloid lineage. Our studies confirm an essential role for ETV2 in endothelial development, but also reveal important differences in hematopoietic development between organisms.
Collapse
Affiliation(s)
- Matthew C Salanga
- Department of Cell Biology and Anatomy, Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona 85724, USA
| | | | | | | |
Collapse
|
30
|
Le Bras A, Samson C, Trentini M, Caetano B, Lelievre E, Mattot V, Beermann F, Soncin F. VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS One 2010; 5:e12156. [PMID: 20808444 PMCID: PMC2922337 DOI: 10.1371/journal.pone.0012156] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 07/20/2010] [Indexed: 11/24/2022] Open
Abstract
Angiogenesis is the process by which new blood vessels arise from existing ones by the budding out of endothelial cell capillaries from the luminal side of blood vessels. Blood vessel formation is essential for organ development during embryogenesis and is associated with several physiological and pathological processes, such as wound healing and tumor development. The VE-statin/egfl7 gene is specifically expressed in endothelial cells during embryonic development and in the adult. We studied here the regulatory mechanisms that control this tissue-specific expression. RT-qPCR analyses showed that the specificity of expression of VE-statin/egfl7 in endothelial cells is not shared with its closest neighbor genes notch1 and agpat2 on the mouse chromosome 2. Chromatin-immunoprecipitation analysis of histone modifications at the VE-statin/egfl7 locus showed that the chromatin is specifically opened in endothelial cells, but not in fibroblasts at the transcription start sites. A 13 kb genomic fragment of promoter was cloned and analyzed by gene reporter assays which showed that two conserved regions are important for the specific expression of VE-statin/egfl7 in endothelial cells; a −8409/−7563 enhancer and the −252/+38 region encompassing the exon-1b transcription start site. The latter contains essential GATA and ETS-binding sites, as assessed by linker-scanning analysis and site-directed mutagenesis. An analysis of expression of the ETS and GATA transcription factors showed that Erg, Fli-1 and GATA-2 are the most highly expressed factors in endothelial cells. Erg and GATA-2 directly control the expression of the endogenous VE-statin/egfl7 while Fli-1 probably exerts an indirect control, as assessed by RNA interference and chromatin immunoprecipitation. This first detailed analysis of the mechanisms that govern the expression of the VE-statin/egfl7 gene in endothelial cells pinpoints the specific importance of ETS and GATA factors in the specific regulation of genes in this cell lineage.
Collapse
Affiliation(s)
- Alexandra Le Bras
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Chantal Samson
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Matteo Trentini
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Bertrand Caetano
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Etienne Lelievre
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Virginie Mattot
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Friedrich Beermann
- Swiss Institute for Experimental Cancer Research (ISREC), Centre de Phénotypage Génomique (CPG), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Fabrice Soncin
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
- * E-mail:
| |
Collapse
|
31
|
Harris ES, Nelson WJ. VE-cadherin: at the front, center, and sides of endothelial cell organization and function. Curr Opin Cell Biol 2010; 22:651-8. [PMID: 20708398 DOI: 10.1016/j.ceb.2010.07.006] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 07/13/2010] [Accepted: 07/14/2010] [Indexed: 11/19/2022]
Abstract
Endothelial cells form cell-cell adhesive structures, called adherens and tight junctions, which maintain tissue integrity, but must be dynamic for leukocyte transmigration during the inflammatory response and cellular remodeling during angiogenesis. This review will focus on Vascular Endothelial (VE)-cadherin, an endothelial-specific cell-cell adhesion protein of the adherens junction complex. VE-cadherin plays a key role in endothelial barrier function and angiogenesis, and consequently VE-cadherin availability and function are tightly regulated. VE-cadherin also participates directly and indirectly in intracellular signaling pathways that control cell dynamics and cell cycle progression. Here we highlight recent work that has advanced our understanding of multiple regulatory and signaling mechanisms that converge on VE-cadherin and have consequences for endothelial barrier function and angiogenic remodeling.
Collapse
Affiliation(s)
- Elizabeth S Harris
- Department of Biology, The James H. Clark Center, Bio-X Program, Stanford University, Stanford, CA 94305, USA.
| | | |
Collapse
|
32
|
Garrido-Martin EM, Blanco FJ, Fernandez-L A, Langa C, Vary CP, Lee UE, Friedman SL, Botella LM, Bernabeu C. Characterization of the human Activin-A receptor type II-like kinase 1 (ACVRL1) promoter and its regulation by Sp1. BMC Mol Biol 2010; 11:51. [PMID: 20587022 PMCID: PMC2906440 DOI: 10.1186/1471-2199-11-51] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 06/29/2010] [Indexed: 11/28/2022] Open
Abstract
Background Activin receptor-like kinase 1 (ALK1) is a Transforming Growth Factor-β (TGF-β) receptor type I, mainly expressed in endothelial cells that plays a pivotal role in vascular remodelling and angiogenesis. Mutations in the ALK1 gene (ACVRL1) give rise to Hereditary Haemorrhagic Telangiectasia, a dominant autosomal vascular dysplasia caused by a haploinsufficiency mechanism. In spite of its patho-physiological relevance, little is known about the transcriptional regulation of ACVRL1. Here, we have studied the different origins of ACVRL1 transcription, we have analyzed in silico its 5'-proximal promoter sequence and we have characterized the role of Sp1 in the transcriptional regulation of ACVRL1. Results We have performed a 5'Rapid Amplification of cDNA Ends (5'RACE) of ACVRL1 transcripts, finding two new transcriptional origins, upstream of the one previously described, that give rise to a new exon undiscovered to date. The 5'-proximal promoter region of ACVRL1 (-1,035/+210) was analyzed in silico, finding that it lacks TATA/CAAT boxes, but contains a remarkably high number of GC-rich Sp1 consensus sites. In cells lacking Sp1, ACVRL1 promoter reporters did not present any significant transcriptional activity, whereas increasing concentrations of Sp1 triggered a dose-dependent stimulation of its transcription. Moreover, silencing Sp1 in HEK293T cells resulted in a marked decrease of ACVRL1 transcriptional activity. Chromatin immunoprecipitation assays demonstrated multiple Sp1 binding sites along the proximal promoter region of ACVRL1 in endothelial cells. Furthermore, demethylation of CpG islands, led to an increase in ACVRL1 transcription, whereas in vitro hypermethylation resulted in the abolishment of Sp1-dependent transcriptional activation of ACVRL1. Conclusions Our results describe two new transcriptional start sites in ACVRL1 gene, and indicate that Sp1 is a key regulator of ACVRL1 transcription, providing new insights into the molecular mechanisms that contribute to the expression of ACVRL1 gene. Moreover, our data show that the methylation status of CpG islands markedly modulates the Sp1 regulation of ACVRL1 gene transcriptional activity.
Collapse
Affiliation(s)
- Eva M Garrido-Martin
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas and Centro de Investigación Biomédica en Red de Enfermedades Raras, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
The soluble fragment of VE-cadherin inhibits angiogenesis by reducing endothelial cell proliferation and tube capillary formation. Cancer Gene Ther 2010; 17:700-7. [PMID: 20559333 DOI: 10.1038/cgt.2010.26] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Vascular endothelial-specific cadherin (VE-cadherin) is an endothelial cell-specific adhesion molecule, localized at cell-cell contact sites. It is involved in physiological and pathological angiogenesis. In this study, we showed that in vitro a soluble N-terminal fragment of VE-cadherin (EC1-3) corresponding to cadherin 1-3 ectodomains inhibited vascular endothelial growth factor-stimulated endothelial cell proliferation and capillary tube structure formation in the matrigel model. In vivo, EC1-3 was tested in a murine colon cancer model. EC1-3-expressing colon cancer C51 cells were subcutaneously grafted into nude mice, and tumor growth and angiogenesis were evaluated. At day 33, the mean volume of the tumors developed was reduced (510±104 versus 990±120 mm(3) for control). Similarly, injection of EC1-3 virus-producing cells into established C51 tumors resulted in an inhibition by 33% of tumor growth. Immunohistological staining of vessels on tumor sections showed a significantly reduced intratumoral angiogenesis. Furthermore, EC1-3 did not induce vessel injury in the lung, liver, spleen, heart and brain in the mice. These results suggest that the soluble N-terminal fragment of VE-cadherin EC1-3 could exert an antitumoral effect by targeting tumor angiogenesis, which included blocking endothelial cell proliferation and capillary tube formation with no obvious toxicity on normal organs.
Collapse
|
34
|
Asano Y, Stawski L, Hant F, Highland K, Silver R, Szalai G, Watson DK, Trojanowska M. Endothelial Fli1 deficiency impairs vascular homeostasis: a role in scleroderma vasculopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1983-98. [PMID: 20228226 DOI: 10.2353/ajpath.2010.090593] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Systemic sclerosis or scleroderma (SSc) is a complex autoimmune connective tissue disease characterized by obliterative vasculopathy and tissue fibrosis. The molecular mechanisms underlying SSc vasculopathy are largely unknown. Friend leukemia integration factor 1 (Fli1), an important regulator of immune function and collagen fibrillogenesis, is expressed at reduced levels in endothelial cells in affected skin of patients with SSc. To develop a disease model and to investigate the function of Fli1 in the vasculature, we generated mice with a conditional deletion of Fli1 in endothelial cells (Fli1 CKO). Fli1 CKO mice showed a disorganized dermal vascular network with greatly compromised vessel integrity and markedly increased vessel permeability. We show that Fli1 regulates expression of genes involved in maintaining vascular homeostasis including VE-cadherin, platelet endothelial cell adhesion molecule 1, type IV collagen, matrix metalloproteinase 9, platelet-derived growth factor B, and S1P(1) receptor. Accordingly, Fli1 CKO mice are characterized by down-regulation of VE-cadherin and platelet endothelial cell adhesion molecule 1, impaired development of basement membrane, and a decreased presence of alpha-smooth muscle actin-positive cells in dermal microvessels. This phenotype is consistent with a role of Fli1 as a regulator of vessel maturation and stabilization. Importantly, vascular characteristics of Fli1 CKO mice are recapitulated by SSc microvasculature. Thus, persistently reduced levels of Fli1 in endothelial cells may play a critical role in the development of SSc vasculopathy.
Collapse
Affiliation(s)
- Yoshihide Asano
- Arthritis Center, Boston University Medical Center, Boston, MA 02118, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Paulis YWJ, Soetekouw PMMB, Verheul HMW, Tjan-Heijnen VCG, Griffioen AW. Signalling pathways in vasculogenic mimicry. Biochim Biophys Acta Rev Cancer 2010; 1806:18-28. [PMID: 20079807 DOI: 10.1016/j.bbcan.2010.01.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/24/2009] [Accepted: 01/07/2010] [Indexed: 12/13/2022]
Abstract
Solid tumour growth is dependent on the development of an adequate blood supply. For years, sprouting angiogenesis has been considered an exclusive mechanism of tumour vascularization. However, over the last years, several other mechanisms have been identified, including vessel-co-option, intussusception, recruitment of endothelial precursor cells (EPCs) and even mechanisms that do not involve endothelial cells, a process called vasculogenic mimicry (VM). The latter describes a mechanism by which highly aggressive tumour cells can form vessel-like structures themselves, by virtue of their high plasticity. VM has been observed in several tumour types and its occurrence is strongly associated with a poor prognosis. This review will focus on signalling molecules and cascades involved in VM. In addition, we will discuss the presence of VM in relation to ongoing cancer research. Finally, we describe the clinical significance of VM regarding anti-angiogenesis treatment modalities.
Collapse
Affiliation(s)
- Yvette W J Paulis
- Department of Internal Medicine, Division of Medical Oncology, School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Nikolova-Krstevski V, Yuan L, Le Bras A, Vijayaraj P, Kondo M, Gebauer I, Bhasin M, Carman CV, Oettgen P. ERG is required for the differentiation of embryonic stem cells along the endothelial lineage. BMC DEVELOPMENTAL BIOLOGY 2009; 9:72. [PMID: 20030844 PMCID: PMC2803788 DOI: 10.1186/1471-213x-9-72] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 12/23/2009] [Indexed: 11/10/2022]
Abstract
BACKGROUND The molecular mechanisms that govern stem cell differentiation along the endothelial lineage remain largely unknown. Ets related gene (ERG) has recently been shown to participate in the transcriptional regulation of a number of endothelial specific genes including VE-cadherin (CD144), endoglin, and von Willebrand's Factor (vWF). The specific role of the ETS factor ERG during endothelial differentiation has not been evaluated. RESULTS ERG expression and function were evaluated during the differentiation of embryonic stem cells into embryoid bodies (EB). The results of our study demonstrate that ERG is first expressed in a subpopulation of vascular endothelial growth factor receptor 2 (VEGF-R2) expressing cells that also express VE-cadherin. During ES cell differentiation, ERG expression remains restricted to cells of the endothelial lineage that eventually coalesce into primitive vascular structures within embryoid bodies. ERG also exhibits an endothelial cell (EC)-restricted pattern during embryogenesis. To further define the role of ERG during ES cell differentiation, we used a knockdown strategy to inhibit ERG expression. Delivery of three independent shRNA led to 70-85% reductions in ERG expression during ES cell differentiation compared to no change with control shRNA. ERG knockdown was associated with a marked reduction in the number of ECs, the expression of EC-restricted genes, and the formation of vascular structures. CONCLUSION The ETS factor ERG appears to be a critical regulator of EC differentiation.
Collapse
|
37
|
Le Bras A, Soncin F. [Genes that make the endothelial identity]. JOURNAL DE LA SOCIETE DE BIOLOGIE 2009; 203:125-41. [PMID: 19527626 DOI: 10.1051/jbio/2009016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The endothelium is a tissue with a distinct identity due to the specific expression of molecular markers by endothelial cells. Further, the endothelium displays a structural heterogeneity illustrated by the expression of specific markers in arteries and in veins. Here, we present a review of the transcriptional and epigenetic mechanisms regulating the expression of the main markers of endothelial cells in man and mouse, demonstrating that there is no common and unique mechanism of specific expression of genes in these cells.
Collapse
Affiliation(s)
- Alexandra Le Bras
- Institut de Biologie de Lille, CNRS UMR8161, Equipe Labellisée Ligue Nationale contre le Cancer 2008, Université de Lille I, Université de Lille II, Institut Pasteur de Lille, 1, rue Calmette, 59021 Lille Cedex, France
| | | |
Collapse
|
38
|
Yuan L, Nikolova-Krstevski V, Zhan Y, Kondo M, Bhasin M, Varghese L, Yano K, Carman CV, Aird WC, Oettgen P. Antiinflammatory effects of the ETS factor ERG in endothelial cells are mediated through transcriptional repression of the interleukin-8 gene. Circ Res 2009; 104:1049-57. [PMID: 19359602 DOI: 10.1161/circresaha.108.190751] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ERG (Ets-related gene) is an ETS transcription factor that has recently been shown to regulate a number of endothelial cell (EC)-restricted genes including VE-cadherin, von Willebrand factor, endoglin, and intercellular adhesion molecule-2. Our preliminary data demonstrate that unlike other ETS factors, ERG exhibits a highly EC-restricted pattern of expression in cultured primary cells and several adult mouse tissues including the heart, lung, and brain. In response to inflammatory stimuli, such as tumor necrosis factor-alpha, we observed a marked reduction of ERG expression in ECs. To further define the role of ERG in the regulation of normal EC function, we used RNA interference to knock down ERG. Microarray analysis of RNA derived from ERG small interfering RNA- or tumor necrosis factor-alpha-treated human umbilical vein (HUV)ECs revealed significant overlap (P<0.01) in the genes that are up- or downregulated. Of particular interest to us was a significant change in expression of interleukin (IL)-8 at both protein and RNA levels. Exposure of ECs to tumor necrosis factor-alpha is known to be associated with increased neutrophil attachment. We observed that knockdown of ERG in HUVECs is similarly associated with increased neutrophil attachment compared to control small interfering RNA-treated cells. This enhanced adhesion could be blocked with IL-8 neutralizing or IL-8 receptor blocking antibodies. ERG can inhibit the activity of the IL-8 promoter in a dose dependent manner. Direct binding of ERG to the IL-8 promoter in ECs was confirmed by chromatin immunoprecipitation. In summary, our findings support a role for ERG in promoting antiinflammatory effects in ECs through repression of inflammatory genes such as IL-8.
Collapse
Affiliation(s)
- Lei Yuan
- Division of Cardiology, Department of Medicine, and the Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Liu F, Patient R. Genome-Wide Analysis of the Zebrafish ETS Family Identifies Three Genes Required for Hemangioblast Differentiation or Angiogenesis. Circ Res 2008; 103:1147-54. [DOI: 10.1161/circresaha.108.179713] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ETS domain transcription factors have been linked to hematopoiesis, vasculogenesis, and angiogenesis. However, their biological functions and the mechanisms of action, remain incompletely understood. Here, we have performed a systematic analysis of zebrafish ETS domain genes and identified 31 in the genome. Detailed gene expression profiling revealed that 12 of them are expressed in blood and endothelial precursors during embryonic development. Combined with a phylogenetic tree assay, this suggests that some of the coexpressed genes may have redundant or additive functions in these cells. Loss-of-function analysis of 3 of them,
erg
,
fli1
, and
etsrp
, demonstrated that
erg
and
fli1
act cooperatively and are required for angiogenesis possibly via direct regulation of an endothelial cell junction molecule, VE-cadherin, whereas
etsrp
is essential for primitive myeloid/endothelial progenitors (hemangioblasts) in zebrafish. Taken together, these results provide a global view of the ETS genes in the zebrafish genome during embryogenesis and provide new insights on the functions and biology of
erg
,
fli1
, and
etsrp
, which could be applicable to higher vertebrates, including mice and humans.
Collapse
Affiliation(s)
- Feng Liu
- From the Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, United Kingdom
| | - Roger Patient
- From the Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, United Kingdom
| |
Collapse
|
40
|
Bernaciak J, Szczałuba K, Derwińska K, Wiśniowiecka-Kowalnik B, Bocian E, Sąsiadek MM, Makowska I, Stankiewicz P, Śmigiel R. Clinical and molecular-cytogenetic evaluation of a family with partial Jacobsen syndrome without thrombocytopenia caused by an ∼5 Mb deletion del(11)(q24.3). Am J Med Genet A 2008; 146A:2449-54. [DOI: 10.1002/ajmg.a.32490] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
41
|
Endo Y, Deonauth K, Prahalad P, Hoxter B, Zhu Y, Byers SW. Role of Sox-9, ER81 and VE-cadherin in retinoic acid-mediated trans-differentiation of breast cancer cells. PLoS One 2008; 3:e2714. [PMID: 18628953 PMCID: PMC2444023 DOI: 10.1371/journal.pone.0002714] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Accepted: 05/19/2008] [Indexed: 11/19/2022] Open
Abstract
Many aspects of development, tumor growth and metastasis depend upon the provision of an adequate vasculature. This can be a result of regulated angiogenesis, recruitment of circulating endothelial progenitors and/or vascular trans-differentiation. The present study demonstrates that treatment of SKBR-3 breast cancer cells with retinoic acid (RA), an important regulator of embryogenesis, cancer and other diseases, stimulates the formation of networks in Matrigel. RA-treatment of SKBR-3 cells co-cultured with human umbilical vein endothelial cells resulted in the formation of mixed structures. RA induces expression of many endothelial genes including vascular endothelial (VE) cadherin. VE-cadherin was also induced by RA in a number of other breast cancer cells. We show that RA-induced VE-cadherin is responsible for the RA-induced morphological changes. RA rapidly induced the expression of Sox-9 and ER81, which in turn form a complex on the VE-cadherin promoter and are required to mediate the transcriptional regulation of VE-cadherin by RA. These data indicate that RA may promote the expression of endothelial genes resulting in endothelial-like differentiation, or provide a mechanism whereby circulating endothelial progenitor cells could be incorporated into a growing organ or tumor.
Collapse
Affiliation(s)
- Yoshimi Endo
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
| | - Kamla Deonauth
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
- Department of Biology, Howard University, Washington D. C., United States of America
| | - Priya Prahalad
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
| | - Becky Hoxter
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
| | - Yuelin Zhu
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
| | - Stephen W. Byers
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington D. C., United States of America
- * E-mail:
| |
Collapse
|
42
|
A GABP-binding element in the Robo4 promoter is necessary for endothelial expression in vivo. Blood 2008; 112:2336-9. [PMID: 18519813 DOI: 10.1182/blood-2008-01-135079] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We recently demonstrated that the 3-kb 5'-flanking region of the human ROBO4 gene directs endothelial cell-specific expression in vitro and in vivo. Moreover, a GA-binding protein (GABP)-binding motif at -119 was necessary for mediating promoter activity in vitro. The goal of the present study was to confirm the functional relevance of the -119 GABP-binding site in vivo. To that end, the Hprt locus of mice was targeted with a Robo4-LacZ transgenic cassette in which the GABP site was mutated. In other studies, the GABP mutation was introduced into the endogenous mouse Robo4 locus in which LacZ was knocked-in. Compared with their respective controls, the mutant promoters displayed a significant reduction in activity in embryoid bodies, embryos, and adult animals. Together, these data provide strong support for the role of the GABP-binding motif in mediating Robo4 expression in the intact endothelium.
Collapse
|
43
|
Transcription factor Erg regulates angiogenesis and endothelial apoptosis through VE-cadherin. Blood 2008; 111:3498-506. [PMID: 18195090 DOI: 10.1182/blood-2007-08-105346] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tight regulation of the balance between apoptosis and survival is essential in angiogenesis. The ETS transcription factor Erg is required for endothelial tube formation in vitro. Inhibition of Erg expression in human umbilical vein endothelial cells (HUVECs), using antisense oligonucleotides, resulted in detachment of cell-cell contacts and increased cell death. Inhibition of Erg expression by antisense in HUVECs also lowered expression of the adhesion molecule vascular endothelial (VE)-cadherin, a key regulator of endothelial intercellular junctions and survival. Using chromatin immunoprecipitation, we showed that Erg binds to the VE-cadherin promoter. Furthermore, Erg was found to enhance VE-cadherin promoter activity in a transactivation assay. Apoptosis induced by inhibition of Erg was partly rescued by overexpression of VE-cadherin-GFP, suggesting that VE-cadherin is involved in the Erg-dependent survival signals. To show the role of Erg in angiogenesis in vivo, we used siRNA against Erg in a Matrigel plug model. Erg inhibition resulted in a significant decrease in vascularization, with increase in caspase-positive endothelial cells (ECs). These results identify a new pathway regulating angiogenesis and endothelial survival, via the transcription factor Erg and the adhesion molecule VE-cadherin.
Collapse
|
44
|
Vestweber D. VE-cadherin: the major endothelial adhesion molecule controlling cellular junctions and blood vessel formation. Arterioscler Thromb Vasc Biol 2007; 28:223-32. [PMID: 18162609 DOI: 10.1161/atvbaha.107.158014] [Citation(s) in RCA: 618] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vascular endothelial (VE)-cadherin is a strictly endothelial specific adhesion molecule located at junctions between endothelial cells. In analogy of the role of E-cadherin as major determinant for epithelial cell contact integrity, VE-cadherin is of vital importance for the maintenance and control of endothelial cell contacts. Mechanisms that regulate VE-cadherin-mediated adhesion are important for the control of vascular permeability and leukocyte extravasation. In addition to its adhesive functions, VE-cadherin regulates various cellular processes such as cell proliferation and apoptosis and modulates vascular endothelial growth factor receptor functions. Consequently, VE-cadherin is essential during embryonic angiogenesis. This review will focus on recent new developments in understanding the role of VE-cadherin in controlling endothelial cell contacts and influencing endothelial cell behavior by various outside-in signaling processes.
Collapse
Affiliation(s)
- Dietmar Vestweber
- Max-Planck-Institute of Molecular Biomedicine, Röntgenstr. 20, D-48149 Münster, Germany.
| |
Collapse
|
45
|
Asija A, Peterson SJ, Stec DE, Abraham NG. Targeting endothelial cells with heme oxygenase-1 gene using VE-cadherin promoter attenuates hyperglycemia-mediated cell injury and apoptosis. Antioxid Redox Signal 2007; 9:2065-74. [PMID: 17883332 DOI: 10.1089/ars.2007.1804] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Risk factors for cardiovascular diseases include hyperglycemia, TNF, and reactive oxygen species (ROS), which collectively contribute to vascular endothelial cell dysfunction and apoptosis. We examined, in vascular endothelial cells, whether the selective expression of heme oxygenase-1 (HO-1) offers cytoprotection against glucose- and TNF-mediated cell death. An adenoviral vector expressing human HO-1 was constructed using a VE-cadherin (VECAD) promotor fragment, and cell-specific expression of the recombinant adenovirus was examined using endothelial and vascular smooth muscle cells. The effects of HO-1 transduction (Ad-VECAD-HO-1 gene) on HO-1 expression, HO activity, and the response to TNF and hyperglycemia were studied. Human HO-1 gene was selectively expressed in endothelial cells after infection with the Ad-VECAD-HO-1 vector. Selective expression of HO-1 prevented TNF- and hyperglycemia-mediated superoxide (O2-) formation, DNA degeneration, and upregulation of caspase, but increased the expression of pAkt and Bcl-xL, proteins responsible for endothelial dysfunction in diabetes. These results demonstrate that endothelial cell survival after oxidative stress injury may be enhanced by targeting HO-1 expression, thus blocking inflammation, apoptosis, and thereby attenuating cardiovascular risk factors.
Collapse
Affiliation(s)
- Amit Asija
- Department of Medicine, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | |
Collapse
|
46
|
Wallez Y, Huber P. Endothelial adherens and tight junctions in vascular homeostasis, inflammation and angiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1778:794-809. [PMID: 17961505 DOI: 10.1016/j.bbamem.2007.09.003] [Citation(s) in RCA: 338] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 09/03/2007] [Accepted: 09/05/2007] [Indexed: 11/25/2022]
Abstract
Endothelial cells lining the vessel wall are connected by adherens, tight and gap junctions. These junctional complexes are related to those found at epithelial junctions but with notable changes in terms of specific molecules and organization. Endothelial junctional proteins play important roles in tissue integrity but also in vascular permeability, leukocyte extravasation and angiogenesis. In this review, we will focus on specific mechanisms of endothelial tight and adherens junctions.
Collapse
Affiliation(s)
- Yann Wallez
- Vascular Pathophysiology Laboratory, Inserm U882 38054 Grenoble, France
| | | |
Collapse
|
47
|
Le Bras A, Lionneton F, Mattot V, Lelièvre E, Caetano B, Spruyt N, Soncin F. HIF-2alpha specifically activates the VE-cadherin promoter independently of hypoxia and in synergy with Ets-1 through two essential ETS-binding sites. Oncogene 2007; 26:7480-9. [PMID: 17563748 DOI: 10.1038/sj.onc.1210566] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mechanisms that are responsible for the restricted pattern of expression of the VE-cadherin gene in endothelial cells are not clearly understood. Regulation of expression is under the control of an approximately 140 bp proximal promoter that provides basal, non-endothelial specific expression. A larger region contained within the 2.5 kb genomic DNA sequence located ahead of the transcription start is involved in the specific expression of the gene in endothelial cells. We show here that the VE-cadherin promoter contains several putative hypoxia response elements (HRE) which are able to bind endothelial nuclear factors under normoxia. The VE-cadherin gene is not responsive to hypoxia but hypoxia-inducible factor (HIF)-2alpha specifically activates the promoter while HIF-1alpha does not. The HRE, that are involved in this activity have been identified. Further, we show that HIF-2alpha cooperates with the Ets-1 transcription factor for activation of the VE-cadherin promoter and that this synergy is dependent on the binding of Ets-1 to DNA. This cooperative action of HIF-2alpha with Ets-1 most probably participates to the transcriptional regulation of expression of the gene in endothelial cells. This mechanism may also be involved in the expression of the VE-cadherin gene by tumor cells in the process of vascular mimicry.
Collapse
Affiliation(s)
- A Le Bras
- CNRS, UMR8161, Lille France; Univ Lille I, Lille, France; Univ Lille II, Lille, France; Inst Pasteur de Lille, Lille, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Okada Y, Yano K, Jin E, Funahashi N, Kitayama M, Doi T, Spokes K, Beeler DL, Shih SC, Okada H, Danilov TA, Maynard E, Minami T, Oettgen P, Aird WC. A three-kilobase fragment of the human Robo4 promoter directs cell type-specific expression in endothelium. Circ Res 2007; 100:1712-22. [PMID: 17495228 DOI: 10.1161/01.res.0000269779.10644.dc] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Robo4, a member of the roundabout family, is expressed exclusively in endothelial cells and has been implicated in endothelial cell migration and angiogenesis. Here we report the cloning and characterization of the human Robo4 promoter. The 3-kb 5'-flanking region directs endothelial cell-specific expression in vitro. Deletion and mutation analyses revealed the functional importance of two 12-bp palindromic DNA sequences at -2528 and -2941, 2 SP1 consensus motifs at -42 and -153, and an ETS consensus motif at -119. In electrophoretic mobility shift assays using supershifting antibodies, the SP1 motifs bound SP1 protein, whereas the ETS site bound a heterodimeric member of the ETS family, GA binding protein (GABP). These DNA-protein interactions were confirmed by chromatin immunoprecipitation assays. Transfection of primary human endothelial cells with small interfering RNA against GABP and SP1 resulted in a significant (approximately 50%) reduction in endogenous Robo4 mRNA expression. The 3-kb Robo4 promoter was coupled to LacZ, and the resulting cassette was introduced into the Hprt locus of mice by homologous recombination. Reporter gene activity was observed in the vasculature of adult organs (particularly in microvessels), tumor xenografts, and embryos, where it colocalized with the endothelial cell-specific marker CD31. LacZ mRNA levels in adult tissues and tumors correlated with mRNA levels for endogenous Robo4, CD31, and vascular endothelial cadherin. Moreover, the pattern of reporter gene expression was similar to that observed in mice in which LacZ was knocked into the endogenous Robo4 locus. Together, these data suggest that 3-kb upstream promoter of human Robo4 contains information for cell type-specific expression in the intact endothelium.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cadherins/metabolism
- Cells, Cultured
- Cloning, Molecular
- DNA/genetics
- DNA Mutational Analysis
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- GA-Binding Protein Transcription Factor/physiology
- Gene Expression Regulation
- Humans
- Lac Operon
- Mice
- Molecular Sequence Data
- Peptide Fragments/genetics
- Peptide Fragments/physiology
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/physiology
- Protein Binding/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/pharmacology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Sequence Analysis, DNA
- Sp1 Transcription Factor/physiology
- Transfection
Collapse
Affiliation(s)
- Yoshiaki Okada
- Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Deleuze V, Chalhoub E, El-Hajj R, Dohet C, Le Clech M, Couraud PO, Huber P, Mathieu D. TAL-1/SCL and its partners E47 and LMO2 up-regulate VE-cadherin expression in endothelial cells. Mol Cell Biol 2007; 27:2687-97. [PMID: 17242194 PMCID: PMC1899886 DOI: 10.1128/mcb.00493-06] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The basic helix-loop-helix TAL-1/SCL essential for hematopoietic development is also required during vascular development for embryonic angiogenesis. We reported that TAL-1 acts positively on postnatal angiogenesis by stimulating endothelial morphogenesis. Here, we investigated the functional consequences of TAL-1 silencing in human primary endothelial cells. We found that TAL-1 knockdown caused the inhibition of in vitro tubulomorphogenesis, which was associated with a dramatic reduction in vascular endothelial cadherin (VE-cadherin) at intercellular junctions. Consistently, silencing of TAL-1 as well as of its cofactors E47 and LMO2 down-regulated VE-cadherin at both the mRNA and the protein level. Endogenous VE-cadherin transcription could be activated in nonendothelial HEK-293 cells by the sole concomitant ectopic expression of TAL-1, E47, and LMO2. Transient transfections in human primary endothelial cells derived from umbilical vein (HUVECs) demonstrated that VE-cadherin promoter activity was dependent on the integrity of a specialized E-box associated with a GATA motif and was maximal with the coexpression of the different components of the TAL-1 complex. Finally, chromatin immunoprecipitation assays showed that TAL-1 and its cofactors occupied the VE-cadherin promoter in HUVECs. Together, these data identify VE-cadherin as a bona fide target gene of the TAL-1 complex in the endothelial lineage, providing a first clue to TAL-1 function in angiogenesis.
Collapse
Affiliation(s)
- Virginie Deleuze
- Institut de Génétique Moléculaire de Montpellier CNRS, UMR5535, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wallez Y, Vilgrain I, Huber P. Angiogenesis: The VE-Cadherin Switch. Trends Cardiovasc Med 2006; 16:55-9. [PMID: 16473763 DOI: 10.1016/j.tcm.2005.11.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Revised: 11/21/2005] [Accepted: 11/30/2005] [Indexed: 11/25/2022]
Abstract
Because angiogenesis is a key step in a number of pathologic processes, including tumor growth and atherosclerosis, many research studies have investigated the regulatory signals active at various stages of vascular invasion. The differential activities of the endothelial junction protein vascular endothelial (VE)-cadherin reflect the versatile behavior of endothelial cells between vascular quiescence and angiogenesis. VE-cadherin function and signaling are deeply modified in proliferating cells, and this conversion is accompanied by phosphorylation of the protein on tyrosine residues and enhanced transcription of its gene. Recent advances in the complex interplay between protein tyrosine kinases and phosphatases regulating VE-cadherin phosphorylation and function are discussed in this review.
Collapse
Affiliation(s)
- Yann Wallez
- Laboratoire de Développement et Vieillissement de l'Endothélium, Université Joseph Fourier, Grenoble, France
| | | | | |
Collapse
|