scholarly journals Role of the AP-1 transcription factor FOSL1 in endothelial cells adhesion and migration

2013 ◽  
Vol 7 (5) ◽  
pp. 408-411 ◽  
Author(s):  
Federico Galvagni ◽  
Maurizio Orlandini ◽  
Salvatore Oliviero
Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 1870
Author(s):  
Klaudia Skrzypek ◽  
Grażyna Adamek ◽  
Marta Kot ◽  
Bogna Badyra ◽  
Marcin Majka

Rhabdomyosarcoma (RMS), is the most frequent soft tissue tumor in children that originates from disturbances in differentiation process. Mechanisms leading to the development of RMS are still poorly understood. Therefore, by analysis of two RMS RH30 cell line subclones, one subclone PAX7 negative, while the second one PAX7 positive, and comparison with other RMS cell lines we aimed at identifying new mechanisms crucial for RMS progression. RH30 subclones were characterized by the same STR profile, but different morphology, rate of proliferation, migration activity and chemotactic abilities in vitro, as well as differences in tumor morphology and growth in vivo. Our analysis indicated a different level of expression of adhesion molecules (e.g., from VLA and ICAM families), myogenic microRNAs, such as miR-206 and transcription factors, such as MYOD, MYOG, SIX1, and ID. Silencing of PAX7 transcription factor with siRNA confirmed the crucial role of PAX7 transcription factor in proliferation, differentiation and migration of RMS cells. To conclude, our results suggest that tumor cell lines with the same STR profile can produce subclones that differ in many features and indicate crucial roles of PAX7 and ID proteins in the development of RMS.


2021 ◽  
Vol 22 (6) ◽  
pp. 2804
Author(s):  
Yasuo Yoshitomi ◽  
Takayuki Ikeda ◽  
Hidehito Saito-Takatsuji ◽  
Hideto Yonekura

Blood vessels are essential for the formation and maintenance of almost all functional tissues. They play fundamental roles in the supply of oxygen and nutrition, as well as development and morphogenesis. Vascular endothelial cells are the main factor in blood vessel formation. Recently, research findings showed heterogeneity in vascular endothelial cells in different tissue/organs. Endothelial cells alter their gene expressions depending on their cell fate or angiogenic states of vascular development in normal and pathological processes. Studies on gene regulation in endothelial cells demonstrated that the activator protein 1 (AP-1) transcription factors are implicated in angiogenesis and vascular development. In particular, it has been revealed that JunB (a member of the AP-1 transcription factor family) is transiently induced in endothelial cells at the angiogenic frontier and controls them on tip cells specification during vascular development. Moreover, JunB plays a role in tissue-specific vascular maturation processes during neurovascular interaction in mouse embryonic skin and retina vasculatures. Thus, JunB appears to be a new angiogenic factor that induces endothelial cell migration and sprouting particularly in neurovascular interaction during vascular development. In this review, we discuss the recently identified role of JunB in endothelial cells and blood vessel formation.


2000 ◽  
Vol 89 (6) ◽  
pp. 2391-2400 ◽  
Author(s):  
Hiroyuki Kito ◽  
Emery L. Chen ◽  
Xiujie Wang ◽  
Masataka Ikeda ◽  
Nobuyoshi Azuma ◽  
...  

The aim of this study was to examine the role of mitogen-activated protein kinases (MAPKs) activation in bovine pulmonary arterial endothelial cells (EC) exposed to cyclic strain. EC were subjected to 10% average strain at 60 cycles/min. Cyclic strain induced activation of extracellular signal-regulated kinase (ERK; 1.5-fold), c-Jun NH2-terminal protein kinase (JNK; 1.9-fold), and p38 (1.5-fold) with a peak at 30 min. To investigate the functional role of the activated MAPKs, we analyzed cells after treatment with PD-98059, a specific ERK kinase inhibitor, or SB-203580, a catalytic inhibitor for p38, and after transient transfection with JNK(K-R), and MEKK(K-M) the respective catalytically inactive mutants of JNK1 and MAPK kinase kinase-1. Cyclic strain increased activator protein-1 (AP-1) binding activity, which was blocked by PD-98059 and SB-203580. Activity of AP-1-dependent luciferase reporter driven by 12- O-tetradecanoyl-phorbol-13-acetate-responsive element (TRE) was induced by cyclic strain, and this was attenuated by PD-98059, MEKK(K-M), JNK(K-R), and SB-203580. PD-98059 and SB-203850 did not inhibit cell alignment and migration induced by cyclic strain. MEKK(K-M) and JNK(K-R) transfection did not block cyclic strain-induced cell alignment. In conclusion, cyclic strain activates ERK, JNK, and p38, and their activation plays a role in transcriptional activation of AP-1/TRE but not in cell alignment and migration changes in bovine pulmonary arterial EC.


2017 ◽  
Vol 95 (10) ◽  
pp. 1263-1270 ◽  
Author(s):  
Vibhuti Sharma ◽  
Nilambra Dogra ◽  
Uma Nahar Saikia ◽  
Madhu Khullar

The etiology of cardiac fibrogenesis is quite diverse, but a common feature is the presence of activated fibroblasts. Experimental evidence suggests that a subset of cardiac fibroblasts is derived via transition of vascular endothelial cells into fibroblasts by endothelial-to-mesenchymal transition (EndMT). During EndMT, endothelial cells lose their endothelial characteristics and acquire a mesenchymal phenotype. Molecular mechanisms and the transcriptional mediators controlling EndMT in heart during development or disease remain relatively undefined. Myocardin-related transcription factor A facilitates the transcription of cytoskeletal genes by serum response factor during fibrosis; therefore, its specific role in cardiac EndMT might be of importance. Activation of activating transcription factor 3 (ATF-3) during cardiac EndMT is speculative, since ATF-3 responds to a transforming growth factor β (TGF-β) stimulus and controls the expression of the primary epithelial-to-mesenchymal transition markers Snail, Slug, and Twist. Although the role of TGF-β in EndMT-mediated cardiac fibrosis has been established, targeting of the TGF-β ligand has not proven to be a viable anti-fibrotic strategy owing to the broad functional importance of this ligand. Thus, targeting of downstream transcriptional mediators may be a useful therapeutic approach in attenuating cardiac fibrosis. Here, we discuss some of the transcription factors that may regulate EndMT-mediated cardiac fibrosis and their involvement in type 2 diabetes.


2009 ◽  
Vol 11 (4) ◽  
pp. 747-764 ◽  
Author(s):  
Srikanth Pendyala ◽  
Irina A Gorshkova ◽  
Peter V. Usatyuk ◽  
Donghong He ◽  
Arjun Pennathur ◽  
...  

2016 ◽  
Vol 2016 ◽  
pp. 1-10 ◽  
Author(s):  
Shuai Wang ◽  
Zewei Sun ◽  
Wenting Zhao ◽  
Zhen Wang ◽  
Mingjie Wu ◽  
...  

CD97/ADGRE5 protein is predominantly expressed on leukocytes and belongs to the EGF-TM7 receptors family. It mediates granulocytes accumulation in the inflammatory tissues and is involved in firm adhesion of PMNC on activated endothelial cells. There have not been any studies exploring the role of CD97 in LPS induced NF-κB activation in macrophages. Therefore, we first measured the CD97 expression in LPS treated human primary macrophages and subsequently analyzed the levels of inflammatory factor TNF-αand transcription factor NF-κB in these macrophages that have been manipulated with either CD97 knockdown or overexpression. We found that a reported anti-inflammatory transcription factor, PPAR-γ, was involved in the CD97 mediated NF-κB suppression. Furthermore, by immunofluorescence staining, we established that CD97 overexpression not only inhibited LPS induced p65 expression in the nucleus but also promoted the PPAR-γexpression. Moreover, using CD97 knockout THP-1 cells, we further demonstrated that CD97 promoted PPAR-γexpression and decreased LPS induced NF-κB activation. In conclusion, CD97 plays a negative role in LPS induced NF-κB activation and TNF-αsecretion, partly through PPAR-γupregulation.


2018 ◽  
Author(s):  
Alethia Villasenor ◽  
Sébastien Gauvrit ◽  
Michelle M. Collins ◽  
Silvia Parajes ◽  
Hans-Martin Maischein ◽  
...  

SUMMARYSignificant efforts have advanced our understanding of foregut-derived organ development; however, little is known about the molecular mechanisms that underlie the formation of the hepatopancreatic ductal (HPD) system. Here, we report a role for the homeodomain transcription factor Hhex in directing HPD progenitor specification in zebrafish. Loss of Hhex function results in impaired HPD system formation. We found that Hhex specifies a distinct population of HPD progenitors that gives rise to the cystic duct, common bile duct, and extra-pancreatic duct. Since hhex is not uniquely expressed in the HPD region but is also expressed in endothelial cells and the yolk syncytial layer (YSL), we tested the role of blood vessels as well as the YSL in HPD formation. We found that blood vessels are required for HPD patterning, but not for HPD progenitor specification. In addition, we found that Hhex is required in both the endoderm and the YSL for HPD development. Our results shed light on the mechanisms necessary to direct endodermal progenitors towards the HPD fate and also advance our understanding of HPD system formation.


2020 ◽  
Author(s):  
Feng Liu ◽  
Hao Wu ◽  
Guangyong Wu ◽  
Jun Long ◽  
Jin Dai ◽  
...  

Abstract Background: Long noncoding RNAs are widely studied in glioma. However, the role of the lncRNA NEAT1 and KDM3A in glioma has not yet been reported. We aimed to reveal the role of these two lncRNAs in the development of glioma through this study.Methods: Samples from glioma patients and normal brain tissues were collected, and the expression of NEAT1 was detected by qRT-PCR. A dual-luciferase reporter gene assay, chromatin immunoprecipitation (ChIP), RNA-binding protein immunoprecipitation (RIP), and RNA pulldown experiments were used to identify the relationship between FOXK1, NEAT1, miR-128, and KDM3A. The CCK8 assay, Transwell assay and flow cytometry were used to detect cell viability, invasion and migration ability, and the cell cycle and apoptosis, respectively. Tumor formation experiments verified the effect of NEAT1 on gliomas in vivo.Results: FOXK1 and NEAT1 were significantly overexpressed in glioma tissues and cells, and NEAT1 was significantly related to WHO classification. FOXK1 bound the NEAT1 gene promoter region in glioma cells, and interference with FOXK1 inhibited NEAT1 expression. NEAT1 inhibited miR-128 expression by binding miR-128; significantly improved cell viability and invasion and migration capabilities; increased the expression of KDM3A and activated the Wnt signaling pathway. Interference with KDM3A reversed the above results. In addition, interference with NEAT1 decreased KDM3A expression and inhibited tumor growth.Conclusion: Interference with NEAT1 promoted the expression of miR-128, thereby suppressing the expression of KDM3A and inhibiting the occurrence and development of glioma, while the expression of NEAT1 was shown to be regulated by the upstream transcription factor FOXK1.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 840-840
Author(s):  
Vesselina G. Cooke ◽  
Meghna U. Naik ◽  
William Skarnes ◽  
Ulhas P. Naik

Abstract Neovascularization is a multistep process that occurs in the body in both physiological and pathological conditions. We have recently shown that Junctional Adhesion Molecule-1 (JAM-1), a member of the Ig superfamily of molecules, is involved in endothelial cell adhesion and migration, leading to angiogenesis. In quiescent endothelial cells, JAM-1 is located at the cell-cell junctions where it forms a complex with integrin αvβ3. Upon treatment of the cells with growth factors, such as bFGF, JAM-1 dissociates from its complex with αvβ3 and redistributes to the cell surface. Blockage of the extracellular domain of JAM-1 inhibits bFGF-induced endothelial cell morphology, proliferation and angiogenesis. Additionally, functional knock-down of JAM-1 using the RNAi technique in endothelial cells showed decreased adhesion and migration of these cells, indicating a possible role for JAM-1 in angiogenesis. In this report, we show that JAM-1 has an important role in bFGF-induced angiogenesis in vivo. Here we present for the first time the generation JAM-1 knock-out mice, using the gene trap strategy. We have successfully confirmed the JAM-1 −/− genotype via Southern, Northern, and Western blot analyses. JAM-1 −/− mice are viable and do not seem to have any external abnormalities, except that they appear to be smaller in size. Retinal fluorescein angiogram revealed no evidence for morphological defects in the vasculature of JAM-1 −/− mice. To evaluate the role of JAM-1 in angiogenesis, we performed an aortic ring assay with both wild type and JAM-1−/− mice. Mouse thoracic aortas were harvested, cross-sectioned into rings of 1-mm thickness, and cultured in a three-dimensional Matrigel supplied with 50 ng/ml bFGF. Vascular sproutings were counted every other day for a period of 7 days at which time they were stained with crystal violet and photographed. Aortic rings from WT mice treated with bFGF showed a 2.8-fold increase in microvessel growth, compared to WT controls with no supplementation of bFGF. In contrast, microvessel sproutings in bFGF treated aortic rings from JAM-1 −/− mice were no more than the vessels in the WT control mice. These results suggest that JAM-1 may be important for bFGF induced angiogenesis. To further confirm the role of JAM-1 in angiogenesis, WT and JAM-1 −/− mice were injected in their flank region with Matrigel containing 80 ng/ml bFGF and 60 U/ml heparin. Two weeks after injection, Matrigel plugs were excised, embedded in paraffin, and the presence of blood vessels was visualized by H&E staining. Matrigel plugs from control WT mice that were not treated with bFGF showed no vascularization, while bFGF supplied Matrigel plugs from WT mice showed a robust vessel growth. Interestingly, bFGF-treated Matrigel plugs form JAM-1−/− mice failed to produce any blood vessels. These ex vivo and in vivo studies using JAM-1−/− mice suggest that JAM-1 has a unique and essential role in bFGF-induced angiogenesis.


Sign in / Sign up

Export Citation Format

Share Document