Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054818

ABSTRACT

Space microgravity condition has great physiological influence on astronauts' health. The interaction of endothelial cells, which control vascular permeability and immune responses, is sensitive to mechanical stress. However, whether microgravity has significant effects on the physiological function of the endothelium has not been investigated. In order to address such a question, a clinostat-based culture model with a HUVEC monolayer being inside the culture vessel under the simulated microgravity (SMG) was established. The transmittance of FITC-tagged dextran was used to estimate the change of integrity of the adherens junction of the HUVEC monolayer. Firstly, we found that the permeability of the HUVEC monolayer was largely increased after SMG treatment. To elucidate the mechanism of the increased permeability of the HUVEC monolayer under SMG, the levels of total expression and activated protein levels of Rap1 and Rap2 in HUVEC cells, which regulate the adherens junction of endothelial cells, were detected by WB and GST pull-down after SMG. As the activation of both Rap1 and Rap2 was significantly decreased under SMG, the expression of Rap1GEF1 (C3G) and Rap1GAP in HUVECs, which regulate the activation of them, was further determined. The results indicate that both C3G and Rap1GAP showed a time-dependent increase with the expression of Rap1GAP being dominant at 48 h after SMG. The down-regulation of the expression of junctional proteins, VE-cadherin and ß-catenin, in HUVEC cells was also confirmed by WB and immunofluorescence after SMG. To clarify whether up-regulation of Rap1GAP is necessary for the increased permeability of the HUVEC monolayer after SMG, the expression of Rap1GAP was knocked down by Rap1GAP-shRNA, and the change of permeability of the HUVEC monolayer was detected. The results indicate that knock-down of Rap1GAP reduced SMG-induced leaking of the HUVEC monolayer in a time-dependent manner. In total, our results indicate that the Rap1GAP-Rap signal axis was necessary for the increased permeability of the HUVEC monolayer along with the down-regulation of junctional molecules including VE-cadherin and ß-catenin.


Subject(s)
GTPase-Activating Proteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Up-Regulation , Weightlessness Simulation , rap GTP-Binding Proteins/metabolism , Actin Cytoskeleton/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Membrane Permeability , Down-Regulation , Endothelium, Vascular/metabolism , Enzyme Activation , GTPase-Activating Proteins/metabolism , Humans , Models, Biological , beta Catenin/metabolism
2.
Int J Mol Sci ; 21(4)2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32079291

ABSTRACT

In order to study the effect of stress changes on cell adhesion, HUVEC, and MCF-7 cells were treated with simulated microgravity effect (SMG) and overloading (OL). METHODS: Rotating Wall Vessel (2D-RWVS) bioreactor was used to create different culture conditions. In addition, the alteration of cell adhesion states, adhesion proteins, and relating factors of adhesion molecules under these two conditions were detected using cell adhesion assay, immunofluorescence, western blot, and qRT-PCR technology. RESULTS: The results showed that the adhesion of cells decreased under SMG, while increased under OL. The expressions of integrin ß1, paxillin, and E-cadherin under SMG condition were down-regulated as compared to that of the control group showing a time-dependent pattern of the decreasing. However, under OL condition, the expressions of adhesion proteins were up-regulated as compared to that of the control group, with a time-dependent pattern of increasing. EMT transcription factors Snail, twist, and ZEB1 were up-regulated under SMG while down-regulated under OL. CONCLUSION: Collectively our results indicated that cells could respond to stress changes to regulate the expressions of adhesion proteins and adapt their adhesion state to the altered mechanical environment. The altered cell adhesion in response to the mechanical stress may involve the changed expression of EMT-inducing factors, Snail, Twist, and ZEB1under the SMG/OL conditions.


Subject(s)
Cadherins/metabolism , Cell Adhesion/physiology , Organic Cation Transport Proteins/metabolism , Transcription Factors/metabolism , Weightlessness Simulation/methods , Antigens, CD , Down-Regulation , Gene Expression , Humans , Integrin beta1/metabolism , MCF-7 Cells , Paxillin/metabolism , Snail Family Transcription Factors , Up-Regulation , Weightlessness , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
3.
ACS Appl Mater Interfaces ; 9(11): 9327-9338, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28240531

ABSTRACT

Hyaluronan (HA), a polymer with various molecular weights (MW) found in tumor microenvironments, is associated with malignant progression of breast cancer. Reducing the amount of high-MW HA in the microenvironment by hyaluronidase is a promising approach for breast cancer treatment. However, whether the generation of HA fragments negatively affects breast cancer cells remains to be determined. Furthermore, HA forms three-dimensional (3D) networks by cross-linking with other extracellular molecules to function. Therefore, a model mimicking the cross-linked HA network is required to determine the effect of HA fragments on breast cancer cells. To clarify the differential roles of low (HA35) versus high (HA117) MW HA on cancer cell phenotype, a 3D culture system was set up by covalently cross-linking HA with alginate and investigating the behavior of 4T-1 and SKBR3 breast cancer cells alongside a two-dimensional (2D) control. The results show the invasion and migration abilities of 4T-1 and SKBR3 cells are significantly enhanced by the presence of HA35 but inhibited by HA117 in both 2D monolayers and 3D spheroids. The differential effects of HA35 and HA117 on cancer cell epithelial-mesenchymal transition (EMT) phenotype were further confirmed in terms of differential regulation of E-cadherin and vimentin as important EMT markers at both the cellular and mRNA levels. Additional experiments show the CD44-Twist signaling pathway might be involved in the differential effects of HA35 and HA117. These results have important implications with respect to understanding the role of HA in breast cancer development and for the design of therapeutic approaches based on the eradication of HA with hyaluronidase.


Subject(s)
Hyaluronic Acid/chemistry , Breast Neoplasms , Cell Line, Tumor , Cell Movement , Humans , Hyaluronan Receptors , Hyaluronoglucosaminidase , Molecular Weight , Neoplasm Proteins , Tumor Microenvironment
4.
PLoS One ; 9(10): e110563, 2014.
Article in English | MEDLINE | ID: mdl-25333650

ABSTRACT

Gene regulatory networks (GRNs) coherently coordinate the expressions of genes and control the behaviors of cellular systems. The complexity in modeling a quantitative GRN usually results from inaccurate parameter estimation, which is mostly due to small sample sizes. For better modeling of GRNs, we have designed a small-sample iterative optimization algorithm (SSIO) to quantitatively model GRNs with nonlinear regulatory relationships. The algorithm utilizes gene expression data as the primary input and it can be applied in case of small-sized samples. Using SSIO, we have quantitatively constructed the dynamic models for the GRNs controlling human and mouse adipogenesis. Compared with two other commonly-used methods, SSIO shows better performance with relatively lower residual errors, and it generates rational predictions on the adipocyte responses to external signals and steady-states. Sensitivity analysis further indicates the validity of our method. Several differences are observed between the GRNs of human and mouse adipocyte differentiations, suggesting the differences in regulatory efficiencies of the transcription factors between the two species. In addition, we use SSIO to quantitatively determine the strengths of the regulatory interactions as well as to optimize regulatory models. The results indicate that SSIO facilitates better investigation and understanding of gene regulatory processes.


Subject(s)
Algorithms , Models, Genetic , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Animals , Bayes Theorem , Cells, Cultured , Gene Regulatory Networks , Humans , Markov Chains , Mice , Monte Carlo Method
5.
PLoS One ; 9(6): e100595, 2014.
Article in English | MEDLINE | ID: mdl-24963972

ABSTRACT

Experimental study was made by keeping human peripheral blood lymphocytes under simulated microgravity in a Rotary Cell Culture System bioreactor to investigate the changes that occur in the number of chromosomes, the expression rate of chromosome fragile site, and the expressions of DNA replication- and repair-related genes. Experimental results indicate simulated microgravity has no effect on the numerical chromosome instability of human peripheral blood lymphocytes, but it enhances the structural chromosome instability of human peripheral blood lymphocytes through the inhibition of DNA replication and the reduction of DNA repair. So, the mechanism of chromosome fragile site induced by simulated microgravity can be explained using the changes that occur in the chromosome structure of human peripheral blood lymphocytes, the DNA replication and repair under the effect of simulated microgravity.


Subject(s)
Chromosomal Instability , Lymphocytes/metabolism , Weightlessness Simulation , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , DNA Repair/genetics , Gene Expression Regulation , Humans , Lymphocytes/cytology , Time Factors
6.
Oncol Rep ; 31(6): 2776-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24700169

ABSTRACT

R-Ras, a member of the Ras superfamily, is expressed in a wide variety of tissues and regulates cell adhesion, migration, differentiation and apoptosis. Research has raised the possibility that R-Ras may function as a positive regulator of cell proliferation and transformation in the breast. To understand the possible role of R-Ras in breast epithelial carcinogenesis, the expression and activation of R-Ras were detected in each of 69 pairs of breast cancer tissues and normal tumor-adjacent tissue samples by qRT-PCR, western blot analysis and GST pull down assay; 12 available cell lines were also subjected to western blot analysis and GST pull down assay. To further address the role of R-Ras in transformation-related phenotype formation of breast cancer cell line MCF-7 in vitro, R-Ras38V, a constitutively activated mutant of R-Ras, was transfected into MCF-7 cells, and the cell proliferation, migration and cell cycle distribution were analyzed. The results showed that although there was slight difference in the protein expression of R-Ras between the breast cancer tissues and normal tissues, the activation of R-Ras was reduced in 63.8% of the cancer tissues when compared to the normal tissue samples. In addition, the results also showed that R-Ras38V inhibited cell proliferation, migration and cell cycle progression in the presence of serum. Contradicting the positive association reported in previous studies, our results indicate that R-Ras activation may negatively regulate the transformation of breast epithelial cells, and the loss of activation of R-Ras may be involved in the carcinogenesis of breast cancer. To solve this controversy, further independent studies are needed to validate our study results.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , ras Proteins/biosynthesis , Adult , Aged , Apoptosis , Breast Neoplasms/pathology , Cell Adhesion/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Middle Aged , Signal Transduction , ras Proteins/genetics
7.
PLoS One ; 8(8): e72704, 2013.
Article in English | MEDLINE | ID: mdl-24009699

ABSTRACT

Aberrant changes in specific glycans have been shown to be associated with immunosurveillance, tumorigenesis, tumor progression and metastasis. In this study, the N-glycan profiling of membrane proteins from human breast cancer cell lines and tissues was detected using modified DNA sequencer-assisted fluorophore-assisted carbohydrate electrophoresis (DSA-FACE). The N-glycan profiles of membrane proteins were analyzed from 7 breast cancer cell lines and MCF 10A, as well as from 100 pairs of breast cancer and corresponding adjacent tissues. The results showed that, compared with the matched adjacent normal tissue samples, two biantennary N-glycans (NA2 and NA2FB) were significantly decreased (p <0.0001) in the breast cancer tissue samples, while the triantennary glycan (NA3FB) and a high-mannose glycan (M8) were dramatically increased (p = 0.001 and p <0.0001, respectively). Moreover, the alterations in these specific N-glycans occurred through the oncogenesis and progression of breast cancer. These results suggested that the modified method based on DSA-FACE is a high-throughput detection technology that is suited for analyzing cell surface N-glycans. These cell surface-specific N-glycans may be helpful in recognizing the mechanisms of tumor cell immunologic escape and could be potential targets for new breast cancer drugs.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Polysaccharides/chemistry , Adult , Age Factors , Aged , Breast Neoplasms/pathology , Cell Line , Cell Membrane/chemistry , Female , Humans , Middle Aged , Neoplasm Staging , Polysaccharides/metabolism , Proteomics/methods , Young Adult
8.
Cancer Sci ; 104(7): 817-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23521319

ABSTRACT

The low-density lipoprotein receptor-related protein 1B (LRP1B) is known as a putative tumor suppressor. The decreased expression of LRP1B has been involved in multiple primary cancers in several studies. However, its expression and function in the carcinogenesis of renal cell cancer (RCC) remain unclear. In this study, we investigated the expression of LRP1B in RCC by in situ hybridization (ISH) and real-time polymerase chain reaction (qRT-PCR). Our results indicated that LRP1B was frequently downexpressed in human RCC tissue and cell lines, which involved both epigenetic events (DNA methylation and histone deacetylation) and N-terminal deletion of LRP1B. Moreover, we testified that knockdown of LRP1B by shRNA significantly promoted anchorage-independent growth, cell migration and invasion in HEK293 cells and renal cancer cells 127 in vitro. We further found that silencing of LRP1B altered the expression of focal adhesion complex-associated proteins, and Cdc42/RhoA activities, which regulate the cytoskeleton dynamics. Taken together, these results strongly support that LRP1B may function as a tumor suppressor against renal cell cancer, and may regulate cell motility via RhoA/Cdc42 pathway and actin cytoskeleton reorganization in RCC.


Subject(s)
Actin Cytoskeleton/genetics , Carcinoma, Renal Cell/genetics , Cell Movement/genetics , Kidney Neoplasms/genetics , Receptors, LDL/genetics , cdc42 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , Acetylation , Actin Cytoskeleton/metabolism , Aged , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , DNA Methylation/genetics , Down-Regulation , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Silencing , Genes, Tumor Suppressor , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Neoplasm Invasiveness , Receptors, LDL/metabolism , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Yi Chuan ; 34(1): 120-5, 2012 Jan.
Article in Chinese | MEDLINE | ID: mdl-22306881

ABSTRACT

Ordered tetrad analysis is important content in the genetics teaching. In particular, the two linkage gene mapping is not only a key point, but also a difficult one. How to explain the content better is a hard nut for the many genetics teachers or editors of the teaching material to crack. Here, based on teaching practice of many years we summarized several key problems, which are difficult to understand by students and frequently neglected by the teachers and the editors of genetics. Furthermore, we deeply analyzed these problems and presented some opinions and suggestions relative to them so as to provide a reference for the teachers of genetics and the editors of teaching materials.


Subject(s)
Chromosome Mapping , Genetic Linkage , Genetics, Microbial/education , Neurospora crassa/genetics , China , Humans , Students
10.
J Biomed Sci ; 18: 58, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849022

ABSTRACT

BACKGROUND: Rab GTPases function as modulators in intracellular transport. Rab5a, a member of the Rab subfamily of small GTPases, is an important regulator of vesicle traffic from the plasma membrane to early endosomes. Recent findings have reported that Rab5a gene was involved in the progression of cancer. In the present study, we investigated the effect of Rab5a on cervical cancer invasion and metastasis and the molecular mechanism underlying the involvement of Rab5a. METHODS: Rab5a expression was assessed by immunohistochemical analysis on a cervical cancer tissue microarray. RNA interference (RNAi) was performed to knock down the endogenous expression of Rab5a gene in HeLa and SiHa cells. Cell motility was evaluated using invasion assay and wound migration assay in vitro. The expression levels of integrin-associated molecules were detected by Western blot and immunofluorescence. RESULTS: We found that Rab5a was expressed at a high level in cervical cancer tissues. Silencing of Rab5a expression significantly decreased cancer cell motility and invasiveness. The down-regulation of integrin-associated focal adhesion signaling molecules was further detected in Rab5a knockdown cells. Meanwhile, active GTP-bound Rac1, Cdc42, and RhoA were also down-regulated, accompanied with the reduction in the number and size of filopodia and lamellipodia. CONCLUSIONS: Taken together, these data suggest that Rab5a functions in regulating the invasion phenotype, and we propose that this regulation may be via integrin-mediated signaling pathway in cervical cancer cells.


Subject(s)
Cell Movement/physiology , Integrins/metabolism , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Signal Transduction/genetics , Uterine Cervical Neoplasms/physiopathology , rab5 GTP-Binding Proteins/metabolism , Blotting, Western , Cell Movement/genetics , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques/methods , HeLa Cells , Humans , Immunohistochemistry , Microarray Analysis , Neoplasm Invasiveness/genetics , RNA Interference , Uterine Cervical Neoplasms/metabolism , rab5 GTP-Binding Proteins/genetics
11.
Mol Cells ; 31(3): 267-74, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21347699

ABSTRACT

MARVEL domain-containing 1 (MARVELD1) is a newly identified nuclear protein; however its function has not been clear until now. Here, we report that mouse MARVELD1 (mMARVELD1), which is highly conserved between mice and humans, exhibits cell cycle-dependent cellular localization. In NIH3T3 cells, MARVELD1 was observed in the nucleus and at the perinuclear region during interphase, but was localized at the mitotic spindle and midbody at metaphase, and a significant fraction of mMARVELD1 translocated to the plasma membrane during anaphase. In addition, treatment of cells with colchicine, a microtubule-depolymerizing agent, resulted in translocation of mMARVELD1 to the plasma membrane, and association of mMARVELD1 and α-tubulin was confirmed by co-immunoprecipitation. Finally, overexpression of mMARVELD1 resulted in a remarkable inhibition of cell proliferation, G1-phase arrest, and reduced cell migration. These findings indicate that mMARVELD1 is a microtubule-associated protein that plays an important role in cell cycle progression and migration.


Subject(s)
Cell Movement , G1 Phase , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Nuclear Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Tubulin/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Consensus Sequence , Membrane Proteins , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle, Skeletal/metabolism , NIH 3T3 Cells , Protein Transport , Sequence Homology, Amino Acid
12.
Cancer Lett ; 282(1): 77-86, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19364627

ABSTRACT

MARVELD1 (MARVEL domain-containing 1) is a member of MARVEL domain-containing proteins and located on human chromosome 10q24.2. MARVELD1 has no significant similarity with other members of MARVEL domain family at amino acid level. Gene expression arrays demonstrated that MARVELD1 is widely expressed in normal human tissues and is down-regulated in primary multiple tumors derived from ovary, vulva, uterus, cervix, breast, testis, kidney, bladder and liver. The down-regulation of MARVELD1 was further identified by real-time PCR and immunohistochemical staining in primary breast cancer. In addition, we identified the reduced expression of MARVELD1 is owing to DNA methylation and could be reversed by pharmacologic demethylation. Finally, our results showed that MARVELD1 protein is located in nucleus instead of cell membrane.


Subject(s)
Chromosomes, Human, Pair 10 , DNA Methylation , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Nuclear Proteins/genetics , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chromosome Mapping , DNA Primers , Down-Regulation , Female , Gene Silencing , Humans , Liver Neoplasms/genetics , Male , Membrane Proteins/metabolism , Microtubule-Associated Proteins , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Testicular Neoplasms/genetics
13.
Cell Signal ; 19(7): 1575-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17382517

ABSTRACT

A cDNA encoding a RalGDS-related protein, Rgl3, was isolated by yeast two-hybrid screening using a small G-protein, Rap1, as a bait. Rgl3 mRNA is commonly detectable in several visceral organs (e.g. kidney, heart, liver, and lung) in the mouse and human. The Rgl3 protein mainly localizes in the cytoplasm when expressed in fibroblasts. Yeast two-hybrid assay indicated that Rgl3 could interact with Rap1, Rap2, H-Ras, N-Ras, and R-Ras but failed to interact efficiently with Ral and Rho. Interestingly, Rgl3 was found to affect cell morphology in two assay systems in culture. First, Rgl3 suppressed cell-spreading induced by Rap1, R-Ras, or C3G-CAAX (a membrane-targeted Rap/R-Ras activator) in HEK-293 cells. Second, Rgl3 enhanced the focus-formation induced by oncogenic H-Ras and N-Ras mutants in NIH3T3 cells. Moreover, we identified profilin II as a potential binding partner for Rgl3 by yeast two-hybrid screening. This interaction requires the characteristic proline cluster in the Rgl3 amino-terminal domain. Profilin II and Rgl3 co-operated in enhancing the N-Ras-induced focus-formation. These findings raise the possibility that Rgl3 mediates interaction between Ras/Rap-family proteins and profilin II, an important activator of actin polymerization.


Subject(s)
Profilins/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , rap1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Transformed , Cell Movement , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subcellular Fractions/metabolism , ral Guanine Nucleotide Exchange Factor/chemistry , ral Guanine Nucleotide Exchange Factor/genetics , ras Proteins/metabolism
14.
Cancer Res ; 64(24): 9062-9, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604273

ABSTRACT

The antiangiogenic function of the tissue inhibitors of metalloproteinases (TIMPs) has been attributed to their matrix metalloproteinase inhibitory activity. Here we demonstrate that TIMP-1 but not Ala+TIMP-1 inhibits both basal and vascular endothelial growth factor (VEGF)-stimulated migration of human microvascular endothelial cells (hMVECs), suggesting that this effect is dependent on direct inhibition of matrix metalloproteinase (MMP) activity. In contrast, TIMP-2 and mutant Ala+TIMP-2, which is devoid of MMP inhibitory activity, block hMVEC migration in response to VEGF-A stimulation. TIMP-2 and Ala+TIMP-2 also suppress basal hMVEC migration via a time-dependent mechanism mediated by enhanced expression of RECK, a membrane-anchored MMP inhibitor, which, in turn, inhibits cell migration. TIMP-2 treatment of hMVECs increases the association of Crk with C3G, resulting in enhanced Rap1 activation. hMVECs stably expressing Rap1 have increased RECK expression and display reduced cell migration compared with those expressing inactive Rap1(38N). RECK-null murine embryo fibroblasts fail to demonstrate TIMP-2-mediated decrease in cell migration despite activation of Rap1. TIMP-2-induced RECK decreases cell-associated MMP activity. Anti-RECK antibody increases MMP activity and reverses the TIMP-2-mediated reduction in cell migration. The effects of TIMP-2 on RECK expression and cell migration were confirmed in A2058 melanoma cells. These results suggest that TIMP-2 can inhibit cell migration via several distinct mechanisms. First, TIMP-2 can inhibit cell migration after VEGF stimulation by direct inhibition of MMP activity induced in response to VEGF stimulation. Secondly, TIMP-2 can disrupt VEGF signaling required for initiation of hMVEC migration. Third, TIMP-2 can enhance expression of RECK via Rap1 signaling resulting in an indirect, time-dependent inhibition of endothelial cell migration.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Membrane Glycoproteins/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Cell Line, Tumor , Endothelium, Vascular/cytology , GPI-Linked Proteins , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Glycoproteins/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , rap1 GTP-Binding Proteins/metabolism
15.
Oncogene ; 23(54): 8711-9, 2004 Nov 18.
Article in English | MEDLINE | ID: mdl-15480424

ABSTRACT

The mutant of Ras protein with serine to asparagine mutation at residue 17 (Ras-17N) is known to interfere with the signaling function of the wild-type Ras protein by sequestering its guanine-nucleotide exchange factors (GEFs). The similar mutant of another Ras family protein Rap1 (Rap1-17N) fails to effectively interfere with the interaction between the wild-type Rap1 and one of its GEFs, C3G, in vitro. In the present study, we have attempted to isolate Rap1 mutants with increased affinity for C3G using random mutagenesis and yeast two-hybrid screening. Based on the pattern of mutations found among these mutants, we could design a potent C3G-binder, named Rap1-AGE, harboring mutations in three sites (17A, 29G, and 117E). The association of Rap1-AGE with C3G in the cells was confirmed by co-immunoprecipitation experiments. The ability of Rap1-AGE to inhibit C3G-mediated Rap1-activation and cell spreading was also demonstrated. On the other hand, Rap1 activation mediated by two other GEFs, Epac and smgGDS, was not inhibited by Rap1-AGE. These results suggest that Rap1-AGE acts as a dominant interfering factor against C3G and serves as a useful tool in analyzing the roles of C3G-Rap1 signaling pathway in various biological processes.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide-Releasing Factor 2/metabolism , rap1 GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Cytoskeletal Proteins , DNA Primers , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Mutagenesis , NIH 3T3 Cells , Protein Binding , Signal Transduction , Two-Hybrid System Techniques , rap1 GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...