201
|
Takeda S, Fujimoto A, Yamauchi E, Hiyoshi M, Kido H, Watanabe T, Kaibuchi K, Ohta T, Konishi H. Role of a tyrosine phosphorylation of SMG-9 in binding of SMG-9 to IQGAP and the NMD complex. Biochem Biophys Res Commun 2011; 410:29-33. [PMID: 21640080 DOI: 10.1016/j.bbrc.2011.05.099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Accepted: 05/17/2011] [Indexed: 12/11/2022]
Abstract
SMG-9 is a component of the NMD complex, a heterotetramer that also includes SMG-1 and SMG-8 in the complex. SMG-9 was also originally identified as a tyrosine-phosphorylated protein but the role of the phosphorylation is not yet known. In this study, we determined that IQGAP protein, an actin cytoskeleton modifier acts as a binding partner with SMG-9 and this binding is regulated by phosphorylation of SMG-9 at Tyr-41. SMG-9 is co-localized with IQGAP1 as a part of the process of actin enrichment in non-stimulated cells, but not in the EGF-stimulated cells. Furthermore, an increase in the ability of SMG-9 to bind to SMG-8 occurs in response to EGF stimulation. These results suggest that tyrosine phosphorylation of SMG-9 may play a role in the formation of the NMD complex in the cells stimulated by the growth factor.
Collapse
Affiliation(s)
- Saori Takeda
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, Shobara, Hiroshima 727-0023, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Mizuno K, Ramalho JS, Izumi T. Exophilin8 transiently clusters insulin granules at the actin-rich cell cortex prior to exocytosis. Mol Biol Cell 2011; 22:1716-26. [PMID: 21441305 PMCID: PMC3093323 DOI: 10.1091/mbc.e10-05-0404] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 02/09/2011] [Accepted: 03/11/2011] [Indexed: 12/16/2022] Open
Abstract
Exophilin8/MyRIP/Slac2-c is an effector protein of the small GTPase Rab27a and is specifically localized on retinal melanosomes and secretory granules. We investigated the role of exophilin8 in insulin granule trafficking. Exogenous expression of exophilin8 in pancreatic β cells or their cell line, MIN6, polarized (exophilin8-positive) insulin granules at the cell corners, where both cortical actin and the microtubule plus-end-binding protein, EB1, were present. Mutation analyses indicated that the ability of exophilin8 to act as a linker between Rab27a and myosin Va is essential for its granule-clustering activity. Moreover, exophilin8 and exophilin8-associated insulin granules were markedly stable and immobile. Total internal reflection fluorescence microscopy indicated that exophilin8 restricts the motion of insulin granules at a region deeper than that where another Rab27a effector, granuphilin, accumulates docked granules directly attached to the plasma membrane. However, the exophilin8-induced immobility of insulin granules was eliminated upon secretagogue stimulation and did not inhibit evoked exocytosis. Furthermore, exophilin8 depletion prevents insulin granules from being transported close to the plasma membrane and inhibits their fusion. These findings indicate that exophilin8 transiently traps insulin granules into the cortical actin network close to the microtubule plus-ends and supplies them for release during the stimulation.
Collapse
Affiliation(s)
- Kouichi Mizuno
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371–8512, Japan
| | - José S. Ramalho
- CEDOC, Faculty of Medical Sciences, NOVA University, 1169–056 Lisbon, Portugal
| | - Tetsuro Izumi
- Laboratory of Molecular Endocrinology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371–8512, Japan
| |
Collapse
|
203
|
Ma Y, Wang B, Li W, Liu X, Wang J, Ding T, Zhang J, Ying G, Fu L, Gu F. Intersectin1-s is involved in migration and invasion of human glioma cells. J Neurosci Res 2011; 89:1079-90. [PMID: 21503949 DOI: 10.1002/jnr.22616] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/24/2010] [Accepted: 01/11/2011] [Indexed: 11/12/2022]
Abstract
Malignant gliomas have a tendency to invade diffusely into surrounding healthy brain tissues, thereby precluding their successful surgical removal. Intersectin1 (ITSN1) as a molecular linker in the central nervous system is well known as an important regulator of endocytosis and exocytosis. ITSN1 has two isoforms: ITSN1-l and ITSN1-s. In this study, we show that siRNA-mediated down regulation of ITSN1-s inhibited migration and invasion of glioma cells. In addition, we demonstrate the possible mechanisms by which ITSN1-s functions in migration and invasion. Several key proteins, including cofilin, LIMK, PAK, FAK, integrin β1, and MMP-9, which are critical for cells migration and invasion, were probably involved in ITSN1-s signaling pathways. These results suggest that ITSN1-s contributes to glioma cells migration and invasion by regulating the formation of cytoskeleton, influencing adhesion and increasing expression of MMP-9. Our results indicate that ITSN1-s is a critical factor in gliomas invasion and identify that ITSN1-s is a new potentially antiinvasion target for therapeutic intervention in gliomas.
Collapse
Affiliation(s)
- Yongjie Ma
- Central Laboratory of Oncology Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Dixon MJ, Gray A, Boisvert FM, Agacan M, Morrice NA, Gourlay R, Leslie NR, Downes CP, Batty IH. A screen for novel phosphoinositide 3-kinase effector proteins. Mol Cell Proteomics 2011; 10:M110.003178. [PMID: 21263009 PMCID: PMC3069342 DOI: 10.1074/mcp.m110.003178] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 12/10/2010] [Indexed: 11/06/2022] Open
Abstract
Class I phosphoinositide 3-kinases exert important cellular effects through their two primary lipid products, phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P(2)). As few molecular targets for PtdIns(3,4)P(2) have yet been identified, a screen for PI 3-kinase-responsive proteins that is selective for these is described. This features a tertiary approach incorporating a unique, primary recruitment of target proteins in intact cells to membranes selectively enriched in PtdIns(3,4)P(2). A secondary purification of these proteins, optimized using tandem pleckstrin homology domain containing protein-1 (TAPP-1), an established PtdIns(3,4)P(2) selective ligand, yields a fraction enriched in proteins of potentially similar lipid binding character that are identified by liquid chromatography-tandem MS. Thirdly, this approach is coupled to stable isotope labeling with amino acids in cell culture using differential isotope labeling of cells stimulated in the absence and presence of the PI 3-kinase inhibitor wortmannin. This provides a ratio-metric readout that distinguishes authentically responsive components from copurifying background proteins. Enriched fractions thus obtained from astrocytoma cells revealed a subset of proteins that exhibited ratios indicative of their initial, cellular responsiveness to PI 3-kinase activation. The inclusion among these of tandem pleckstrin homology domain containing protein-1, three isoforms of Akt, switch associated protein-70, early endosome antigen-1 and of additional proteins expressing recognized lipid binding domains demonstrates the utility of this strategy and lends credibility to the novel candidate proteins identified. The latter encompass a broad set of proteins that include the gene product of TBC1D2A, a putative Rab guanine nucleotide triphosphatase activating protein (GAP) and IQ motif containing GAP1, a potential tumor promoter. A sequence comparison of the former protein indicates the presence of a pleckstrin homology domain whose lipid binding character remains to be established. IQ motif containing GAP1 lacks known lipid interacting components and a preliminary analysis here indicates that this may exemplify a novel class of atypical phosphoinositide (aPI) binding domain.
Collapse
Affiliation(s)
| | | | | | - Mark Agacan
- ‖The Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow St., Dundee, DD1 5EH, Scotland, UK
| | | | | | | | | | | |
Collapse
|
205
|
Logue JS, Whiting JL, Tunquist B, Langeberg LK, Scott JD. Anchored protein kinase A recruitment of active Rac GTPase. J Biol Chem 2011; 286:22113-21. [PMID: 21460214 PMCID: PMC3121355 DOI: 10.1074/jbc.m111.232660] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Protein kinase A-anchoring proteins (AKAPs) influence fundamental cellular processes by directing the cAMP-dependent protein kinase (PKA) toward its intended substrates. In this report we describe the identification and characterization of a ternary complex of AKAP220, the PKA holoenzyme, and the IQ domain GTPase-activating protein 2 isoform (IQGAP2) that is enriched at cortical regions of the cell. Formation of an IQGAP2-AKAP220 core complex initiates a subsequent phase of protein recruitment that includes the small GTPase Rac. Biochemical and molecular biology approaches reveal that PKA phosphorylation of Thr-716 on IQGAP2 enhances association with the active form of the Rac GTPase. Cell-based experiments indicate that overexpression of an IQGAP2 phosphomimetic mutant (IQGAP2 T716D) enhances the formation of actin-rich membrane ruffles at the periphery of HEK 293 cells. In contrast, expression of a nonphosphorylatable IQGAP2 T716A mutant or gene silencing of AKAP220 suppresses formation of membrane ruffles. These findings imply that IQGAP2 and AKAP220 act synergistically to sustain PKA-mediated recruitment of effectors such as Rac GTPases that impact the actin cytoskeleton.
Collapse
Affiliation(s)
- Jeremy S Logue
- Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | | | | | | | |
Collapse
|
206
|
Kim H, White CD, Sacks DB. IQGAP1 in microbial pathogenesis: Targeting the actin cytoskeleton. FEBS Lett 2011; 585:723-9. [PMID: 21295032 PMCID: PMC3085995 DOI: 10.1016/j.febslet.2011.01.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 11/18/2022]
Abstract
Microbial pathogens cause widespread morbidity and mortality. Central to the pathogens' virulence is manipulation of the host cell's cytoskeleton, which facilitates microbial invasion, multiplication, and avoidance of the innate immune response. IQGAP1 is a ubiquitously expressed scaffold protein that integrates diverse signaling cascades. Research has shown that IQGAP1 binds to and modulates the activity of multiple proteins that participate in bacterial invasion. Here, we review data that support a role for IQGAP1 in infectious disease via its ability to regulate the actin cytoskeleton. In addition, we explore other mechanisms by which IQGAP1 may be exploited by microbial pathogens.
Collapse
Affiliation(s)
- Hugh Kim
- Department of Translational Medicine, Brigham and Women's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA 02115, USA
| | | | | |
Collapse
|
207
|
McNulty DE, Li Z, White CD, Sacks DB, Annan RS. MAPK scaffold IQGAP1 binds the EGF receptor and modulates its activation. J Biol Chem 2011; 286:15010-21. [PMID: 21349850 DOI: 10.1074/jbc.m111.227694] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cellular responses produced by EGF are mediated through the receptor (EGFR) and by various enzymes and scaffolds. Recent studies document IQGAP1 as a scaffold for the MAPK cascade, binding directly to B-Raf, MEK, and ERK and regulating their activation in response to EGF. We previously showed that EGF is unable to activate B-Raf in cells lacking IQGAP1. However, the mechanism by which IQGAP1 links B-Raf to EGFR was unknown. Here we report that endogenous EGFR and IQGAP1 co-localize and co-immunoprecipitate in cells. EGF has no effect on the association, but Ca(2+) attenuates binding. In vitro analysis demonstrated a direct association mediated through the IQ and kinase domains of IQGAP1 and EGFR, respectively. Calmodulin disrupts this interaction. Using a mass spectrometry-based assay, we show that EGF induces phosphorylation of IQGAP1 Ser(1443), a residue known to be phosphorylated by PKC. This phosphorylation is eliminated by pharmacological inhibition of either EGFR or PKC and transfection with small interfering RNA directed against the PKCα isoform. In IQGAP1-null cells, EGF-stimulated tyrosine phosphorylation of EGFR is severely attenuated. Normal levels of autophosphorylation are restored by reconstituting wild type IQGAP1 and enhanced by an IQGAP1 S1443D mutant. Collectively, these data demonstrate a functional interaction between IQGAP1 and EGFR and suggest that IQGAP1 modulates EGFR activation.
Collapse
Affiliation(s)
- Dean E McNulty
- Proteomic and Biological Mass Spectrometry Laboratory, GlaxoSmithKline, Collegeville, Pennsylvania 19426, USA
| | | | | | | | | |
Collapse
|
208
|
Secretory and endo/exocytic trafficking in invadopodia formation: The MT1-MMP paradigm. Eur J Cell Biol 2011; 90:108-14. [DOI: 10.1016/j.ejcb.2010.04.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Revised: 04/29/2010] [Accepted: 04/29/2010] [Indexed: 11/22/2022] Open
|
209
|
Gladue DP, Holinka LG, Fernandez-Sainz IJ, Prarat MV, O'Donnell V, Vepkhvadze NG, Lu Z, Risatti GR, Borca MV. Interaction between Core protein of classical swine fever virus with cellular IQGAP1 protein appears essential for virulence in swine. Virology 2011; 412:68-74. [PMID: 21262517 DOI: 10.1016/j.virol.2010.12.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/28/2010] [Accepted: 12/30/2010] [Indexed: 02/08/2023]
Abstract
Here we show that IQGAP1, a cellular protein that plays a pivotal role as a regulator of the cytoskeleton interacts with Classical Swine Fever Virus (CSFV) Core protein. Sequence analyses identified residues within CSFV Core protein (designated as areas I, II, III and IV) that maintain homology to regions within the matrix protein of Moloney Murine Leukemia Virus (MMLV) that mediate binding to IQGAP1 [EMBO J, 2006 25:2155]. Alanine-substitution within Core regions I, II, III and IV identified residues that specifically mediate the Core-IQGAP1 interaction. Recombinant CSFV viruses harboring alanine substitutions at residues (207)ATI(209) (I), (210)VVE(212) (II), (213)GVK(215) (III), or (232)GLYHN(236) (IV) have defective growth in primary swine macrophage cultures. In vivo, substitutions of residues in areas I and III yielded viruses that were completely attenuated in swine. These data shows that the interaction of Core with an integral component of cytoskeletal regulation plays a role in the CSFV cycle.
Collapse
Affiliation(s)
- D P Gladue
- Plum Island Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Greenport, NY 11944, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Johnson M, Sharma M, Brocardo MG, Henderson BR. IQGAP1 translocates to the nucleus in early S-phase and contributes to cell cycle progression after DNA replication arrest. Int J Biochem Cell Biol 2011; 43:65-73. [DOI: 10.1016/j.biocel.2010.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Accepted: 09/20/2010] [Indexed: 01/03/2023]
|
211
|
Heil A, Nazmi AR, Koltzscher M, Poeter M, Austermann J, Assard N, Baudier J, Kaibuchi K, Gerke V. S100P is a novel interaction partner and regulator of IQGAP1. J Biol Chem 2010; 286:7227-38. [PMID: 21177863 DOI: 10.1074/jbc.m110.135095] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Ca(2+)-binding proteins of the S100 family participate in intracellular Ca(2+) signaling by binding to and regulating specific cellular targets in their Ca(2+)-loaded conformation. Because the information on specific cellular targets of different S100 proteins is still limited, we developed an affinity approach that selects for protein targets only binding to the physiologically active dimer of an S100 protein. Using this approach, we here identify IQGAP1 as a novel and dimer-specific target of S100P, a member of the S100 family enriched in the cortical cytoskeleton. The interaction between S100P and IQGAP1 is strictly Ca(2+)-dependent and characterized by a dissociation constant of 0.2 μM. Binding occurs primarily through the IQ domain of IQGAP1 and the first EF hand loop of S100P, thus representing a novel structural principle of S100-target protein interactions. Upon cell stimulation, S100P and IQGAP1 co-localize at or in close proximity to the plasma membrane, and complex formation can be linked to altered signal transduction properties of IQGAP1. Specifically, the EGF-induced tyrosine phosphorylation of IQGAP1 that is thought to function in assembling signaling intermediates at IQGAP1 scaffolds in the subplasmalemmal region is markedly reduced in cells overexpressing S100P but not in cells expressing an S100P mutant deficient in IQGAP1 binding. Furthermore, B-Raf binding to IQGAP1 and MEK1/2 activation occurring downstream of IQGAP1 in EGF-triggered signaling cascades are compromised at elevated S100P levels. Thus, S100P is a novel Ca(2+)-dependent regulator of IQGAP1 that can down-regulate the function of IQGAP1 as a signaling intermediate by direct interaction.
Collapse
Affiliation(s)
- Annika Heil
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Strasse 56, D-48149 Muenster, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Rho GTPases and exocytosis: what are the molecular links? Semin Cell Dev Biol 2010; 22:27-32. [PMID: 21145407 DOI: 10.1016/j.semcdb.2010.12.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 12/01/2010] [Accepted: 12/01/2010] [Indexed: 02/08/2023]
Abstract
Delivery of proteins or lipids to the plasma membrane or into the extracellular space occurs through exocytosis, a process that requires tethering, docking, priming and fusion of vesicles, as well as F-actin rearrangements in response to specific extracellular cues. GTPases of the Rho family have been implicated as important regulators of exocytosis, but how Rho proteins control this process is an open question. In this review, we focus on molecular connections that drive Rho-dependent exocytosis in polarized and regulated exocytosis. Specifically, we present data showing that Rho proteins interaction with the exocyst complex and IQGAP mediates polarized exocytosis, whereas interaction with actin-binding proteins like N-WASP mediates regulated exocytosis.
Collapse
|
213
|
Skandalis SS, Kozlova I, Engström U, Hellman U, Heldin P. Proteomic identification of CD44 interacting proteins. IUBMB Life 2010; 62:833-40. [DOI: 10.1002/iub.392] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
214
|
Lee MR, Kim JS, Kim KS. miR-124a is important for migratory cell fate transition during gastrulation of human embryonic stem cells. Stem Cells 2010; 28:1550-9. [PMID: 20665740 DOI: 10.1002/stem.490] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Precise control of gene expression is of paramount importance for proper embryonic development. Although a number of microRNAs (miRNAs) has been implicated in fine-tuning mRNA translation during development, their exact roles for gastrulation, particularly in connection with functional targets, have yet to be clarified, with regard to stage-specific cell migration to form three embryonic germ layers. We found that miR-124a is expressed in human embryonic stem cells (hESC), but is gradually downregulated during embryoid body (EB) formation in vitro. We also provide evidence that SLUG and IQGAP1, which modulates rearrangement of the migratory cytoskeleton, are specific targets for miR-124a during EB formation. Furthermore, we show that the beginning of cell migration, a hallmark event in gastrulation, is tightly coupled with downregulation of miR-124a during EB formation and induction of SLUG and IQGAP1. Overexpressed miR-124a in hESC reduced expression of SLUG and IQGAP1 and blocked migratory cell behavior in EB. An expression level of MIXL1, associated with gastulation process, was also inversely correlated with expression of miR-124a. Taken together, our results strongly suggest that miR-124a may play an active role in inhibiting hESCs from differentiation into EB by downregulating expression of SLUG and IQGAP1, thereby maintaining stemness.
Collapse
Affiliation(s)
- Man Ryul Lee
- Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul, Korea
| | | | | |
Collapse
|
215
|
White CD, Khurana H, Gnatenko DV, Li Z, Odze RD, Sacks DB, Schmidt VA. IQGAP1 and IQGAP2 are reciprocally altered in hepatocellular carcinoma. BMC Gastroenterol 2010; 10:125. [PMID: 20977743 PMCID: PMC2988069 DOI: 10.1186/1471-230x-10-125] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2010] [Accepted: 10/26/2010] [Indexed: 01/16/2023] Open
Abstract
Background IQGAP1 and IQGAP2 are homologous members of the IQGAP family of scaffold proteins. Accumulating evidence implicates IQGAPs in tumorigenesis. We recently reported that IQGAP2 deficiency leads to the development of hepatocellular carcinoma (HCC) in mice. In the current study we extend these findings, and investigate IQGAP1 and IQGAP2 expression in human HCC. Methods IQGAP1 and IQGAP2 protein expression was assessed by Western blotting and immunohistochemistry. IQGAP mRNA was measured by quantitative RT-PCR. The methylation status of the Iqgap2 promoter was determined by pyrosequencing of bisulfite-treated genomic DNA. Results IQGAP1 and IQGAP2 expression was reciprocally altered in 6/6 liver cancer cell lines. Similarly, immunohistochemical staining of 82 HCC samples showed that IQGAP2 protein expression was reduced in 64/82 (78.0%), while IQGAP1 was present in 69/82 (84.1%). No IQGAP1 staining was detected in 23/28 (82.1%) normal livers, 4/4 (100.0%) hepatic adenomas and 23/23 (100.0%) cirrhosis cases, while IQGAP2 was increased in 22/28 (78.6%), 4/4 (100.0%) and 23/23 (100.0%), respectively. Although the Iqgap2 promoter was not hypermethylated in HCC at any of the 25 CpG sites studied (N = 17), IQGAP2 mRNA levels were significantly lower in HCC specimens (N = 23) than normal livers (N = 6). Conclusions We conclude that increased IQGAP1 and/or decreased IQGAP2 contribute to the pathogenesis of human HCC. Furthermore, downregulation of IQGAP2 in HCC occurs independently of hypermethylation of the Iqgap2 promoter. Immunostaining of IQGAP1 and IQGAP2 may aid in the diagnosis of HCC, and their pharmacologic modulation may represent a novel therapeutic strategy for the treatment of liver cancer.
Collapse
Affiliation(s)
- Colin D White
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
216
|
Urao N, Razvi M, Oshikawa J, McKinney RD, Chavda R, Bahou WF, Fukai T, Ushio-Fukai M. IQGAP1 is involved in post-ischemic neovascularization by regulating angiogenesis and macrophage infiltration. PLoS One 2010; 5:e13440. [PMID: 20976168 PMCID: PMC2955540 DOI: 10.1371/journal.pone.0013440] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Accepted: 09/24/2010] [Indexed: 11/18/2022] Open
Abstract
Background Neovascularization is an important repair mechanism in response to ischemic injury and is dependent on inflammation, angiogenesis and reactive oxygen species (ROS). IQGAP1, an actin-binding scaffold protein, is a key regulator for actin cytoskeleton and motility. We previously demonstrated that IQGAP1 mediates vascular endothelial growth factor (VEGF)-induced ROS production and migration of cultured endothelial cells (ECs); however, its role in post-ischemic neovascularization is unknown. Methodology/Principal Findings Ischemia was induced by left femoral artery ligation, which resulted in increased IQGAP1 expression in Mac3+ macrophages and CD31+ capillary-like ECs in ischemic legs. Mice lacking IQGAP1 exhibited a significant reduction in the post-ischemic neovascularization as evaluated by laser Doppler blood flow, capillary density and α-actin positive arterioles. Furthermore, IQGAP1−/− mice showed a decrease in macrophage infiltration and ROS production in ischemic muscles, leading to impaired muscle regeneration and increased necrosis and fibrosis. The numbers of bone marrow (BM)-derived cells in the peripheral blood were not affected in these knockout mice. BM transplantation revealed that IQGAP1 expressed in both BM-derived cells and tissue resident cells, such as ECs, is required for post-ischemic neovascularization. Moreover, thioglycollate-induced peritoneal macrophage recruitment and ROS production were inhibited in IQGAP1−/− mice. In vitro, IQGAP1−/− BM-derived macrophages showed inhibition of migration and adhesion capacity, which may explain the defective macrophage recruitment into the ischemic tissue in IQGAP1−/− mice. Conclusions/Significance IQGAP1 plays a key role in post-ischemic neovascularization by regulating, not only, ECs-mediated angiogenesis but also macrophage infiltration as well as ROS production. Thus, IQGAP1 is a potential therapeutic target for inflammation- and angiogenesis-dependent ischemic cardiovascular diseases.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Masooma Razvi
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jin Oshikawa
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ronald D. McKinney
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Rupal Chavda
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Wadie F. Bahou
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Tohru Fukai
- Departments of Medicine and Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Masuko Ushio-Fukai
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
217
|
Watanabe T, Sato K, Kaibuchi K. Cadherin-mediated intercellular adhesion and signaling cascades involving small GTPases. Cold Spring Harb Perspect Biol 2010; 1:a003020. [PMID: 20066109 DOI: 10.1101/cshperspect.a003020] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epithelia form physical barriers that separate the internal milieu of the body from its external environment. The biogenesis of functional epithelia requires the precise coordination of many cellular processes. One of the key events in epithelial biogenesis is the establishment of cadherin-dependent cell-cell contacts, which initiate morphological changes and the formation of other adhesive structures. Cadherin-mediated adhesions generate intracellular signals that control cytoskeletal reorganization, polarity, and vesicle trafficking. Among such signaling pathways, those involving small GTPases play critical roles in epithelial biogenesis. Assembly of E-cadherin activates several small GTPases and, in turn, the activated small GTPases control the effects of E-cadherin-mediated adhesions on epithelial biogenesis. Here, we focus on small GTPase signaling at E-cadherin-mediated epithelial junctions.
Collapse
Affiliation(s)
- Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | | | | |
Collapse
|
218
|
Romero AM, Esteban-Pretel G, Marín MP, Ponsoda X, Ballestín R, Canales JJ, Renau-Piqueras J. Chronic ethanol exposure alters the levels, assembly, and cellular organization of the actin cytoskeleton and microtubules in hippocampal neurons in primary culture. Toxicol Sci 2010; 118:602-12. [PMID: 20829428 DOI: 10.1093/toxsci/kfq260] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The organization and dynamics of microtubules (MTs) and the actin cytoskeleton are critical for the correct development and functions of neurons, including intracellular traffic and signaling. In vitro ethanol exposure impairs endocytosis, exocytosis, and nucleocytoplasmic traffic in astrocytes and alters endocytosis in cultured neurons. In astrocytes, these effects relate to changes in the organization and/or function of MTs and the actin cytoskeleton. To evaluate this possibility in hippocampal cultured neurons, we analyzed if chronic ethanol exposure affects the levels, assembly, and cellular organization of both cytoskeleton elements and the possible underlying mechanisms of these effects by morphological and biochemical methods. In the experiments described below, we provide the first evidence that chronic alcohol exposure decreases the amount of both filamentous actin and polymerized tubulin in neurons and that the number of MTs in dendrites lowers in treated cells. Alcohol also diminishes the MT-associated protein-2 levels, which mainly localizes in the somatodendritic compartment in neurons. Ethanol decreases the levels of total Rac, Cdc42, and RhoA, three small guanosine triphosphatases (GTPases) involved in the organization and dynamics of the actin cytoskeleton and MTs. Yet when alcohol decreases the levels of the active forms (GTP bound) of Rac1 and Cdc42, it does not affect the active form of RhoA. We also investigated the levels of several effector and regulator molecules of these GTPases to find that alcohol induces heterogeneous results. In conclusion, our results show that MT, actin cytoskeleton organization, and Rho GTPase signaling pathways are targets for the toxic effects of ethanol in neurons.
Collapse
Affiliation(s)
- Ana M Romero
- Section of Biología y Patología Celular, Centro Investigación, Hospital La Fe, E-46009 Valencia, Spain
| | | | | | | | | | | | | |
Collapse
|
219
|
Zhou J, Liang S, Fang L, Chen L, Tang M, Xu Y, Fu A, Yang J, Wei Y. Quantitative proteomic analysis of HepG2 cells treated with quercetin suggests IQGAP1 involved in quercetin-induced regulation of cell proliferation and migration. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2010; 13:93-103. [PMID: 19207037 DOI: 10.1089/omi.2008.0075] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Quercetin, a wild distributed bioflavonoid, exhibits antitumor effects on murine models by inducing apoptosis and inhibiting growth of many cancer cell lines, while proteins involved in antitumor effects at proteomic level are still unclear. In our study, we used a quantitative proteomic strategy termed stable isotope labeling by amino acids in cell culture (SILAC)-mass spectrometry (MS) to study the differential proteomic profiling of HepG2 cells treated by quercetin. In all, there were 70 changed proteins among those quantified proteins in HepG2 cells treated by 50 microM quercetin for 48 h, and 14 proteins showed significant upregulation, whereas 56 proteins were downregulated. The functional classification of changed proteins includes signaling protein, protein synthesis, cytoskeleton, metabolism, etc. Of these, Ras GTPase-activating-like protein (IQGAP1) and beta-tubulin were found to be reduced at a large degree. The migration inhibition of HepG2 cells can be induced by quercetin, and the RNA and protein expression level of IQGAP1 and beta-tubulin were respectively decreased obviously in HepG2 cells exposed to quercetin for 48 h in the scratch migration assay. The downregulated expression of IQGAP1 and beta-tubulin by quercetin treatment correlated with cell migration ability, and quercetin probably inhibits HepG2 proliferation and migration through IQGAP1 and beta-tubulin expression changes and their interactions with other proteins.
Collapse
Affiliation(s)
- Jin Zhou
- State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, West China Medical School, Sichuan University, #1 Keyuan Street 4, Gaopeng Street, Chengdu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Liang S, Fu A, Zhang Q, Tang M, Zhou J, Wei Y, Chen L. Honokiol inhibits HepG2 migration via down-regulation of IQGAP1 expression discovered by a quantitative pharmaceutical proteomic analysis. Proteomics 2010; 10:1474-83. [PMID: 20127691 DOI: 10.1002/pmic.200900649] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Honokiol (HNK), a natural small molecular product, inhibited proliferation of HepG2 cells and exhibited anti-tumor activity in nude mice. In this article, we applied a novel sensitive stable isotope labeling with amino acids in cell culture-based quantitative proteomic method and a model of nude mice to investigate the correlation between HNK and the hotspot migration molecule Ras GTPase-activating-like protein (IQGAP1). The quantitative proteomic analysis showed that IQGAP1 was 0.53-fold down-regulated under 10 microg/mL HNK exposure for 24 h on HepG2 cells. Migration ability of HepG2 cells under HNK treatment was correlated with its expression level of IQGAP1. In addition, the biochemical validation on HepG2 cells and the tumor xenograft model further demonstrated that HNK decreased the expression level of IQGAP1 and its upstream proteins Cdc42/Rac1. These data supported that HNK can modulate cell adhesion and cell migration by acting on Cdc42/Rac1 signaling via IQGAP1 interactions with its upstream Cdc42/Rac1 proteins, which is a new molecular mechanism of HNK to exert its anti-tumor activity.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, West China Medical School, Sichuan University, Chengdu, PR China
| | | | | | | | | | | | | |
Collapse
|
221
|
Klees RF, De Marco PC, Salasznyk RM, Ahuja D, Hogg M, Antoniotti S, Kamath L, Dordick JS, Plopper GE. Apocynin derivatives interrupt intracellular signaling resulting in decreased migration in breast cancer cells. J Biomed Biotechnol 2010; 2006:87246. [PMID: 16883056 PMCID: PMC1460968 DOI: 10.1155/jbb/2006/87246] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cancer cells are defined by their ability to divide uncontrollably
and metastasize to secondary sites in the body. Consequently,
tumor cell migration represents a promising target for anticancer
drug development. Using our high-throughput cell migration assay,
we have screened several classes of compounds for noncytotoxic
tumor cell migration inhibiting activity. One such compound,
apocynin (4-acetovanillone), is oxidized by peroxidases to yield a
variety of oligophenolic and quinone-type compounds that are
recognized inhibitors of NADPH oxidase and may be inhibitors of
the small G protein Rac1 that controls cell migration. We report
here that while apocynin itself is not effective, apocynin
derivatives inhibit migration of the breast cancer cell line
MDA-MB-435 at subtoxic concentrations; the migration of
nonmalignant MCF10A breast cells is unaffected. These compounds
also cause a significant rearrangement of the actin cytoskeleton,
cell rounding, and decreased levels of active Rac1 and its related
G protein Cdc42. These results may suggest a promising new route
to the development of novel anticancer therapeutics.
Collapse
Affiliation(s)
- Robert F. Klees
- Department of Biology, Rensselaer Polytechnic Institute,
Troy, NY 12180, USA
| | - Paul C. De Marco
- Department of Biology, Rensselaer Polytechnic Institute,
Troy, NY 12180, USA
| | - Roman M. Salasznyk
- Department of Biology, Rensselaer Polytechnic Institute,
Troy, NY 12180, USA
| | - Disha Ahuja
- Department of Chemical and Biological Engineering, Rensselaer
Polytechnic Institute, Troy, NY 12180, USA
| | - Michael Hogg
- Department of Chemical and Biological Engineering, Rensselaer
Polytechnic Institute, Troy, NY 12180, USA
| | - Sylvain Antoniotti
- Department of Chemical and Biological Engineering, Rensselaer
Polytechnic Institute, Troy, NY 12180, USA
| | - Lakshmi Kamath
- Life Sciences Division, Millipore Corporation,
Danvers, MA 01923, USA
| | - Jonathan S. Dordick
- Department of Chemical and Biological Engineering, Rensselaer
Polytechnic Institute, Troy, NY 12180, USA
| | - George E. Plopper
- Department of Biology, Rensselaer Polytechnic Institute,
Troy, NY 12180, USA
- *George E. Plopper:
| |
Collapse
|
222
|
Osman M. An emerging role for IQGAP1 in regulating protein traffic. ScientificWorldJournal 2010; 10:944-53. [PMID: 20495773 PMCID: PMC3217317 DOI: 10.1100/tsw.2010.85] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/14/2010] [Accepted: 04/15/2010] [Indexed: 02/07/2023] Open
Abstract
IQGAP1, an effector of CDC42p GTPase, is a widely conserved, multifunctional protein that bundles F-actin through its N-terminus and binds microtubules through its C-terminus to modulate the cell architecture. It has emerged as a potential oncogene associated with diverse human cancers. Therefore, IQGAP1 has been heavily investigated; regardless, its precise cellular function remains unclear. Work from yeast suggests that IQGAP1 plays an important role in directed cell growth, which is a conserved feature crucial to morphogenesis, division axis, and body plan determination. New evidence suggests a conserved role for IQGAP1 in protein synthesis and membrane traffic, which may help to explain the diversity of its cellular functions. Membrane traffic mediates infections by intracellular pathogens and a range of degenerative human diseases arise from dysfunctions in intracellular traffic; thus, elucidating the mechanisms of cellular traffic will be important in order to understand the basis of a wide range of inherited and acquired human diseases. Recent evidence suggests that IQGAP1 plays its role in cell growth through regulating the conserved mTOR pathway. The mTOR signaling cascade has been implicated in membrane traffic and is activated in nearly all human cancers, but clinical response to the mTOR-specific inhibitor rapamycin has been disappointing. Thus, understanding the regulators of this pathway will be crucial in order to identify predictors of rapamycin sensitivity. In this review, I discuss emerging evidence that supports a potential role of IQGAP1 in regulating membrane traffic via regulating the mTOR pathway.
Collapse
Affiliation(s)
- Mahasin Osman
- Department of Molecular Pharmacology, Physiology and Biotechnology, Division of Biology and Medicine, Alpert School of Medicine, Brown University, Providence, RI, USA.
| |
Collapse
|
223
|
Hayashi H, Nabeshima K, Aoki M, Hamasaki M, Enatsu S, Yamauchi Y, Yamashita Y, Iwasaki H. Overexpression of IQGAP1 in advanced colorectal cancer correlates with poor prognosis-critical role in tumor invasion. Int J Cancer 2010; 126:2563-74. [PMID: 19856315 DOI: 10.1002/ijc.24987] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
IQGAP1 is a multifunctional protein involved in actin cytoskeleton assembly and E-cadherin-mediated cell adhesion. We reported previously IQGAP1 overexpression in human colorectal carcinomas especially at the invasion front (IF) and that such overexpression tended to correlate with lymph node metastasis in advanced cases. Thus, in this study, we investigated the clinicopathological significance of IQGAP1 expression in 85 cases of pT2-3 colorectal carcinomas with special reference to its expression pattern and prognosis, followed by analysis of the role of IQGAP1 in cancer invasion in vitro. Quantitative reverse transcription-PCR showed significant upregulation of IQGAP1 in colorectal carcinomas compared with normal mucosa. Immunohistochemically, IQGAP1 expression pattern was classified into diffuse (20%), IF-associated (35.3%) and focal (44.7%). The diffuse pattern was associated with higher rates of distant metastasis. Patients with IQGAP1 overexpression and diffuse pattern had significantly shorter survival (p < 0.0001) than others, and the diffuse pattern was an independent predictor of poor survival by multivariate analysis. In vitro invasion assays using three human colon carcinoma cell lines showed that IQGAP1 siRNA significantly suppressed hepatocyte growth factor (HGF)-stimulated cell invasion. HGF reduced membranous localization of alpha-catenin, but did not alter localization of E-cadherin, beta-catenin and IQGAP1 in membranes. Suppression of IQGAP1 expression by siRNA did not alter membranous localization of alpha-catenin even in the presence of HGF. Our results indicate that IQGAP1 plays a critical role in colon cancer cell invasion, and therefore diffuse and high expression of IQGAP1 predicts poor prognosis in patients with colorectal carcinoma.
Collapse
Affiliation(s)
- Hiroyuki Hayashi
- Department of Pathology, Fukuoka University Hospital and School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
224
|
Spindler V, Schlegel N, Waschke J. Role of GTPases in control of microvascular permeability. Cardiovasc Res 2010; 87:243-53. [DOI: 10.1093/cvr/cvq086] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
225
|
Poincloux R, Lizárraga F, Chavrier P. Matrix invasion by tumour cells: a focus on MT1-MMP trafficking to invadopodia. J Cell Sci 2009; 122:3015-24. [PMID: 19692588 DOI: 10.1242/jcs.034561] [Citation(s) in RCA: 370] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
When migrating away from a primary tumour, cancer cells interact with and remodel the extracellular matrix (ECM). Matrix metalloproteinases (MMPs), and in particular the transmembrane MT1-MMP (also known as MMP-14), are key enzymes in tumour-cell invasion. Results from recent in vitro studies highlight that MT1-MMP is implicated both in the breaching of basement membranes by tumour cells and in cell invasion through interstitial type-I collagen tissues. Remarkably, MT1-MMP accumulates at invadopodia, which are specialized ECM-degrading membrane protrusions of invasive cells. Here we review current knowledge about MT1-MMP trafficking and its importance for the regulation of protease activity at invadopodia. In invasive cells, endocytosis of MT1-MMP by clathrin- and caveolae-dependent pathways can be counteracted by several mechanisms, which leads to protease stabilization at the cell surface and increased pericellular degradation of the matrix. Furthermore, the recent identification of cellular components that control delivery of MT1-MMP to invadopodia brings new insight into mechanisms of cancer-cell invasion and reveals potential pharmacological targets.
Collapse
Affiliation(s)
- Renaud Poincloux
- CNRS, UMR144, Membrane and Cytoskeleton Dynamics, and Institut Curie, Paris, France
| | | | | |
Collapse
|
226
|
Tsubota A, Matsumoto K, Mogushi K, Nariai K, Namiki Y, Hoshina S, Hano H, Tanaka H, Saito H, Tada N. IQGAP1 and vimentin are key regulator genes in naturally occurring hepatotumorigenesis induced by oxidative stress. Carcinogenesis 2009; 31:504-11. [PMID: 20015863 DOI: 10.1093/carcin/bgp313] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
To identify key genes involved in the complex multistep process of hepatotumorigenesis, we reduced multivariate clinicopathological variables by using the Long-Evans Cinnamon rat, a model with naturally occurring and oxidative stress-induced hepatotumorigenesis. Gene expression patterns were analyzed serially by profiling liver tissues from rats of a naive status (4 weeks old), through to those with chronic hepatitis (26 and 39 weeks old) to tumor development (67 weeks old). Of 31 099 probe sets used for microarray analysis, 87 were identified as being upregulated in a stepwise manner during disease progression and tumor development. Quantitative real-time reverse transcription-polymerase chain reaction and statistical analyses verified that IQGAP1 and vimentin mRNA expression levels increased significantly throughout hepatotumorigenesis. A hierarchical clustering algorithm showed both genes clustered together and in the same cluster group. Immunohistochemical and western blot analyses showed similar increases in protein levels of IAGAP1 and vimentin. Finally, pathway analyses using text-mining technology with more comprehensive and recent gene-gene interaction data identified IQGAP1 and vimentin as important nodes in underlying gene regulatory networks. These findings enhance our understanding of the multistep hepatotumorigenesis and identification of target molecules for novel treatments.
Collapse
Affiliation(s)
- Akihito Tsubota
- Institute of Clinical Medicine and Research, Jikei University School of Medicine, 163-1 Kashiwa-shita, Kashiwa, Chiba 277-8567, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Miller PM, Folkmann AW, Maia AR, Efimova N, Efimov A, Kaverina I. Golgi-derived CLASP-dependent microtubules control Golgi organization and polarized trafficking in motile cells. Nat Cell Biol 2009; 11:1069-80. [PMID: 19701196 PMCID: PMC2748871 DOI: 10.1038/ncb1920] [Citation(s) in RCA: 237] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 05/22/2009] [Indexed: 01/10/2023]
Abstract
Microtubules are indispensable for Golgi complex assembly and maintenance, which are integral parts of cytoplasm organization during interphase in mammalian cells. Here, we show that two discrete microtubule subsets drive two distinct, yet simultaneous, stages of Golgi assembly. In addition to the radial centrosomal microtubule array, which positions the Golgi in the centre of the cell, we have identified a role for microtubules that form at the Golgi membranes in a manner dependent on the microtubule regulators CLASPs. These Golgi-derived microtubules draw Golgi ministacks together in tangential fashion and are crucial for establishing continuity and proper morphology of the Golgi complex. We propose that specialized functions of these two microtubule arrays arise from their specific geometries. Further, we demonstrate that directional post-Golgi trafficking and cell migration depend on Golgi-associated CLASPs, suggesting that correct organization of the Golgi complex by microtubules is essential for cell polarization and motility.
Collapse
Affiliation(s)
- Paul M. Miller
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew W. Folkmann
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ana R.R. Maia
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Institute for Molecular and Cell Biology, University of Porto, Porto 4150-180, Portugal
| | - Nadia Efimova
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrey Efimov
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Now at Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
228
|
Doehn U, Hauge C, Frank SR, Jensen CJ, Duda K, Nielsen JV, Cohen MS, Johansen JV, Winther BR, Lund LR, Winther O, Taunton J, Hansen SH, Frödin M. RSK is a principal effector of the RAS-ERK pathway for eliciting a coordinate promotile/invasive gene program and phenotype in epithelial cells. Mol Cell 2009; 35:511-22. [PMID: 19716794 PMCID: PMC3784321 DOI: 10.1016/j.molcel.2009.08.002] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2008] [Revised: 05/18/2009] [Accepted: 08/07/2009] [Indexed: 10/20/2022]
Abstract
The RAS-stimulated RAF-MEK-ERK pathway confers epithelial cells with critical motile and invasive capacities during development, tissue regeneration, and carcinoma progression, often via promoting the epithelial-mesenchymal transition (EMT). Many mechanisms by which ERK exerts this control remain elusive. We demonstrate that the ERK-activated kinase RSK is necessary to induce mesenchymal motility and invasive capacities in nontransformed epithelial and carcinoma cells. RSK is sufficient to induce certain motile responses. Expression profiling analysis revealed that a primary role of RSK is to induce transcription of a potent promotile/invasive gene program by FRA1-dependent and -independent mechanisms. The program enables RSK to coordinately modulate the extracellular environment, the intracellular motility apparatus, and receptors mediating communication between these compartments to stimulate motility and invasion. These findings uncover a mechanism whereby the RAS-ERK pathway controls epithelial cell motility by identifying RSK as a key effector, from which emanate multiple highly coordinate transcription-dependent mechanisms for stimulation of motility and invasive properties.
Collapse
Affiliation(s)
- Ulrik Doehn
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Camilla Hauge
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Scott R. Frank
- GI Cell Biology Laboratory, Children’s Hospital and Harvard Medical School, 300 Longwood Ave., Boston, MA 02115, USA
| | - Claus J. Jensen
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Katarzyna Duda
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Jakob V. Nielsen
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Michael S. Cohen
- Howard Hughes Medical Institute, Dept. of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158-2280, USA
| | - Jens V. Johansen
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
- Bioinformatics Centre, University of Copenhagen
| | - Benny R. Winther
- Dept. of Clinical Biochemistry, Glostrup Hospital, 2600 Glostrup, Denmark
| | - Leif R. Lund
- The Finsen Laboratory, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Ole Winther
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
- Bioinformatics Centre, University of Copenhagen
| | - Jack Taunton
- Howard Hughes Medical Institute, Dept. of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158-2280, USA
| | - Steen H. Hansen
- GI Cell Biology Laboratory, Children’s Hospital and Harvard Medical School, 300 Longwood Ave., Boston, MA 02115, USA
| | - Morten Frödin
- Biotech Research & Innovation Centre (BRIC) and Centre for Epigenetics, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
229
|
Zhang B, Gu F, She C, Guo H, Li W, Niu R, Fu L, Zhang N, Ma Y. Reduction of Akt2 inhibits migration and invasion of glioma cells. Int J Cancer 2009; 125:585-595. [PMID: 19330838 DOI: 10.1002/ijc.24314] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malignant gliomas have a tendency to invade diffusely into surrounding healthy brain tissues, thereby precluding their successful surgical removal. The serine/threonine kinase Akt2 is well known as an important regulator of cell survival and growth. In this study, we show that siRNA-mediated depletion of Akt2 inhibited migration and invasion of glioma cells. In addition, we demonstrate the mechanisms by which Akt2 functions to promote cell migration and invasion. Phosphorylation of cofilin, a critical step of actin polymerization, and phosphorylation of Girdin, essential for the integrity of the actin cytoskeleton and cell migration, were impaired. Furthermore, epidermal growth factor-induced ACAP1 phosphorylation and integrin beta1 phosphorylation were also blocked, consistent with defects in adhesion. Thus, Akt2 regulates both cell adhesion and cytoskeleton rearrangement during migration. Decreased MMP-9 expression in Akt2 knocked-down glioma cells was subsequently confirmed by Western blotting, consistent with the decreased invasion in vitro and in vivo. These results suggest that Akt2 contributes to glioma cells migration and invasion by regulating the formation of cytoskeleton, influencing adhesion and increasing expression of MMP-9. Our immunohistochemistry results by using human gliomas tissue sections also indicated that Akt2 expression was closely related with the malignancy of gliomas. This is coincident with our in vivo and in vitro results from cell lines. All of these results indicate that Akt2 is a critical factor in gliomas invasion. This study identifies that Akt2 is a potentially antiinvasion target for therapeutic intervention in gliomas.
Collapse
Affiliation(s)
- Baogang Zhang
- Department of Core Laboratory, Tianjin Medical University, Cancer Institute and Hospital, Tianjin, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Kunimoto K, Nojima H, Yamazaki Y, Yoshikawa T, Okanoue T, Tsukita S. Involvement of IQGAP3, a regulator of Ras/ERK-related cascade, in hepatocyte proliferation in mouse liver regeneration and development. J Cell Physiol 2009; 220:621-31. [PMID: 19452445 DOI: 10.1002/jcp.21798] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The spatio-temporal regulation of hepatocyte proliferation is a critical issue in liver regeneration. Here, in normal and regenerating liver as well as in developing liver, we examined its expression/localization of IQGAP3, which was most recently reported as a Ras/Rac/Cdc42-binding proliferation factor associated with cell-cell contacts in epithelial-type cells. In parallel, the expression/localization of Rac/Cdc42-binding IQGAP1/2 was examined. IQGAP3 showed a specific expression in proliferating hepatocytes positive for the proliferating marker Ki-67, the levels of expressions of mRNAs and proteins were significantly increased in hepatocytes in liver regeneration and development. In immunofluorescence, IQGAP3 was highly enriched at cell-cell contacts of hepatocytes. IQGAP1 and IQGAP2 were exclusively expressed in Kupffer and sinusoidal endothelial cells, respectively, in normal, regenerating, and developing liver. The expression of IQGAP1, but not of IQGAP2, was increased in CCl4-induced (but not in partial hepatectomy-induced) liver regeneration. Exclusive expression/localization of IQGAP3 to hepatocytes in the liver likely reflects the specific involvement of the IQGAP3/Ras/ERK signaling cascade in hepatocyte proliferation in addition to the previously identified signaling pathways, possibly by integrating cell-cell contact-related proliferating signaling events. On the other hand, the Rac/Cdc42-binding properties of IQGAP1/2/3 may be related to the distinct modes of remodeling due to the different strategies which induced proliferation of liver cells; partial hepatectomy, CCl4 injury, or embryonic development. Thus, the functional orchestration of Ras and the Ras homologous (Rho) family proteins Rac/Cdc42 likely plays a critical role in liver regeneration and development.
Collapse
Affiliation(s)
- Koshi Kunimoto
- Laboratory of Biological Science, Graduate School of Frontier Biosciences and Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
231
|
Liu SC, Jen YM, Jiang SS, Chang JL, Hsiung CA, Wang CH, Juang JL. G(alpha)12-mediated pathway promotes invasiveness of nasopharyngeal carcinoma by modulating actin cytoskeleton reorganization. Cancer Res 2009; 69:6122-30. [PMID: 19602597 DOI: 10.1158/0008-5472.can-08-3435] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The molecular mechanisms behind the aggressiveness of nasopharyngeal carcinoma (NPC), a highly invasive and metastatic head and neck malignancy, have not been made clear. In this study investigating these mechanisms, guanine nucleotide-binding protein alpha(12) subunit (G(alpha)(12)) signaling was found by microarray analysis to be increased in primary NPC cells and NPC-derived cell lines. Using small interfering RNA to knock down G(alpha)(12) in NPC cells resulted in a reduction in cell migration and invasion as well as a reversal in fibroblastoid morphology. Using microarray analysis, we also found a reduction in expression of key actin dynamics regulators and several epithelial-to-mesenchymal transition-related genes in G(alpha)(12)-depleted NPC cells. Knocking down one of those genes, IQ motif containing GTPase activating protein 1, reduced the migration and formation of adherens junctions and reversed the fibroblastoid morphology of NPC cells, as knocking down G(alpha)(12) was found to do. Immunohistochemical analysis found NPC tumors to have significantly greater levels of G(alpha)(12) protein than the normal basal epithelial cells. Quantitative real-time PCR analysis revealed a significant correlation between G(alpha)(12) mRNA levels and NPC lymph node metastasis. Together, our findings support a model in which activation of G(alpha)(12) signaling promotes tumorigenesis and progression of NPC by modulating actin cytoskeleton reorganization and expression of epithelial-to-mesenchymal transition-related genes. =
Collapse
Affiliation(s)
- Shu-Chen Liu
- Department of Radiation Oncology, Graduate Institute of Life Sciences, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
232
|
Deplazes J, Fuchs M, Rauser S, Genth H, Lengyel E, Busch R, Luber B. Rac1 and Rho contribute to the migratory and invasive phenotype associated with somatic E-cadherin mutation. Hum Mol Genet 2009; 18:3632-44. [PMID: 19584084 DOI: 10.1093/hmg/ddp312] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Recent evidence suggests a close association between extracellular E-cadherin mutation in diffuse-type gastric carcinoma and the acquisition of a migratory phenotype of tumour cells. To characterize the cellular machinery that mediates the gain of motility of tumour cells with mutant E-cadherin, we turned to the small Rho GTPases Rac1 and Rho because they have been implicated in pathological processes including tumour cell migration and invasion. In the present study, we analyse the activity of Rac1 and Rho in relation to E-cadherin harbouring an in-frame deletion of exon 8 and prove for the first time that the mutation reduces the ability of E-cadherin to activate Rac1 and to inhibit Rho. We provide evidence that the lack of Rac1 activation observed in response to mutant E-cadherin influences the downstream signalling of Rac1, as is shown by the decrease in the binding of the Rac1 effector protein IQGAP1 to Rac1-GTP. Moreover, reduced membranous localization of p120-catenin in mutant E-cadherin expressing cells provides an explanation for the lack of negative regulation of Rho by mutant E-cadherin. Further, we show by time-lapse laser scanning microscopy and invasion assay that the enhanced motility and invasion associated with mutant E-cadherin is sensitive to the inhibition of Rac1 and Rho. Together, these findings present evidence that the mutation of E-cadherin influences Rac1 and Rho activation in opposite directions and that Rac1 and Rho are involved in the establishment of the migratory and invasive phenotype of tumour cells that have an E-cadherin mutation.
Collapse
Affiliation(s)
- Joëlle Deplazes
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Technische Universität München, Klinikum rechts der Isar, 81675 München, Germany
| | | | | | | | | | | | | |
Collapse
|
233
|
Kim D, Lokuta MA, Huttenlocher A, Beebe DJ. Selective and tunable gradient device for cell culture and chemotaxis study. LAB ON A CHIP 2009; 9:1797-800. [PMID: 19495465 PMCID: PMC2804468 DOI: 10.1039/b901613a] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
This article describes a microfluidic device for cell culture and chemotaxis studies under various temporal and spatial concentration gradients of the medium or chemoattractant. Vertical membranes formed using in situ fabrication are employed to avoid fluid flow inside the cell observation chamber. Thus, the medium and chemoattractants are primarily provided by diffusion, maintaining cell-cell communication via secreted factors. Neutrophils were used to demonstrate the capability of the device for chemotaxis research. Experiments exhibited successful migration up a concentration gradient of interleukin 8.
Collapse
Affiliation(s)
- Dongshin Kim
- Department of Mechanical Engineering, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biomedical Engineering, University of Wisconsin, 2142 Engineering Centers Building, 1550 Engineering Drive, Madison, WI, 53706, USA
| | - Mary A. Lokuta
- Department of Pediatrics, 4205 Microbial Sciences Building, 1550 Linden Dr, Madison, WI, 53706, USA
| | - Anna Huttenlocher
- Department of Pediatrics, 4205 Microbial Sciences Building, 1550 Linden Dr, Madison, WI, 53706, USA
- Department of Pharmacology, University of Wisconsin, Madison, WI, 53706 USA
| | - David J. Beebe
- Department of Mechanical Engineering, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biomedical Engineering, University of Wisconsin, 2142 Engineering Centers Building, 1550 Engineering Drive, Madison, WI, 53706, USA
| |
Collapse
|
234
|
White CD, Brown MD, Sacks DB. IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis. FEBS Lett 2009; 583:1817-24. [PMID: 19433088 PMCID: PMC2743239 DOI: 10.1016/j.febslet.2009.05.007] [Citation(s) in RCA: 247] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 04/28/2009] [Accepted: 05/02/2009] [Indexed: 12/13/2022]
Abstract
The IQGAP family comprises three proteins in humans. The best characterized is IQGAP1, which participates in protein-protein interactions and integrates diverse signaling pathways. IQGAP2 and IQGAP3 harbor all the domains identified in IQGAP1, but their biological roles are poorly defined. Proteins that bind IQGAP1 include Cdc42 and Rac1, E-cadherin, beta-catenin, calmodulin and components of the mitogen-activated protein kinase pathway, all of which are involved in cancer. Here, we summarize the biological functions of IQGAPs that may contribute to neoplasia. Additionally, we review published data which implicate IQGAPs in cancer and tumorigenesis. The cumulative evidence suggests IQGAP1 is an oncogene while IQGAP2 may be a tumor suppressor.
Collapse
Affiliation(s)
- Colin D. White
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Matthew D. Brown
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - David B. Sacks
- Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
235
|
Abstract
Reactive oxygen species (ROS) are generated in response to growth factors, cytokines, G protein-coupled receptor agonists, or shear stress, and function as signaling molecules in nonphagocytes. However, it is poorly understood how freely diffusible ROS can activate specific signaling, so-called "redox signaling." NADPH oxidases are a major source of ROS and now recognized to have specific subcellular localizations, and this targeting to specific compartments is required for localized ROS production. One important mechanism may involve the interaction of oxidase subunits with various targeting proteins localized in lamellipodial leading edge and focal adhesions/complexes. ROS are believed to inactivate protein tyrosine phosphatases, thereby establishing a positive-feedback system that promotes activation of specific redox signaling pathways involved in various functions. Additionally, ROS production may be localized through interactions of NADPH oxidase with signaling platforms associated with caveolae/lipid rafts, endosomes, and nucleus. These indicate that the specificity of ROS-mediated signal transduction may be modulated by the localization of Nox isoforms and their regulatory subunits within specific subcellular compartments. This review summarizes the recent progress on compartmentalization of redox signaling via activation of NADPH oxidase, which is implicated in cell biology and pathophysiologies.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Department of Pharmacology, Center for Lung and Vascular Biology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois 60612, USA.
| |
Collapse
|
236
|
Gigante A, Cesari E, Busilacchi A, Manzotti S, Kyriakidou K, Greco F, Di Primio R, Mattioli-Belmonte M. Collagen I membranes for tendon repair: effect of collagen fiber orientation on cell behavior. J Orthop Res 2009; 27:826-32. [PMID: 19058185 DOI: 10.1002/jor.20812] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Tendons have poor spontaneous regenerative capabilities, and complete regeneration is never achieved despite intensive remodeling. In this in vitro study, we characterized two multilamellar collagen I membranes differing in the arrangement of collagen fiber deposition (oriented vs. nonoriented) and compared their mechanical properties. Human dermal fibroblasts and tenocytes were seeded on the two membranes to evaluate the effect of fiber orientation on cell viability and cytoskeletal organization. Results demonstrate that the multilamellar collagen I membrane with oriented fibers has the better mechanical properties and affords optimum cell proliferation and adhesion. Its fiber arrangement provides an instructive pattern for cell growth and may serve to guide the alignment of cells migrating from the ends of a crushed or frayed tendon to obtain a strong, correctly structured tendon, thus providing a viable clinical option for tendon repair.
Collapse
Affiliation(s)
- Antonio Gigante
- Orthopaedics Clinic, School of Medicine, Marche Polytechnic University, Via Tronto 10/A, Ancona 60020, Italy
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Marcos S, Moreau J, Backer S, Job D, Andrieux A, Bloch-Gallego E. Tubulin tyrosination is required for the proper organization and pathfinding of the growth cone. PLoS One 2009; 4:e5405. [PMID: 19404406 PMCID: PMC2672595 DOI: 10.1371/journal.pone.0005405] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Accepted: 03/18/2009] [Indexed: 12/03/2022] Open
Abstract
Background During development, neuronal growth cones integrate diffusible and contact guidance cues that are conveyed to both actin and microtubule (MT) cytoskeletons and ensure axon outgrowth and pathfinding. Although several post-translational modifications of tubulin have been identified and despite their strong conservation among species, their physiological roles during development, especially in the nervous sytem, are still poorly understood. Methodology/Findings Here, we have dissected the role of a post-translational modification of the last amino acid of the α-tubulin on axonal growth by analyzing the phenotype of precerebellar neurons in Tubulin tyrosin ligase knock-out mice (TTL−/−) through in vivo, ex vivo and in vitro analyses. TTL−/− neurons are devoid of tyrosinated tubulin. Their pathway shows defects in vivo, ex vivo, in hindbrains open-book preparations or in vitro, in a collagen matrix. Their axons still orient toward tropic cues, but they emit supernumerary branches and their growth cones are enlarged and exhibit an emission of mis-oriented filopodia. Further analysis of the TTL−/− growth cone intracellular organization also reveals that the respective localization of actin and MT filaments is disturbed, with a decrease in the distal accumulation of Myosin IIB, as well as a concomitant Rac1 over-activation in the hindbrain. Pharmacological inhibition of Rac1 over-activation in TTL−/− neurons can rescue Myosin IIB localization. Conclusions/Significance In the growth cone, we propose that tubulin tyrosination takes part in the relative arrangement of actin and MT cytoskeletons, in the regulation of small GTPases activity, and consequently, in the proper morphogenesis, organization and pathfinding of the growth cone during development.
Collapse
Affiliation(s)
- Séverine Marcos
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- Inserm, U567, Département Génétique et Développement, Paris, France
| | - Julie Moreau
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- Inserm, U567, Département Génétique et Développement, Paris, France
| | - Stéphanie Backer
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- Inserm, U567, Département Génétique et Développement, Paris, France
| | - Didier Job
- Grenoble Institut des Neurosciences, Centre de Recherche Inserm U.836 – UJF-CEA-CHU, Bâtiment Edmond J. Safra, Université Joseph Fourier, Site Santé à La Tronche, Grenoble, France
| | - Annie Andrieux
- Grenoble Institut des Neurosciences, Centre de Recherche Inserm U.836 – UJF-CEA-CHU, Bâtiment Edmond J. Safra, Université Joseph Fourier, Site Santé à La Tronche, Grenoble, France
| | - Evelyne Bloch-Gallego
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), Paris, France
- Inserm, U567, Département Génétique et Développement, Paris, France
- * E-mail:
| |
Collapse
|
238
|
Abstract
The mitogen-activated protein kinase (MAPK) pathway allows cells to interpret external signals and respond in an appropriate way. Diverse cellular functions, ranging from differentiation and proliferation to migration and inflammation, are regulated by MAPK signalling. Therefore, cells have developed mechanisms by which this single pathway modulates numerous cellular responses from a wide range of activating factors. This specificity is achieved by several mechanisms, including temporal and spatial control of MAPK signalling components. Key to this control are protein scaffolds, which are multidomain proteins that interact with components of the MAPK cascade in order to assemble signalling complexes. Studies conducted on different scaffolds, in different biological systems, have shown that scaffolds exert substantial control over MAPK signalling, influencing the signal intensity, time course and, importantly, the cellular responses. Protein scaffolds, therefore, are integral elements to the modulation of the MAPK network in fundamental physiological processes.
Collapse
Affiliation(s)
- Matthew D. Brown
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - David B. Sacks
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
239
|
Johnson M, Sharma M, Henderson BR. IQGAP1 regulation and roles in cancer. Cell Signal 2009; 21:1471-8. [PMID: 19269319 DOI: 10.1016/j.cellsig.2009.02.023] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2009] [Accepted: 02/26/2009] [Indexed: 01/01/2023]
Abstract
IQGAP1 is a key mediator of several distinct cellular processes, in particular cytoskeletal rearrangements. Recent studies have implicated a potential role for IQGAP1 in cancer, supported by the over-expression and distinct membrane localisation of IQGAP1 observed in a range of tumours. IQGAP1 is thought to contribute to the transformed cancer cell phenotype by regulating signalling pathways involved in cell proliferation and transformation, weakening of cell:cell adhesion contacts and stimulation of cell motility and invasion. In this review we discuss these different functional and regulatory roles of IQGAP1 and its homologues in relation to their potential impact on tumourigenesis.
Collapse
Affiliation(s)
- Michael Johnson
- Westmead Institute for Cancer Research, Westmead Millennium Institute at Westmead Hospital, University of Sydney, NSW 2145, Australia
| | | | | |
Collapse
|
240
|
Kalantari F, Auguste P, Ziafazeli T, Tzimas G, Malmström L, Bioulac-Sage P, Boismenu D, Vali H, Chevet E. Proteomics analysis of liver pathological calcification suggests a role for the IQ motif containing GTPase activating protein 1 in myofibroblast function. Proteomics Clin Appl 2009; 3:307-21. [PMID: 26238749 DOI: 10.1002/prca.200780133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Indexed: 12/14/2022]
Abstract
To date the cellular and molecular mechanisms by which liver pathological calcifications occur and are regulated are poorly investigated. To study the mechanisms linked to their appearance, we performed a proteomics analysis of calcified liver samples. To this end, human liver biopsies collected in noncalcified (N), precalcified (P), and calcified (C) areas of the liver were subjected to weak ion exchange chromatography, SDS-PAGE, and LC-ESI MS/MS analyses. As we previously demonstrated that alpha-smooth muscle actin (α-SMA) expressing myofibroblasts were involved in liver pathological calcification, we performed a targeted analysis of actin cytoskeleton remodeling-related proteins. This revealed dramatic changes in protein expression patterns in the periphery of the calcified areas. More particularly, we found that IQGAP1 and IQGAP2 proteins were subjected to major expression changes. We show that IQGAP1 expression within P and C areas of the liver correlates with the high abundance of myofibroblasts and that IQGAP1 is specifically expressed in these cells. In addition, we find that IQGAP1 is part of a protein complex including β-catenin and Rac1 mainly in P and C regions of the liver. These results suggest that IQGAP1 may play a critical role in the regulation of cytoskeleton remodeling in liver myofibroblasts in response to liver injury and consequently impact on their function.
Collapse
Affiliation(s)
- Fariba Kalantari
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Patrick Auguste
- Equipe Avenir, INSERM, Bordeaux, France. .,Université Bordeaux 2, Bordeaux, France. .,Université Bordeaux 1, Talence, France.
| | - Tahereh Ziafazeli
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - George Tzimas
- Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Lars Malmström
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | | | - Daniel Boismenu
- Montreal Proteomics Network, McGill University, Montreal, Quebec, Canada
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Eric Chevet
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada. .,Equipe Avenir, INSERM, Bordeaux, France. .,Université Bordeaux 2, Bordeaux, France. .,Department of Surgery, McGill University, Montreal, Quebec, Canada. .,Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA. .,Montreal Proteomics Network, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
241
|
Yan J, Yang Y, Zhang H, King C, Kan HM, Cai Y, Yuan CX, Bloom GS, Hua X. Menin interacts with IQGAP1 to enhance intercellular adhesion of beta-cells. Oncogene 2009; 28:973-82. [PMID: 19079338 PMCID: PMC2645484 DOI: 10.1038/onc.2008.435] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 10/10/2008] [Accepted: 11/01/2008] [Indexed: 12/20/2022]
Abstract
Multiple endocrine neoplasia type 1 (MEN1) is a dominantly inherited tumor syndrome that results from the mutation of the MEN1 gene that encodes protein menin. Stable overexpression of MEN1 has been shown to partially suppress the Ras-mediated morphological changes of fibroblast cells. Little is known about the molecular mechanisms by which menin decreases the oncogenic effects on cell morphology and other phenotypes. Here we showed that ectopic expression of menin in pretumor beta-cells increases islet cell adhesion and reduces cell migration. Our further studies revealed that menin interacts with the scaffold protein, IQ motif containing GTPase activating protein 1 (IQGAP1), reduces GTP-Rac1 interaction with IQGAP1 but increases epithelial cadherin (E-cadherin)/beta-catenin interaction with IQGAP1. Consistent with an essential role for menin in regulating beta-cell adhesion in vivo, accumulations of beta-catenin and E-cadherin are reduced at cell junctions in the islets from Men1-excised mice. Together, these results define a novel menin-IQGAP1 pathway that controls cell migration and cell-cell adhesion in endocrine cells.
Collapse
Affiliation(s)
- Jizhou Yan
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
| | - Yuqing Yang
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
| | - Hui Zhang
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Catrina King
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
| | - Ho-Man Kan
- Department of Biology, University of Virginia; Charlottesville, VA 22903
| | - Ying Cai
- Department of Biology, University of Virginia; Charlottesville, VA 22903
| | - Chao-Xing Yuan
- Proteomic Facility: University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
| | - George S Bloom
- Department of Biology, University of Virginia; Charlottesville, VA 22903
- Department of Cell Biology, University of Virginia; Charlottesville, VA 22903
| | - Xianxin Hua
- Abramson Family Cancer Research Institute and Department of Cancer Biology, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, PA19014
| |
Collapse
|
242
|
Hu B, Shi B, Jarzynka MJ, Yiin JJ, D'Souza-Schorey C, Cheng SY. ADP-ribosylation factor 6 regulates glioma cell invasion through the IQ-domain GTPase-activating protein 1-Rac1-mediated pathway. Cancer Res 2009; 69:794-801. [PMID: 19155310 PMCID: PMC2633432 DOI: 10.1158/0008-5472.can-08-2110] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A common pathobiological feature of malignant gliomas is the insidious infiltration of single tumor cells into the brain parenchyma, rendering these deadly tumors virtually incurable with available therapies. In this study, we report that ADP-ribosylation factor 6 (ARF6), a Ras superfamily small GTPase, is abundantly expressed in invasive human glioma cells. Cellular depletion of ARF6 by small interfering RNA decreased Rac1 activation, impaired HGF-stimulated and serum-stimulated glioma cell migration in vitro, and markedly decreased the invasive capacity of invasive glioma in the brain. Furthermore, ectopic expression of ARF6 in glioma cells promoted cell migration via the activation of Rac1. Upon stimulation of glioma cells with HGF, we show that IQ-domain GTPase-activating protein 1 (IQGAP1) is recruited and overlaps with ARF6 at the leading edge of migrating cells. However, cellular depletion of ARF6 abrogated this recruitment of IQGAP1 and attenuated the formation of surface protrusions. ARF6 forms complexes with Rac1 and IQGAP1 in glioma cells upon HGF stimulation, and knockdown of IQGAP1 significantly inhibits ARF6-induced Rac1 activation and cell migration. Taken together, these data suggest that ARF6-mediated Rac1 activation is essential for glioma cell invasion via a signaling pathway that requires IQGAP1.
Collapse
Affiliation(s)
- Bo Hu
- Departments of Medicine and Pathology, Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | |
Collapse
|
243
|
Akhmanova A, Stehbens SJ, Yap AS. Touch, grasp, deliver and control: functional cross-talk between microtubules and cell adhesions. Traffic 2009; 10:268-74. [PMID: 19175539 DOI: 10.1111/j.1600-0854.2008.00869.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cross-talk between microtubule networks and sites of cell-matrix and cell-cell adhesion has profound impact on these structures and is essential for proper cell organization, polarization and motility. Components of adhesion sites can interact directly with microtubules or with proteins that specifically associate with microtubule plus ends and minus ends and in this way capture, stabilize or destabilize microtubules. In their turn, microtubules can serve as routes for delivery of structural and regulatory factors that control adhesion site turnover. In addition, the microtubule lattice or growing microtubule plus ends can serve as diffusional sinks that accumulate and scaffold regulatory molecules, thereby affecting their activity in the vicinity of adhesions. Combination of these mechanisms underlies the functional co-operation between microtubules and adhesion sites and defines their dynamic behavior.
Collapse
Affiliation(s)
- Anna Akhmanova
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | |
Collapse
|
244
|
Grise F, Bidaud A, Moreau V. Rho GTPases in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2009; 1795:137-51. [PMID: 19162129 DOI: 10.1016/j.bbcan.2008.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 12/21/2008] [Accepted: 12/24/2008] [Indexed: 01/05/2023]
Abstract
Rho GTPases are major regulators of signal transduction pathways and play key roles in processes including actin dynamics, cell cycle progression, cell survival and gene expression, whose deregulation may lead to tumorigenesis. A growing number of in vitro and in vivo studies using tumor-derived cell lines, primary tumors and animal cancer models strongly suggest that altered Rho GTPase signaling plays an important role in the initiation as well as in the progression of hepatocellular carcinoma (HCC), one of the deadliest human cancers in the world. These alterations can occur at the level of the GTPases themselves or of one of their regulators or effectors. The participation into the tumorigenic process can occur either through the over-expression of one of these components which presents an oncogenic activity as illustrated with RhoA and C or through the attenuation of the expression of a component presenting tumor suppressor activity as for Cdc42 or the RhoGAP, DLC-1. Consequently, these observations reflect the heterogeneity and the complexity of liver carcinogenesis. Recently, pharmacological approaches targeting Rho GTPase signaling have been used in HCC-derived models with relative success but remain to be validated in more physiologically relevant systems. Therefore, therapeutic approaches targeting Rho GTPase signaling may provide a novel alternative for anti-HCC therapy.
Collapse
Affiliation(s)
- Florence Grise
- INSERM, U889, Bordeaux, 33076 Bordeaux, France; Université Victor Segalen Bordeaux 2, Bordeaux, 33076 Bordeaux, France
| | | | | |
Collapse
|
245
|
Buss C, Müller D, Rüter C, Heusipp G, Schmidt MA. Identification and characterization of Ibe, a novel type III effector protein of A/E pathogens targeting human IQGAP1. Cell Microbiol 2009; 11:661-77. [PMID: 19134119 DOI: 10.1111/j.1462-5822.2009.01284.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Enteropathogenic Escherichia coli (EPEC), atypical enteropathogenic Escherichia coli (ATEC) and enterohemorrhagic Escherichia coli (EHEC) belong to the family of attaching and effacing (A/E) pathogens. Pathogenicity is mediated by subversion of host cell functions involving type III secretion system (TTSS)-dependent effector proteins. In this study, we have identified and characterized a novel TTSS-dependent effector protein encoded at the 5'-end of the locus of enterocyte effacement (LEE) pathogenicity island (PAI) of ATEC strain 3431-4/86 (O8:H(-)). Using affinity purification we identified IQGAP1, a scaffolding protein involved in the regulation of the actin cytoskeleton, as a putative host cell target. Accordingly, we termed the novel effector protein 'Ibe' for IQGAP1-binding effector. The interaction of Ibe and IQGAP1 was confirmed by co-immunoprecipitation from ATEC-infected cells and immunofluorescence analysis, which revealed colocalization of Ibe and IQGAP1 in ATEC-induced pedestals and actin-rich membrane ruffles. This suggests that the putative effector function of Ibe is mediated via IQGAP1. The Ibe-independent recruitment of IQGAP1 to ATEC-induced pedestals implies a general role for IQGAP1 in the subversion of host cell functions during infection. Homologues of the novel effector Ibe are widely distributed among EPEC, ATEC and EHEC strains but are not necessarily genetically linked to the LEE as they have occasionally also been found to be encoded within lambdoid prophages.
Collapse
Affiliation(s)
- Christoph Buss
- Institut für Infektiologie, Zentrum für Molekularbiologie der Entzündung (ZMBE), Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | | | | | | |
Collapse
|
246
|
Meunier B, Quaranta M, Daviet L, Hatzoglou A, Leprince C. The membrane-tubulating potential of amphiphysin 2/BIN1 is dependent on the microtubule-binding cytoplasmic linker protein 170 (CLIP-170). Eur J Cell Biol 2008; 88:91-102. [PMID: 19004523 DOI: 10.1016/j.ejcb.2008.08.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 08/07/2008] [Accepted: 08/07/2008] [Indexed: 01/02/2023] Open
Abstract
Amphiphysins are BIN-amphiphysin-RVS (BAR) domain-containing proteins that influence membrane curvature in sites such as T-tubules in muscular cells, endocytic pits in neuronal as well as non-neuronal cells, and possibly cytoplasmic endosomes. This effect on lipid membranes is fulfilled by diverse amphiphysin 2/BIN1 isoforms, generated by alternative splicing and showing distinct structural and functional properties. In this study, our goal was to characterize the functional role of a ubiquitously expressed amphiphysin 2/BIN1 by the characterization of new molecular partners. We performed a two-hybrid screen with an isoform of amphiphysin 2/BIN1 expressed in HeLa cells. We identified CLIP-170 as an amphiphysin 2/BIN1-interacting molecule. CLIP-170 is a plus-end tracking protein involved in microtubule (MT) stability and recruitment of dynactin. The binding between amphiphysin 2/BIN1 and CLIP-170 is dependent on the N-terminal part of amphiphysin 2 (mostly the BAR domain) and an internal coiled-coil region of CLIP-170. This partnership was confirmed by GST pull-down assay and by co-immunoprecipitation in HeLa cells that express endogenous amphiphysin 2 (mostly isoforms 6, 9 and 10). When overexpressed in HeLa cells, amphiphysin 2/BIN1 leads to the formation of intracellular tubules which can closely align with MTs. After MT depolymerization by nocodazole, amphiphysin 2-stained tubules disappear, and reappear after nocodazole washout. Furthermore, depletion of CLIP-170 by RNAi induced a decrease in the proportion of cells with amphiphysin 2-stained tubules and an increase in the proportion of cells with no tubules. This result suggests the existence of a mechanistic link between the two types of tubules, which is likely to involve the +TIP protein, CLIP-170. Amphiphysin 2/BIN1 may be an anchoring point on membranes for CLIP-170, and consequently for MT. Then, the pushing force of polymerizing MT could help amphiphysin 2/BIN1 in its tubulation potential. We propose that amphiphysin 2/BIN1 participates in the tubulation of traffic intermediates and intracellular organelles first via its intrinsic tubulating potential and second via its ability to bind CLIP-170 and MT.
Collapse
Affiliation(s)
- Brigitte Meunier
- Analysis of Signal Transduction Group, INSERM U830, Institut Curie, Paris, France
| | | | | | | | | |
Collapse
|
247
|
The IQGAP Iqg1 is a regulatory target of CDK for cytokinesis in Candida albicans. EMBO J 2008; 27:2998-3010. [PMID: 18923418 DOI: 10.1038/emboj.2008.219] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2008] [Accepted: 09/23/2008] [Indexed: 01/05/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) drive and coordinate multiple cell-cycle events, including construction and contraction of the actomyosin ring during cytokinesis. However, it remains unclear whether CDKs regulate cytokinesis by directly targeting components of the ring. In a search for proteins containing consensus CDK phosphorylation sites in Candida albicans, we found that the IQGAP Iqg1 contains two dense clusters of 19 such sites flanking the actin-interacting CH domain. Here, we show that Iqg1 is indeed a phosphoprotein that undergoes cell-cycle-dependent phosphorylation and can be phosphorylated by purified Clb-Cdc28 kinases in vitro. Mass spectrometry identified several phosphoserine and phosphothreonine residues among these CDK sites. Mutating 15 of the CDK phosphorylation sites with alanine markedly reduced Iqg1 phosphorylation in vivo. The 15A mutation greatly stabilized Iqg1, caused both premature assembly and delayed disassembly of the actomyosin ring, blocked Iqg1 interaction with the actin-nucleating proteins Bni1 and Bnr1, and resulted in defects in cytokinesis. Our data therefore strongly support the idea that the Cdc28 CDK regulates cytokinesis partly by directly phosphorylating the actomyosin ring component Iqg1.
Collapse
|
248
|
Tesař V, Zima T. Recent Progress in the Pathogenesis of Nephrotic Proteinuria. Crit Rev Clin Lab Sci 2008; 45:139-220. [DOI: 10.1080/10408360801934865] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
249
|
Investigating the action mechanism for a natural active compound honokiol by quantitative proteomics. J Biotechnol 2008. [DOI: 10.1016/j.jbiotec.2008.07.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
250
|
Gloushankova NA. Changes in regulation of cell-cell adhesion during tumor transformation. BIOCHEMISTRY (MOSCOW) 2008; 73:742-50. [PMID: 18707582 DOI: 10.1134/s000629790807002x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Cadherin-mediated cell-cell adhesion defines the integrity of most tissues. Cell-cell adherens junctions are dynamic structures whose functional state is regulated by interactions of cadherin with beta-catenin, p120, and actin cytoskeleton structures. Small GTPases of the Rho family and GTPase Rap1 play the central role in the formation and maintenance of cell-cell adhesion. Aberrant activation of signaling pathways, transcriptional repression of the E-cadherin gene, ectopic expression of N-cadherin, and disturbances in regulation of adhesive and transcriptional functions of beta-catenin stimulate tumor progression.
Collapse
Affiliation(s)
- N A Gloushankova
- Blokhin Cancer Research Center, Russian Academy of Medical Sciences, Moscow, 115478, Russia.
| |
Collapse
|