Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Naunyn Schmiedebergs Arch Pharmacol ; 394(1): 107-115, 2021 01.
Article in English | MEDLINE | ID: mdl-32840651

ABSTRACT

Tryptase is a serine protease that is released from mast cells during allergic responses. Tryptase inhibitors are being explored as treatments for allergic inflammation in the skin and respiratory system, most notably asthma. Here we report direct tryptase inhibition by natural product compounds. Candidate inhibitors were identified by computational screening of a large (98,000 compounds) virtual library of natural product compounds for tryptase enzymatic site binding. Biochemical assays were used to validate the predicted anti-tryptase activity in vitro, revealing a high (four out of six) success rate for predicting binding using the computational docking model. We further assess tryptase inhibition by a biflavonoid scaffold, whose structure-activity relationship is partially defined by assessing the potency of structurally similar analogs.


Subject(s)
Biflavonoids/pharmacology , Biological Products/pharmacology , Tryptases/antagonists & inhibitors , Biflavonoids/chemistry , Biological Products/chemistry , Molecular Docking Simulation , Structure-Activity Relationship , Tryptases/metabolism
2.
Bioorg Med Chem Lett ; 30(2): 126813, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31831383

ABSTRACT

A series of novel 3,6-di-substituted or 3-substituted pyrazolo[1,5-a]pyrimidines were prepared via a microwave-assisted approach that generated a broad array of derivatives in good yields (20-93%, ave. = 59%). The straightforward synthesis involved sequential treatment of commercially-available acetonitrile derivatives with DMF-dimethylacetal (120 °C, 20 min), followed by treatment with NH2NH2·HBr (120 °C, 20 min), and 1,1,3,3-tetramethoxypropane or 2-aryl-substituted malondialdehdyes (120 °C, 20 min). Compounds were screened for antimitotic activities against MCF7 breast cancer and/or A2780 ovarian cancer cell lines in vitro. The most active compounds exhibited EC50 values ranging from 0.5 to 4.3 µM, with the 3-(4-(trifluoromethyl)phenyl)-6-[4-(2-(piperidin-1-yl)ethoxy]phenyl analogue (34e) and the 3-(2-fluorophenyl)-6-[4-(2-(4-methylpiperizin-1-yl)ethoxy]phenyl analogue (35a) being two to three fold more active than Compound C (Dorsomorphin) in A2780 and MCF7 assays, respectively. Importantly, a monosubstituted 3-(benzothiazol-2-yl) derivative (13) was equipotent with the more synthetically challenging 3,6-disubstituted derivatives (34a-e and 35a-e), and exhibited a promising and unique selectivity profile when screened against a panel consisting of 403 protein kinases (Kinomescan™ selectivity score = 0.005, Kd = 0.55 ± 0.055 µM and 0.410 ± 0.20 µM for JAK1 JH2 pseudokinase and VPS34, respectively).


Subject(s)
Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Janus Kinase 1/antagonists & inhibitors , Pyrimidines/chemical synthesis , Humans , Molecular Structure , Structure-Activity Relationship
3.
Bioorg Med Chem Lett ; 29(13): 1647-1653, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31047749

ABSTRACT

Despite their clinical importance, drug resistance remains problematic for microtubule targeting drugs. D4-9-31, a novel microtubule destabilizing agent, has pharmacology that suggests it can overcome common resistance mechanisms and has been shown to remain efficacious in cell and animal models with acquired taxane resistance. To better understand resistance mechanisms and the breadth of cross-resistance with D4-9-31, this study examines the A2780 ovarian cancer cell line as it develops acquired resistance with continuous exposure to D4-9-31. Analyzing cellular responses to D4-9-31 reveals that D4-9-31 resistance is associated with increased mitochondrial respiration, but no cross-resistance to other microtubule targeting agents is observed. Sequencing of transcripts of parental cells and resistant counterparts reveals mutations and altered expression of microtubule-associated genes, but not in genes commonly associated with resistance to microtubule targeting drugs. Additionally, our findings suggest distinct mechanisms drive short- and long-term drug resistance.


Subject(s)
Amides/therapeutic use , Microtubules/drug effects , Polymerization/drug effects , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Amides/pharmacology , Humans , Pyridines/pharmacology , Pyrimidines/pharmacology
4.
Bioorg Med Chem Lett ; 28(5): 934-941, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29433928

ABSTRACT

Microtubule-targeting agents are important tools in cancer treatment. Generating novel microtubule targeting agents with novel pharmacology could dramatically expand the utility of this class of drugs. Here we characterize the pharmacology of recently described small molecule microtubule polymerization inhibitors. Pharmacokinetic experiments show oral bioavailability through gastric absorption. In vitro assays designed to predict absorption, distribution, metabolism, and excretion (ADME) and safety reveal a scaffold that is metabolically stable, evades P-glycoprotein, does not inhibit CYP enzymes, occurs as a significant free fraction in serum, and has exceptionally high cellular permeability. Together with in vivo efficacy models, pharmacology supports further development as a treatment for solid tumors.


Subject(s)
Amides/pharmacology , Microtubules/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Amides/chemical synthesis , Amides/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Microtubules/metabolism , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Polymerization/drug effects , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Rats , Structure-Activity Relationship
5.
Bioorg Med Chem Lett ; 27(17): 3992-4000, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28780159

ABSTRACT

Stimulation of cultured epithelial cells with scatter factor/hepatocyte growth factor (HGF) results in individual cells detaching and assuming a migratory and invasive phenotype. Epithelial scattering recapitulates cancer progression and studies have implicated HGF signaling as a driver of cancer metastasis. Inhibitors of HGF signaling have been proposed to act as anti-cancer agents. We previously screened a small molecule library for compounds that block HGF-induced epithelial scattering. Most hits identified in this screen exhibit anti-mitotic properties. Here we assess the biological mechanism of a compound that blocks HGF-induced scattering with limited anti-mitotic activity. Analogs of this compound have one of two distinct activities: inhibiting either cell migration or cell proliferation with cell cycle arrest in G2/M. Each activity bears unique structure-activity relationships. The mechanism of action of anti-mitotic compounds is by inhibition of microtubule polymerization; these compounds entropically and enthalpically bind tubulin in the colchicine binding site, generating a conformational change in the tubulin dimer.


Subject(s)
Amides/pharmacology , Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Hepatocyte Growth Factor/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Amides/chemical synthesis , Amides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial Cells/metabolism , Hepatocyte Growth Factor/metabolism , Humans , MCF-7 Cells , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
6.
J Theor Biol ; 430: 141-156, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28648562

ABSTRACT

Epithelial cells can be triggered to actively detach from epithelial tissues and become solitary, migratory and invasive. This process occurs repeatedly in development, where it is termed epithelial-mesenchymal transition (EMT), and can be recapitulated as epithelial scattering in cell culture models. Detachment of cell-cell junctions involves changes in contractile forces, actin cytoskeletal organization, changes in cell-substrate adhesion properties, surface presentation of cell-cell adhesion molecules, and gene expression. That these cellular processes affect each other and share molecular components creates difficulties in generating hypotheses and designing experiments to understand the mechanics of epithelial scattering. Computational modeling is proving a powerful too in such instances. Here we develop a cellular automaton to reveal insights into how cells rupture epithelial cell-cell junctions during scattering. The model is optimized for realistic and stable recapitulation of behavior of single cells, then for realistic simulation of multiple cells forming epithelial colonies. With a workable model of epithelial cell behavior, we then alter model parameters and assess whether we can realistically mimic epithelial scattering. Adjusting model parameters to recapitulate epithelial scattering reveals that induction of cell migration is the major driver of epithelial scattering.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Epithelial Cells/cytology , Animals , Biomechanical Phenomena , Cell Adhesion , Epithelial-Mesenchymal Transition , Humans , Intercellular Junctions/metabolism , Models, Biological
7.
Pharm Biol ; 55(1): 1586-1591, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28395583

ABSTRACT

CONTEXT: Kalanchoe pinnata (Lam.) Pers. (Crassulaceae) is a succulent plant that is known for its traditional antivirus and antibacterial usage. OBJECTIVE: This work examines two compounds identified from the K. pinnata plant for their antivirus activity against human alphaherpesvirus (HHV) 1 and 2 and vaccinia virus (VACV). MATERIALS AND METHODS: Compounds KPB-100 and KPB-200 were isolated using HPLC and were identified using NMR and MS. Both compounds were tested in plaque reduction assay of HHV-2 wild type (WT) and VACV. Both compounds were then tested in virus spread inhibition and virus yield reduction (VYR) assays of VACV. KPB-100 was further tested in viral cytopathic effect (CPE) inhibition assay of HHV-2 TK-mutant and VYR assay of HHV-1 WT. RESULTS: KPB-100 and KPB-200 inhibited HHV-2 at IC50 values of 2.5 and 2.9 µg/mL, respectively, and VACV at IC50 values of 3.1 and 7.4 µg/mL, respectively, in plaque reduction assays. In virus spread inhibition assay of VACV KPB-100 and KPB-200 yielded IC50 values of 1.63 and 13.2 µg/mL, respectively, and KPB-100 showed a nearly 2-log reduction in virus in VYR assay of VACV at 20 µg/mL. Finally, KPB-100 inhibited HHV-2 TK- at an IC50 value of 4.5 µg/mL in CPE inhibition assay and HHV-1 at an IC90 of 3.0 µg/mL in VYR assay. DISCUSSION AND CONCLUSION: Both compounds are promising targets for synthetic optimization and in vivo study. KPB-100 in particular showed strong inhibition of all viruses tested.


Subject(s)
Antiviral Agents/pharmacology , Kalanchoe/chemistry , Plant Extracts/pharmacology , Antiviral Agents/administration & dosage , Antiviral Agents/isolation & purification , Chromatography, High Pressure Liquid , Cytopathogenic Effect, Viral/drug effects , HeLa Cells , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Vaccinia virus/drug effects
8.
Tissue Barriers ; 4(3): e1187326, 2016.
Article in English | MEDLINE | ID: mdl-27583192

ABSTRACT

Epithelial tissues use adherens junctions to maintain tight interactions and coordinate cellular activities. Adherens junctions are remodeled during epithelial morphogenesis, including instances of epithelial-mesenchymal transition, or EMT, wherein individual cells detach from the tissue and migrate as individual cells. EMT has been recapitulated by growth factor induction of epithelial scattering in cell culture. In culture systems, cells undergo a highly reproducible series of cell morphology changes, most notably cell spreading followed by cellular compaction and cell migration. These morphology changes are accompanied by striking actin rearrangements. The current evidence suggests that global changes in actomyosin-based cellular contractility, first a loss of contractility during spreading and its activation during cell compaction, are the main drivers of epithelial scattering. In this review, we focus on how spreading and contractility might be controlled during epithelial scattering. While we propose a central role for RhoA, which is well known to control cellular contractility in multiple systems and whose role in epithelial scattering is well accepted, we suggest potential roles for additional cellular systems whose role in epithelial cell biology has been less well documented. In particular, we propose critical roles for vesicle recycling, calcium channels, and calcium-dependent kinases.


Subject(s)
Calcium Signaling , Epithelial-Mesenchymal Transition , Epithelium/metabolism , rhoA GTP-Binding Protein/metabolism , Adherens Junctions/metabolism , Animals , Epithelium/pathology , Humans
9.
J Biomol Screen ; 21(7): 671-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27245142

ABSTRACT

Stimulation of cultured epithelial cells with scatter factor/hepatocyte growth factor (HGF) results in the detachment of cell-cell junctions and initiation of cell migration. Instead of coordinating collective cell behavior within a tissue, cells become solitary and have few cell-cell interactions. Since epithelial scattering is recapitulated in cancer progression and since HGF signaling drives cancer metastasis in many cases, inhibitors of HGF signaling have been proposed to act as anticancer agents. We previously sought to better understand critical components required for HGF-induced epithelial scattering by performing a forward chemical genetics screen, which resulted in the identification of compounds with no previously reported biological activity that we report here. In efforts to determine the mechanism of these compounds, we find that many compounds have broad antiproliferative effects on cancer cell lines by arrest of cell division in G2/M with minimal induction of apoptosis. This effect is reminiscent of microtubule-targeting agents, and we find that several of these scaffolds directly inhibit microtubule polymerization. Compounds are assessed for their toxicity and pharmacokinetics in vivo. The identification of novel small-molecule inhibitors of microtubule polymerization highlights the role of the microtubule cytoskeleton in HGF-induced epithelial scattering.


Subject(s)
Antineoplastic Agents/isolation & purification , Hepatocyte Growth Factor/antagonists & inhibitors , High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Small Molecule Libraries/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Hepatocyte Growth Factor/genetics , Humans , Intercellular Junctions/drug effects , Microtubules/drug effects , Neoplasm Metastasis , Neoplasms/pathology , Polymerization/drug effects , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology
10.
Biochim Biophys Acta ; 1853(2): 388-95, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450971

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) and Zyxin are interacting proteins involved in cellular adhesion and motility. PKA phosphorylates VASP at serine 157, regulating VASP cellular functions. VASP interacts with ABL and is a substrate of the BCR-ABL oncoprotein. The presence of BCR-ABL protein drives oncogenesis in patients with chronic myeloid leukemia (CML) due to a constitutive activation of tyrosine kinase activity. However, the function of VASP and Zyxin in BCR-ABL pathway and the role of VASP in CML cells remain unknown. In vitro experiments using K562 cells showed the involvement of VASP in BCR-ABL signaling. VASP and Zyxin inhibition decreased the expression of anti-apoptotic proteins, BCL2 and BCL-XL. Imatinib induced an increase in phosphorylation at Ser157 of VASP and decreased VASP and BCR-ABL interaction. VASP did not interact with Zyxin in K562 cells; however, after Imatinib treatment, this interaction was restored. Corroborating our data, we demonstrated the absence of phosphorylation at Ser157 in VASP in the bone marrow of CML patients, in contrast to healthy donors. Phosphorylation of VASP on Ser157 was restored in Imatinib responsive patients though not in the resistant patients. Therefore, we herein identified a possible role of VASP in CML pathogenesis, through the regulation of BCR-ABL effector proteins or the absence of phosphorylation at Ser157 in VASP.


Subject(s)
Benzamides/pharmacology , Cell Adhesion Molecules/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Zyxin/metabolism , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Proliferation/drug effects , Clone Cells , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Imatinib Mesylate , K562 Cells , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , bcl-X Protein/metabolism
11.
Exp Cell Res ; 326(2): 187-200, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24780819

ABSTRACT

Epithelial scattering occurs when cells disassemble cell-cell junctions, allowing individual epithelial cells to act in a solitary manner. Epithelial scattering occurs frequently in development, where it accompanies epithelial-mesenchymal transitions and is required for individual cells to migrate and invade. While migration and invasion have received extensive research focus, how cell-cell junctions are detached remains poorly understood. An open debate has been whether disruption of cell-cell interactions occurs by remodeling of cell-cell adhesions, increased traction forces through cell substrate adhesions, or some combination of both processes. Here we seek to examine how changes in adhesion and contractility are coupled to drive detachment of individual epithelial cells during hepatocyte growth factor (HGF)/scatter factor-induced EMT. We find that HGF signaling does not alter the strength of cell-cell adhesion between cells in suspension, suggesting that changes in cell-cell adhesion strength might not accompany epithelial scattering. Instead, cell-substrate adhesion seems to play a bigger role, as cell-substrate adhesions are stronger in cells treated with HGF and since rapid scattering in cells treated with HGF and TGFß is associated with a dramatic increase in focal adhesions. Increases in the pliability of the substratum, reducing cells ability to generate traction on the substrate, alter cells׳ ability to scatter. Further consistent with changes in substrate adhesion being required for cell-cell detachment during EMT, scattering is impaired in cells expressing both active and inactive RhoA mutants, though in different ways. In addition to its roles in driving assembly of both stress fibers and focal adhesions, RhoA also generates myosin-based contractility in cells. We therefore sought to examine how RhoA-dependent contractility contributes to cell-cell detachment. Inhibition of Rho kinase or myosin II induces the same effect on cells, namely an inhibition of cell scattering following HGF treatment. Interestingly, restoration of myosin-based contractility in blebbistatin-treated cells results in cell scattering, including global actin rearrangements. Scattering is reminiscent of HGF-induced epithelial scattering without a concomitant increase in cell migration or decrease in adhesion strength. This scattering is dependent on RhoA, as blebbistatin-induced scattering is reduced in cells expressing dominant-negative RhoA mutants. This suggests that induction of myosin-based cellular contractility may be sufficient for cell-cell detachment during epithelial scattering.


Subject(s)
Cell Movement/physiology , Epithelial Cells/physiology , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Line , Dogs , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/physiology , Focal Adhesions/physiology , Hepatocyte Growth Factor/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Intercellular Junctions/physiology , Mutation , Myosin Type II/metabolism , Signal Transduction , Stress Fibers/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
12.
Biomedicines ; 2(4): 384-402, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-28548077

ABSTRACT

Signaling from the c-Met receptor tyrosine kinase is associated with progression and metastasis of epithelial tumors. c-Met, the receptor for hepatocyte growth factor, triggers epithelial-mesenchymal transition (EMT) of cultured cells, which is thought to drive migration of tumor cells and confer on them critical stem cell properties. Here, we employ mathematical modeling to better understand how EMT affects population dynamics in metastatic tumors. We find that without intervention, micrometastatic tumors reach a steady-state population. While the rates of proliferation, senescence and death only have subtle effects on the steady state, changes in the frequency of EMT dramatically alter population dynamics towards exponential growth. We also find that therapies targeting cell proliferation or cell death are markedly more successful when combined with one that prevents EMT, though such therapies do little when used alone. Stochastic modeling reveals the probability of tumor recurrence from small numbers of residual differentiated tumor cells. EMT events in metastatic tumors provide a plausible mechanism by which clinically detectable tumors can arise from dormant micrometastatic tumors. Modeling the dynamics of this process demonstrates the benefit of a treatment that eradicates tumor cells and reduces the rate of EMT simultaneously.

13.
Int Rev Cell Mol Biol ; 303: 1-25, 2013.
Article in English | MEDLINE | ID: mdl-23445807

ABSTRACT

The regulated assembly and organization of actin filaments allows the cell to construct a large diversity of actin-based structures specifically suited to a range of cellular processes. A vast array of actin regulatory proteins must work in concert to form specific actin networks within cells, and spatial and temporal requirements for actin assembly necessitate rapid regulation of protein activity. This chapter explores a common mechanism of controlling the activity of actin binding proteins: allosteric autoinhibition by interdomain head-tail interactions. Intramolecular interactions maintain these proteins in a closed conformation that masks protein domains needed to regulate actin dynamics. Autoinhibition is typically relieved by two or more ligand binding and/or posttranslational modification events that expose key protein domains. Regulation through multiple inputs permits precise temporal and spatial control of protein activity to guide actin network formation.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Allosteric Regulation , Animals , Humans , Models, Biological
14.
Cell Mol Biol Lett ; 18(1): 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23076992

ABSTRACT

Cell-cell and cell-substrate adhesions are sites of dramatic actin rearrangements and where actin-membrane connections are tightly regulated. Zyxin-VASP complexes localize to sites of cell-cell and cell-substrate adhesion and function to regulate actin dynamics and actin-membrane connections at these sites. To accomplish these functions, zyxin recruits VASP to cellular sites via proline-rich binding sites near zyxin's amino terminus. While the prevailing thought has been that zyxin simply acts as a scaffold protein for VASP binding, the identification of a LIM domain-VASP interaction could complicate this view. Here we assess how zyxin-VASP binding through both the proline rich motifs and the LIM domains alters specific VASP functions. We find that neither individual interaction alters VASP's actin regulatory activities. In contrast, however, we find that full-length zyxin dramatically reduces VASP-mediated actin bundling and actin assembly. Taken together, these results suggest a model where zyxin-VASP complexes occur in complex organizations with suppressed actin regulatory activity.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Zyxin/metabolism , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/genetics , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cell Communication , Eukaryotic Cells/chemistry , Eukaryotic Cells/metabolism , Humans , Microfilament Proteins/chemistry , Phosphoproteins/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Zyxin/chemistry
15.
J Biol Chem ; 288(4): 2179-89, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23235160

ABSTRACT

Cell-cell adhesions and the cytoskeletons play important and coordinated roles in cell biology, including cell differentiation, development, and migration. Adhesion and cytoskeletal dynamics are regulated by Rho-GTPases. ARHGAP21 is a negative regulator of Rho-GTPases, particularly Cdc42. Here we assess the function of ARHGAP21 in cell-cell adhesion, cell migration, and scattering. We find that ARHGAP21 is localized in the nucleus, cytoplasm, or perinuclear region but is transiently redistributed to cell-cell junctions 4 h after initiation of cell-cell adhesion. ARHGAP21 interacts with Cdc42, and decreased Cdc42 activity coincides with the appearance of ARHGAP21 at the cell-cell junctions. Cells lacking ARHGAP21 expression show weaker cell-cell adhesions, increased cell migration, and a diminished ability to undergo hepatocyte growth factor-induced epithelial-mesenchymal transition (EMT). In addition, ARHGAP21 interacts with α-tubulin, and it is essential for α-tubulin acetylation in EMT. Our findings indicate that ARHGAP21 is a Rho-GAP involved in cell-cell junction remodeling and that ARHGAP21 affects migration and EMT through α-tubulin interaction and acetylation.


Subject(s)
Epithelial-Mesenchymal Transition , Epithelium/metabolism , GTPase-Activating Proteins/physiology , Tubulin/metabolism , Acetylation , Animals , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cell Movement , Dogs , GTPase-Activating Proteins/metabolism , Humans , Madin Darby Canine Kidney Cells , Neoplasm Metastasis , RNA Interference , Time Factors , cdc42 GTP-Binding Protein/metabolism
16.
J Cell Sci ; 125(Pt 17): 4001-13, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22685327

ABSTRACT

Hepatocyte growth factor (HGF) signaling drives epithelial cells to scatter by breaking cell-cell adhesions and causing them to migrate as solitary cells, a process that parallels epithelial-mesenchymal transition. HGF binds and activates the c-met receptor tyrosine kinase, but downstream signaling required for scattering remains poorly defined. We have applied a chemical biology approach to identify components of HGF signaling that are required for scattering in an in vitro model system. This approach yields a number of small molecules that block HGF-induced scattering, including a calcium channel blocker. We show that HGF stimulation results in sudden and transient increases in ion channel influxes at the plasma membrane. Although multiple channels occur in the membranes of our model system, we find that TrpC6 is specifically required for HGF-induced scattering. We further demonstrate that HGF-induced ion influxes through TrpC6 channels coincide with a transient increase in nuclear factor of activated T-cells (NFAT)-dependent gene transcription and that NFAT-dependent gene transcription is required for HGF-induced cell scattering.


Subject(s)
Cell Membrane/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Ions/metabolism , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Transcription, Genetic , Actins/metabolism , Animals , Calcium/metabolism , Cell Membrane/drug effects , Dogs , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hepatocyte Growth Factor/pharmacology , Madin Darby Canine Kidney Cells , Microtubules/drug effects , Microtubules/metabolism , Reproducibility of Results , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , TRPC Cation Channels/metabolism , TRPV Cation Channels/metabolism , Transcription, Genetic/drug effects
17.
Biochem Biophys Res Commun ; 415(3): 485-9, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-22056563

ABSTRACT

Cell-cell junction remodeling is associated with dramatic actin reorganizations. Several actin regulatory systems have been implicated in actin remodeling events as cell-cell contacts are assembled and disassembled, including zyxin/LPP-VASP complexes. These complexes facilitate strong cell-cell adhesion by maintaining actin-membrane connections. It has been proposed that zyxin and LPP localize to cell-cell junctions via a well-defined interaction with alpha-actinin. This was recently confirmed for LPP, but zyxin localization at cell-cell contacts occurs independently of alpha-actinin binding. Here we seek to map the zyxin sequence responsible for localization to cell-cell contacts and identify the protein that docks zyxin at this cellular location. Previous results have shown that a zyxin fragment excluding the alpha-actin binding site and the LIM domains (amino acids 51-392) can independently localize to cell-cell contacts. Here, expression of smaller zyxin fragments show that zyxin localization requires amino acids 230-280. A yeast-two-hybrid screen, using the central region of zyxin as bait, resulted in the identification of the cell-cell adhesion receptor nectin-4 as a zyxin binding partner. Further demonstrating zyxin-nectin interactions, zyxin binds the intracellular domain of nectin-2 in vitro. Depletion of nectin-2 from L cells expressing E-cadherin results in a loss of zyxin localization to cell-cell contacts, demonstrating that the zyxin-nectin interaction plays a critical role in zyxin targeting to these sites.


Subject(s)
Cell Adhesion Molecules/metabolism , Zyxin/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Line , Dogs , Humans , Nectins , Protein Structure, Tertiary , Two-Hybrid System Techniques , Zyxin/chemistry , Zyxin/genetics
18.
Biochem Biophys Res Commun ; 414(1): 215-9, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21945940

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) is an actin regulatory protein that functions in adhesion and migration. In epithelial cells, VASP participates in cell-cell adhesion. At the molecular level, VASP drives actin bundling and polymerization. VASP activity is primarily regulated by phosphorylation. Three physiologically relevant phosphorylation sites significantly reduce actin regulatory activity and are targeted by several kinases, most notable Abl and protein kinases A and G (PKA and PKG). AMP-dependent kinase (AMPK) is best characterized as a cellular sensor of ATP depletion, but also alters actin dynamics in epithelial cells and participates in cell polarity pathways downstream of LKB1. While little is known about how AMPK direct changes in actin dynamics, AMPK has been shown to phosphorylate VASP at one of these three well-characterized PKA/PKG phosphorylation sites. Here we show that phosphorylation of VASP by AMPK occurs at a novel site, serine 322, and that phosphorylation at this site alters actin filament binding. We also show that inhibition of AMPK activity results in the accumulation of VASP at cell-cell adhesions and a concomitant increase in cell-cell adhesion.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Actins/metabolism , Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Line , Dogs , Mice , Microfilament Proteins/genetics , Molecular Sequence Data , Phosphoproteins/genetics , Phosphorylation , Serine/genetics
19.
Biochem Biophys Res Commun ; 404(3): 780-4, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21168386

ABSTRACT

Zyxin is an actin regulatory protein that is concentrated at sites of actin-membrane association, particularly cell junctions. Zyxin participates in actin dynamics by binding VASP, an interaction that occurs via proline-rich N-terminal ActA repeats. An intramolecular association of the N-terminal LIM domains at or near the ActA repeats can prevent VASP and other binding partners from binding full-length zyxin. Such a head-tail interaction likely accounts for how zyxin function in actin dynamics, cell adhesion, and cell migration can be regulated by the cell. Since zyxin binding to several partners, via the LIM domains, requires phosphorylation, it seems likely that zyxin phosphorylation might alter the head-tail interaction and, thus, zyxin activity. Here we show that zyxin point mutants at a known phosphorylation site, serine 142, alter the ability of a zyxin fragment to directly bind a separate zyxin LIM domains fragment protein. Further, expression of the zyxin phosphomimetic mutant results in increased localization to cell-cell contacts of MDCK cells and generates a cellular phenotype, namely inability to disassemble cell-cell contacts, precisely like that produced by expression of zyxin mutants that lack the entire regulatory LIM domain region. These data suggest that zyxin phosphorylation at serine 142 results in release of the head-tail interaction, changing zyxin activity at cell-cell contacts.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/metabolism , Serine/metabolism , Animals , Cell Adhesion , Cytoskeletal Proteins/genetics , Dogs , Phosphorylation , Point Mutation , Protein Structure, Tertiary , Serine/genetics
20.
Exp Cell Res ; 317(3): 307-18, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21075102

ABSTRACT

HGF signaling induces epithelial cells to disassemble cadherin-based adhesion and increase cell motility and invasion, a process termed epithelial-mesenchymal transition (EMT). EMT plays a major role in cancer metastasis, allowing individual cells to detach from the primary tumor, invade local tissue, and colonize distant tissues with new tumors. While invasion of vascular and lymphatic networks is the predominant route of metastasis, nerves also can act as networks for dissemination of cancer cell to distant sites in a process termed perineual invasion (PNI). Signaling between nerves and invasive cancer cells remains poorly understood, as does cellular decision making that selects the specific route of invasion. Here we examine how HGF signaling contributes to PNI using reductionist culture model systems. We find that TGFß, produced by PC12 cells, enhances scattering in response to HGF stimulation, increasing both cell-cell junction disassembly and cell migration. Further, gradients of TGFß induce migratory mesenchymal cells to undergo chemotaxis towards the source of TGFß. Interestingly, VEGF suppresses TGFß-induced enhancement of scattering. These results have broad implications for how combinatorial growth factor signaling contributes to cancer metastasis, suggesting that VEGF and TGFß might modulate HGF signaling to influence route selection during cancer progression.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hepatocyte Growth Factor/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Cell Movement , Neoplasm Metastasis , PC12 Cells , Rats , Signal Transduction , Transforming Growth Factor beta/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...