Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Cancer Res ; 78(22): 6509-6522, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30279244

ABSTRACT

Glioblastoma (GBM) is an aggressive and incurable primary brain tumor that causes severe neurologic, cognitive, and psychologic symptoms. Symptoms are caused and exacerbated by the infiltrative properties of GBM cells, which enable them to pervade the healthy brain and disrupt normal function. Recent research has indicated that although radiotherapy (RT) remains the most effective component of multimodality therapy for patients with GBM, it can provoke a more infiltrative phenotype in GBM cells that survive treatment. Here, we demonstrate an essential role of the actin-myosin regulatory kinase myotonic dystrophy kinase-related CDC42-binding kinase (MRCK) in mediating the proinvasive effects of radiation. MRCK-mediated invasion occurred via downstream signaling to effector molecules MYPT1 and MLC2. MRCK was activated by clinically relevant doses per fraction of radiation, and this activation was concomitant with an increase in GBM cell motility and invasion. Furthermore, ablation of MRCK activity either by RNAi or by inhibition with the novel small-molecule inhibitor BDP-9066 prevented radiation-driven increases in motility both in vitro and in a clinically relevant orthotopic xenograft model of GBM. Crucially, treatment with BDP-9066 in combination with RT significantly increased survival in this model and markedly reduced infiltration of the contralateral cerebral hemisphere.Significance: An effective new strategy for the treatment of glioblastoma uses a novel, anti-invasive chemotherapeutic to prevent infiltration of the normal brain by glioblastoma cells.Cancer Res; 78(22); 6509-22. ©2018 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Myotonin-Protein Kinase/antagonists & inhibitors , Actins/chemistry , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/radiotherapy , Cardiac Myosins/metabolism , Cell Line, Tumor , Cell Movement , Female , Glioblastoma/radiotherapy , Humans , Mice , Mice, Nude , Microscopy, Fluorescence , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Myosins/chemistry , Neoplasm Invasiveness , Phenotype , RNA Interference , RNA, Small Interfering/metabolism
3.
Cancer Res ; 78(8): 2096-2114, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29382705

ABSTRACT

The myotonic dystrophy-related Cdc42-binding kinases MRCKα and MRCKß contribute to the regulation of actin-myosin cytoskeleton organization and dynamics, acting in concert with the Rho-associated coiled-coil kinases ROCK1 and ROCK2. The absence of highly potent and selective MRCK inhibitors has resulted in relatively little knowledge of the potential roles of these kinases in cancer. Here, we report the discovery of the azaindole compounds BDP8900 and BDP9066 as potent and selective MRCK inhibitors that reduce substrate phosphorylation, leading to morphologic changes in cancer cells along with inhibition of their motility and invasive character. In over 750 human cancer cell lines tested, BDP8900 and BDP9066 displayed consistent antiproliferative effects with greatest activity in hematologic cancer cells. Mass spectrometry identified MRCKα S1003 as an autophosphorylation site, enabling development of a phosphorylation-sensitive antibody tool to report on MRCKα status in tumor specimens. In a two-stage chemical carcinogenesis model of murine squamous cell carcinoma, topical treatments reduced MRCKα S1003 autophosphorylation and skin papilloma outgrowth. In parallel work, we validated a phospho-selective antibody with the capability to monitor drug pharmacodynamics. Taken together, our findings establish an important oncogenic role for MRCK in cancer, and they offer an initial preclinical proof of concept for MRCK inhibition as a valid therapeutic strategy.Significance: The development of selective small-molecule inhibitors of the Cdc42-binding MRCK kinases reveals their essential roles in cancer cell viability, migration, and invasive character. Cancer Res; 78(8); 2096-114. ©2018 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Drug Discovery , Myotonin-Protein Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/enzymology , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Nude , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Skin Neoplasms/enzymology , Xenograft Model Antitumor Assays
4.
Oncotarget ; 6(36): 38469-86, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26540348

ABSTRACT

The actin and microtubule cytoskeletons are critically important for cancer cell proliferation, and drugs that target microtubules are widely-used cancer therapies. However, their utility is compromised by toxicities due to dose and exposure. To overcome these issues, we characterized how inhibition of the actin and microtubule cytoskeleton regulatory LIM kinases could be used in drug combinations to increase efficacy. A previously-described LIMK inhibitor (LIMKi) induced dose-dependent microtubule alterations that resulted in significant mitotic defects, and increased the cytotoxic potency of microtubule polymerization inhibitors. By combining LIMKi with 366 compounds from the GSK Published Kinase Inhibitor Set, effective combinations were identified with kinase inhibitors including EGFR, p38 and Raf. These findings encouraged a drug discovery effort that led to development of CRT0105446 and CRT0105950, which potently block LIMK1 and LIMK2 activity in vitro, and inhibit cofilin phosphorylation and increase αTubulin acetylation in cells. CRT0105446 and CRT0105950 were screened against 656 cancer cell lines, and rhabdomyosarcoma, neuroblastoma and kidney cancer cells were identified as significantly sensitive to both LIMK inhibitors. These large-scale screens have identified effective LIMK inhibitor drug combinations and sensitive cancer types. In addition, the LIMK inhibitory compounds CRT0105446 and CRT0105950 will enable further development of LIMK-targeted cancer therapy.


Subject(s)
Lim Kinases/antagonists & inhibitors , Mitosis/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , MCF-7 Cells , Microtubules/metabolism , Mitosis/physiology , Neoplasms/enzymology , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Neuroblastoma/pathology
5.
Proc Natl Acad Sci U S A ; 112(3): 773-8, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25568088

ABSTRACT

(Macro)autophagy delivers cellular constituents to lysosomes for degradation. Although a cytoplasmic process, autophagy-deficient cells accumulate genomic damage, but an explanation for this effect is currently unclear. We report here that inhibition of autophagy causes elevated proteasomal activity leading to enhanced degradation of checkpoint kinase 1 (Chk1), a pivotal factor for the error-free DNA repair process, homologous recombination (HR). We show that loss of autophagy critically impairs HR and that autophagy-deficient cells accrue micronuclei and sub-G1 DNA, indicators of diminished genomic integrity. Moreover, due to impaired HR, autophagy-deficient cells are hyperdependent on nonhomologous end joining (NHEJ) for repair of DNA double-strand breaks. Consequently, inhibition of NHEJ following DNA damage in the absence of autophagy results in persistence of genomic lesions and rapid cell death. Because autophagy deficiency occurs in several diseases, these findings constitute an important link between autophagy and DNA repair and highlight a synthetic lethal strategy to kill autophagy-deficient cells.


Subject(s)
Autophagy , DNA Repair/genetics , Genes, Lethal , Animals , Base Sequence , Cells, Cultured , DNA Primers , Homologous Recombination , Mice , Real-Time Polymerase Chain Reaction
6.
Cell Commun Signal ; 12: 54, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25288205

ABSTRACT

BACKGROUND: The myotonic dystrophy kinase-related CDC42-binding kinases MRCKα and MRCKß regulate actin-myosin contractility and have been implicated in cancer metastasis. Along with the related ROCK1 and ROCK2 kinases, the MRCK proteins initiate signalling events that lead to contractile force generation which powers cancer cell motility and invasion. A potential strategy for cancer therapy is to reduce metastasis by blocking MRCK activity, either alone or in combination with ROCK inhibition. However, to date no potent small molecule inhibitors have been developed with selectivity towards MRCK. RESULTS: Screening a kinase-focused small molecule chemical library resulted in the identification of compounds with inhibitory activity towards MRCK. Medicinal chemistry combined with in vitro enzyme profiling led to the discovery of 4-chloro-1-(4-piperidyl)-N-[5-(2-pyridyl)-1H-pyrazol-4-yl]pyrazole-3-carboxamide (BDP00005290; abbreviated as BDP5290) as a potent MRCK inhibitor. X-ray crystallography of the MRCKß kinase domain in complex with BDP5290 revealed how this ligand interacts with the nucleotide binding pocket. BDP5290 demonstrated marked selectivity for MRCKß over ROCK1 or ROCK2 for inhibition of myosin II light chain (MLC) phosphorylation in cells. While BDP5290 was able to block MLC phosphorylation at both cytoplasmic actin stress fibres and peripheral cortical actin bundles, the ROCK selective inhibitor Y27632 primarily reduced MLC phosphorylation on stress fibres. BDP5290 was also more effective at reducing MDA-MB-231 breast cancer cell invasion through Matrigel than Y27632. Finally, the ability of human SCC12 squamous cell carcinoma cells to invade a three-dimensional collagen matrix was strongly inhibited by 2 µM BDP5290 but not the identical concentration of Y27632, despite equivalent inhibition of MLC phosphorylation. CONCLUSIONS: BDP5290 is a potent MRCK inhibitor with activity in cells, resulting in reduced MLC phosphorylation, cell motility and tumour cell invasion. The discovery of this compound will enable further investigations into the biological activities of MRCK proteins and their contributions to cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Myotonin-Protein Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Amides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Humans , Myotonin-Protein Kinase/metabolism , Neoplasm Invasiveness , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
7.
Biomaterials ; 33(9): 2701-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22200536

ABSTRACT

The potential of gene therapy to treat cancer is hampered by the lack of safe and efficacious gene delivery systems able to selectively deliver therapeutic genes to tumors by intravenous administration. With the long-term aim of developing an efficacious cancer-targeted gene medicine, we demonstrated that transferrin-bearing polypropylenimine dendrimer complexed to a plasmid DNA encoding p73 led to an enhanced anti-proliferative activity in vitro, by up to 120-fold in A431 compared to the unmodified dendriplex. In vivo, the intravenous administration of this p73-encoding dendriplex resulted in a rapid and sustained inhibition of tumor growth over one month, with complete tumor suppression for 10% of A431 and B16-F10 tumors and long-term survival of the animals. The treatment was well tolerated by the animals, with no apparent signs of toxicity. These results suggest that the p73-encoding tumor-targeted polypropylenimine dendrimer should be further explored as a therapeutic strategy for cancer therapy.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/therapeutic use , Gene Transfer Techniques , Genetic Therapy/methods , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Nuclear Proteins/genetics , Nuclear Proteins/therapeutic use , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/therapeutic use , Aminobutyrates/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , DNA/metabolism , Dendrimers/administration & dosage , Dendrimers/chemistry , Diagnostic Imaging , Electrophoretic Mobility Shift Assay , Female , Humans , Injections, Intravenous , Luminescent Measurements , Mice , Mice, Inbred BALB C , Remission Induction , Transfection , Transferrin/metabolism , Tumor Protein p73 , Xenograft Model Antitumor Assays
8.
Autophagy ; 7(11): 1295-301, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21808150

ABSTRACT

(Macro)autophagy is a membrane-trafficking process that serves to sequester cellular constituents in organelles termed autophagosomes, which target their degradation in the lysosome. Autophagy operates at basal levels in all cells where it serves as a homeostatic mechanism to maintain cellular integrity. The levels and cargoes of autophagy can, however, change in response to a variety of stimuli, and perturbations in autophagy are known to be involved in the aetiology of various human diseases. Autophagy must therefore be tightly controlled. We report here that the Drosophila cyclin-dependent kinase PITSLRE is a modulator of autophagy. Loss of the human PITSLRE orthologue, CDK11, initially appears to induce autophagy, but at later time points CDK11 is critically required for autophagic flux and cargo digestion. Since PITSLRE/CDK11 regulates autophagy in both Drosophila and human cells, this kinase represents a novel phylogenetically conserved component of the autophagy machinery.


Subject(s)
Autophagy , Cyclin-Dependent Kinases/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Animals , Cell Line, Tumor , Humans , Lysosomes/metabolism , Phagosomes/metabolism , Sequence Homology, Amino Acid
9.
Cancer Cell ; 19(6): 776-91, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21665151

ABSTRACT

Tumors and associated stroma manifest mechanical properties that promote cancer. Mechanosensation of tissue stiffness activates the Rho/ROCK pathway to increase actomyosin-mediated cellular tension to re-establish force equilibrium. To determine how actomyosin tension affects tissue homeostasis and tumor development, we expressed conditionally active ROCK2 in mouse skin. ROCK activation elevated tissue stiffness via increased collagen. ß-catenin, a key element of mechanotranscription pathways, was stabilized by ROCK activation leading to nuclear accumulation, transcriptional activation, and consequent hyperproliferation and skin thickening. Inhibiting actomyosin contractility by blocking LIMK or myosin ATPase attenuated these responses, as did FAK inhibition. Tumor number, growth, and progression were increased by ROCK activation, while ROCK blockade was inhibitory, implicating actomyosin-mediated cellular tension and consequent collagen deposition as significant tumor promoters.


Subject(s)
Actomyosin/physiology , Epidermis/pathology , Skin Neoplasms/etiology , beta Catenin/physiology , Animals , Biomechanical Phenomena , Cell Proliferation , Cells, Cultured , Humans , Hyperplasia , Mice , Papilloma/etiology , Signal Transduction , rho-Associated Kinases/analysis , rho-Associated Kinases/genetics , rho-Associated Kinases/physiology
10.
Cell Res ; 21(4): 666-82, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21079653

ABSTRACT

The central arbiter of cell fate in response to DNA damage is p53, which regulates the expression of genes involved in cell cycle arrest, survival and apoptosis. Although many responses initiated by DNA damage have been characterized, the role of actin cytoskeleton regulators is largely unknown. We now show that RhoC and LIM kinase 2 (LIMK2) are direct p53 target genes induced by genotoxic agents. Although RhoC and LIMK2 have well-established roles in actin cytoskeleton regulation, our results indicate that activation of LIMK2 also has a pro-survival function following DNA damage. LIMK inhibition by siRNA-mediated knockdown or selective pharmacological blockade sensitized cells to radio- or chemotherapy, such that treatments that were sub-lethal when administered singly resulted in cell death when combined with LIMK inhibition. Our findings suggest that combining LIMK inhibitors with genotoxic therapies could be more efficacious than single-agent administration, and highlight a novel connection between actin cytoskeleton regulators and DNA damage-induced cell survival mechanisms.


Subject(s)
Actins/metabolism , Gene Expression Regulation , Lim Kinases/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Cytoskeleton , DNA Damage , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunoblotting , Mice , Microarray Analysis , RNA, Small Interfering , Signal Transduction/drug effects , Transcription, Genetic , rho-Associated Kinases/metabolism , rhoC GTP-Binding Protein
11.
Transcription ; 2(5): 211-5, 2011.
Article in English | MEDLINE | ID: mdl-22231116

ABSTRACT

Signaling through the Rho family of small GTPases regulates a variety of cellular processes via changes in the actin cytoskeleton. Here we discuss recent findings that show the transcription factor p53 regulates the expression of several Rho pathway signaling molecules, and how mutation of p53 in cancer dramatically alters signaling output through this pathway.


Subject(s)
Signal Transduction , Transcription, Genetic , rho GTP-Binding Proteins/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Apoptosis , Cytoskeleton/metabolism , DNA Damage , Humans , Lim Kinases/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
12.
J Cell Biol ; 191(1): 169-85, 2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20876278

ABSTRACT

LIM kinases 1 and 2 (LIMK1/2) are centrally positioned regulators of actin cytoskeleton dynamics. Using siRNA-mediated knockdown or a novel small molecule inhibitor, we show LIMK is required for path generation by leading tumor cells and nontumor stromal cells during collective tumor cell invasion. LIMK inhibition lowers cofilin phosphorylation, F-actin levels, serum response factor transcriptional activity and collagen contraction, and reduces invasion in three-dimensional invasion assays. Although motility was unaffected, LIMK inhibition impairs matrix protein degradation and invadopodia formation associated with significantly faster recovery times in FRAP assays indicative of reduced F-actin stability. When LIMK is knocked down in MDA-MB-231 cells, they lose the ability to lead strands of collectively invading cells. Similarly, when LIMK activity is blocked in cancer-associated fibroblasts, they are unable to lead the collective invasion of squamous carcinoma cells in an organotypic skin model. These results show that LIMK is required for matrix remodeling activities for path generation by leading cells in collective invasion.


Subject(s)
Lim Kinases/physiology , Neoplasm Invasiveness , Stromal Cells/enzymology , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Lim Kinases/antagonists & inhibitors , Phosphorylation , Protein Stability , RNA Interference
13.
Cancer Cell ; 14(5): 349-51, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18977323

ABSTRACT

Switching between elongated and rounded modes of movement allows invasive tumor cells to adapt to varying microenvironments. In a recent issue of Cell, Sanz-Moreno et al. identify DOCK3, NEDD9, WAVE2, and ARHGAP22 as key molecules regulating Rac and Rho signaling that determine the mode of movement driving melanoma cell metastasis.


Subject(s)
Cell Movement/physiology , Melanoma/pathology , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , Humans , Melanoma/metabolism , Melanoma/secondary , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism
14.
Mol Cell Biol ; 26(12): 4612-27, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16738326

ABSTRACT

The members of the Rho GTPase family are well known for their regulation of actin cytoskeletal structures. In addition, they influence progression through the cell cycle. The RhoA and RhoC proteins regulate numerous effector proteins, with a central and vital signaling role mediated by the ROCK I and ROCK II serine/threonine kinases. The requirement for ROCK function in the proliferation of numerous cell types has been revealed by studies utilizing ROCK-selective inhibitors such as Y-27632. However, the mechanisms by which ROCK signaling promotes cell cycle progression have not been thoroughly characterized. Using a conditionally activated ROCK-estrogen receptor fusion protein, we found that ROCK activation is sufficient to stimulate G1/S cell cycle progression in NIH 3T3 mouse fibroblasts. Further analysis revealed that ROCK acts via independent pathways to alter the levels of cell cycle regulatory proteins: cyclin D1 and p21(Cip1) elevation via Ras and the mitogen-activated protein kinase pathway, increased cyclin A via LIM kinase 2, and reduction of p27(Kip1) protein levels. Therefore, the influence of ROCK on cell cycle regulatory proteins occurs by multiple independent mechanisms.


Subject(s)
Cyclin A/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Cycle/physiology , Cell Line , Focal Adhesions , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Mice , Models, Biological , NIH 3T3 Cells , Recombinant Fusion Proteins/metabolism , Signal Transduction , rho-Associated Kinases
15.
Methods Enzymol ; 406: 541-53, 2006.
Article in English | MEDLINE | ID: mdl-16472686

ABSTRACT

The Rho-associated kinases ROCK I and ROCK II are serine/threonine kinases that play central and critical roles in regulating the actin cytoskeleton. Activation of ROCK proteins contributes positively to the phosphorylation of myosin II light chains (MLC), myosin ATPase activity, stabilization of filamentous actin, and coupling of the actin-myosin filaments to the plasma membrane, thereby leading to the increased actin-myosin force generation and contractility. We have constructed a conditionally-activated form of ROCK II (called ROCK:ER) by fusing the ROCK II kinase domain to the estrogen receptor hormone-binding domain. In this chapter, we describe the construction and characterization of this regulatable ROCK:ER fusion protein.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Receptors, Estrogen/metabolism , Animals , Cattle , Cell Line , Green Fluorescent Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , NIH 3T3 Cells , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , rho-Associated Kinases
16.
Mol Cell Biol ; 26(4): 1480-95, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449658

ABSTRACT

The transcription factor AP-1, which is composed of Fos and Jun family proteins, plays an essential role in tumor cell invasion by altering gene expression. We report here that Krp1, the AP-1 up-regulated protein that has a role in pseudopodial elongation in v-Fos-transformed rat fibroblast cells, forms a novel interaction with the nondifferentially expressed actin binding protein Lasp-1. Krp1 and Lasp-1 colocalize with actin at the tips of pseudopodia, and this localization is maintained by continued AP-1 mediated down-regulation of fibronectin that in turn suppresses integrin and Rho-ROCK signaling and allows pseudopodial protrusion and mesenchyme-like invasion. Mutation analysis of Lasp-1 demonstrates that its SH3 domain is necessary for pseudopodial extension and invasion. The results support the concept of an AP-1-regulated multigenic invasion program in which proteins encoded by differentially expressed genes direct the function, localization, and activity of proteins that are not differentially expressed to enhance the invasiveness of cells.


Subject(s)
Carrier Proteins/metabolism , Fibronectins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factor AP-1/metabolism , Animals , Carrier Proteins/genetics , Cell Line , Cell Transformation, Neoplastic , Cytoskeletal Proteins , Fibronectins/genetics , Genes, fos , Intracellular Signaling Peptides and Proteins , Mesoderm/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Models, Biological , Mutagenesis, Site-Directed , Neoplasm Invasiveness , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Pseudopodia/metabolism , RNA, Small Interfering/genetics , Rats , Signal Transduction , Transcription Factor AP-1/genetics , rho-Associated Kinases
17.
J Cell Biol ; 168(2): 245-55, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657395

ABSTRACT

Membrane blebbing during the apoptotic execution phase results from caspase-mediated cleavage and activation of ROCK I. Here, we show that ROCK activity, myosin light chain (MLC) phosphorylation, MLC ATPase activity, and an intact actin cytoskeleton, but not microtubular cytoskeleton, are required for disruption of nuclear integrity during apoptosis. Inhibition of ROCK or MLC ATPase activity, which protect apoptotic nuclear integrity, does not affect caspase-mediated degradation of nuclear proteins such as lamins A, B1, or C. The conditional activation of ROCK I was sufficient to tear apart nuclei in lamin A/C null fibroblasts, but not in wild-type fibroblasts. Thus, apoptotic nuclear disintegration requires actin-myosin contractile force and lamin proteolysis, making apoptosis analogous to, but distinct from, mitosis where nuclear disintegration results from microtubule-based forces and from lamin phosphorylation and depolymerization.


Subject(s)
Actins/metabolism , Apoptosis/physiology , Cell Nucleus/metabolism , Lamins/metabolism , Myosins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amides/pharmacology , Animals , Apoptosis/drug effects , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cycloheximide/pharmacology , Cytochalasin D/pharmacology , Cytoskeletal Proteins , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Intracellular Signaling Peptides and Proteins , Lamins/genetics , Lim Kinases , Mice , Microscopy, Electron, Transmission , Microtubules/drug effects , Microtubules/metabolism , Mutation/physiology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Myosins/antagonists & inhibitors , NIH 3T3 Cells , Nocodazole/pharmacology , Nuclear Lamina/drug effects , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyridines/pharmacology , Transfection , Tumor Necrosis Factor-alpha/pharmacology , rho-Associated Kinases
18.
Cancer Res ; 64(24): 8994-9001, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604264

ABSTRACT

Progression of tumors to invasive and metastatic forms requires that tumor cells undergo dramatic morphologic changes, a process regulated by Rho GTPases. Elevated expression of RhoA and RhoC, as well as the Rho effector proteins ROCK I and ROCK II, are commonly observed in human cancers and are often associated with more invasive and metastatic phenotypes. To examine how ROCK contributes to the progression of solid tumors, we established a conditionally activated form of ROCK II by fusing the kinase domain to the estrogen receptor hormone-binding domain (ROCK:ER). ROCK:ER-expressing colon carcinoma cells grown as tumors in immunocompromised nude mice organized into discrete clusters surrounding blood vessels. However, ROCK:ER activation resulted in the aggressive dissemination of tumor cells into the surrounding stroma, indicating that increased ROCK signaling is sufficient to promote invasion from solid tumors. In addition, tumors in which ROCK:ER was activated were more highly vascularized, indicating that ROCK contributes to tumor angiogenesis. ROCK:ER activation resulted in changes to epithelial morphology and organization that facilitated motility in vitro, likely by inducing the redistribution of proteins such as ezrin, as well as adherens junction and extracellular matrix-binding proteins. These results suggest that ROCK inhibitors would be useful antimetastatic and antiangiogenic chemotherapeutic agents in tumors associated with elevated RhoA, RhoC, ROCK I, or ROCK II expression.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Tamoxifen/analogs & derivatives , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cytoskeleton/enzymology , Cytoskeleton/pathology , Enzyme Activation , HCT116 Cells , Humans , Hyaluronan Receptors/metabolism , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tamoxifen/pharmacology , rho-Associated Kinases
19.
Curr Biol ; 13(24): 2185-9, 2003 Dec 16.
Article in English | MEDLINE | ID: mdl-14680635

ABSTRACT

The epidermis comprises multiple layers of specialized epithelial cells called keratinocytes. As cells are lost from the outermost epidermal layers, they are replaced through terminal differentiation, in which keratinocytes of the basal layer cease proliferating, migrate upwards, and eventually reach the outermost cornified layers. Normal homeostasis of the epidermis requires that the balance between proliferation and differentiation be tightly regulated. The GTP binding protein RhoA plays a fundamental role in the regulation of the actin cytoskeleton and in the adhesion events that are critically important to normal tissue homeostasis. Two central mediators of the signals from RhoA are the ROCK serine/threonine kinases ROCK-I and ROCK-II. We have analyzed ROCK's role in the regulation of epidermal keratinocyte function by using a pharmacological inhibitor and expressing conditionally active or inactive forms of ROCK-II in primary human keratinocytes. We report that blocking ROCK function results in inhibition of keratinocyte terminal differentiation and an increase in cell proliferation. In contrast, activation of ROCK-II in keratinocytes results in cell cycle arrest and an increase in the expression of a number of genes associated with terminal differentiation. Thus, these results indicate that ROCK plays a critical role in regulating the balance between proliferation and differentiation in human keratinocytes.


Subject(s)
Cell Differentiation/physiology , Keratinocytes/physiology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism , Amides/metabolism , Blotting, Western , Cell Cycle/physiology , Electrophoresis, Polyacrylamide Gel , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins , Keratinocytes/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/metabolism , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...