Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Gastroenterol Hepatol ; 34(10): 1766-1776, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30972813

ABSTRACT

BACKGROUND: Hyaluronidases (HAases), enzymes that degrade hyaluronan, have been widely investigated in cancer biology. However, whether HAases serve as tumor promoters or suppressors has been controversial in different cancers, and the exact role of HAases in colorectal cancer (CRC) has not been elucidated. METHODS: The expression levels of HYAL1, HYAL2, and HYAL3 in cancer and corresponding normal tissues from CRC patients were examined via immunohistochemistry. Then the correlation between HAases levels and pathological characteristics of CRC patients was analyzed. To verify the clinical data, HYAL1 and HYAL2 were downregulated or overexpressed in colon cancer cells LOVO and HCT116 to observe their influences on cell invasion and migration. For the mechanism study, we investigated the effects of HYAL1 and HYAL2 on the expression of matrix metalloproteases (MMPs)/tissue inhibitor of metalloproteases (TIMPs) and distribution of F-actin. RESULTS: All the three HAases were abnormally elevated in cancer tissues. Interestingly, HYAL1 and HYAL2, but not HYAL3, were negatively correlated with lymphatic metastasis and TNM stage. When HYAL1 and HYAL2 were knocked down, the invasion and migration abilities of colon cancer cells were accelerated, whereas overexpression of HYAL1 and HYAL2 had the opposite effects. In addition, colon cancer cells with HYAL1 and HYAL2 downregulation showed increased levels of MMP2 and MMP9, decreased levels of TIMP1 and TIMP2, and more intense F-actin stress fibers. CONCLUSIONS: Our study suggests that HYAL1 and HYAL2 suppress CRC metastasis through regulating MMPs/TIMPs balance and rearranging F-actin distribution, further inhibiting invasion and migration of cancer cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement , Colorectal Neoplasms/enzymology , Hyaluronoglucosaminidase/metabolism , Aged , Cell Adhesion Molecules/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Hyaluronoglucosaminidase/genetics , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Stress Fibers/enzymology , Stress Fibers/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
2.
Fertil Steril ; 107(3): 803-812, 2017 03.
Article in English | MEDLINE | ID: mdl-28069177

ABSTRACT

OBJECTIVE: To study the impact of integrin-linked kinase (ILK) in endometrial stromal cells (ESCs) during decidualization. DESIGN: Laboratory study with the use of human endometrium. SETTING: University hospital. PATIENT(S): Fertile reproductive-age women who had not received hormonal treatment for 3 months before tissue collection. INTERVENTION(S): Endometrium tissue collection, in vitro decidualization of isolated ESCs, and small interfering (si) RNA transfection. MAIN OUTCOME MEASURE(S): Immunohistochemistry, ELISA, Western blot analysis, methylthiazolyl tetrazolium assay, and immunofluorescence staining. RESULT(S): In vivo expression of ILK is significantly increased in distended-fusiform stromal cells of late secretory endometrium and in cobblestone-shaped decidual cells of early pregnancy. During in vitro decidualization for up to 8 days, confluent cultures of isolated ESCs consistently displayed increased ILK expression and morphologic transformation from fibroblast-like to polygonal cells. Subsequent ILK knockdown by siRNA transfection reversed this transformation, accompanied by decreased phosphorylation of glycogen synthase kinase (GSK) 3ß and decreased viable cell numbers. Immunofluorescence staining of the decidualized ESCs demonstrated linkage of increased levels of ILK at the tips of the fan-shaped organization of actin stress fibers located in the submembranous area, which expanded the decidual cells into a typical polygonal appearance. Knock-down of ILK abrogated the polymerization and organization of actin fibers, which reverted the cells to their undecidualized morphology. CONCLUSION(S): During human endometrial decidualization, ILK is essential for morphologic transformation of ESCs through organization of the actin cytoskeleton; it may also function through subsequent GSK3ß signaling, which requires further studies.


Subject(s)
Cell Movement , Cell Shape , Decidua/enzymology , Embryo Implantation , Protein Serine-Threonine Kinases/metabolism , Stromal Cells/enzymology , Actins/metabolism , Cell Survival , Cells, Cultured , Decidua/pathology , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Menstrual Cycle/metabolism , Phosphorylation , Pregnancy , Protein Serine-Threonine Kinases/genetics , RNA Interference , Stress Fibers/enzymology , Stromal Cells/pathology , Time Factors , Transfection
3.
J Cell Physiol ; 232(9): 2447-2460, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27591737

ABSTRACT

Glaucoma, a leading cause of irreversible blindness, is commonly associated with elevated intraocular pressure (IOP) due to impaired aqueous humor (AH) drainage through the trabecular meshwork (TM). Although dysregulated production and organization of extracellular matrix (ECM) is presumed to increase resistance to AH outflow and elevate IOP by altering TM cell contractile and adhesive properties, it is not known whether regulation of ECM protein phosphorylation via the secretory vertebrate lonesome kinase (VLK) influences TM cellular characteristics. Here, we tested this possibility. Experiments carried out in this study reveal that the 32 kDa protein is a prominent VLK isoform detectable in lysates and conditioned media (CM) of human TM cells. Increased levels of VLK were observed in CM of TM cells subjected to cyclic mechanical stretch, or treated with dexamethasone, TGF-ß2, and TM cells expressing constitutively active RhoA GTPase. Downregulation of VLK expression in TM cells using siRNA decreased tyrosine phosphorylation (TyrP) of ECM proteins and focal adhesions, and induced changes in cell shape in association with reduced levels of actin stress fibers and phospho-paxillin. VLK was also demonstrated to regulate TGF-ß2-induced TyrP of ECM proteins. Taken together, these results suggest that VLK secretion can be regulated by external cues, intracellular signal proteins, and mechanical stretch, and VLK can in turn regulate TyrP of ECM proteins secreted by TM cells and control cell shape, actin stress fibers, and focal adhesions. These observations indicate a potential role for VLK in homeostasis of AH outflow and IOP, and in the pathobiology of glaucoma. J. Cell. Physiol. 232: 2447-2460, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Adhesion , Cell Shape , Extracellular Matrix Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Trabecular Meshwork/enzymology , Adult , Aged , Aqueous Humor/metabolism , Cell Adhesion/drug effects , Cell Shape/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , Dexamethasone/pharmacology , Focal Adhesions/enzymology , Glaucoma/enzymology , Glaucoma/pathology , Glaucoma/physiopathology , Humans , Intraocular Pressure , Mechanotransduction, Cellular , Middle Aged , Mutation , Paxillin/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , RNA Interference , Stress Fibers/enzymology , Time Factors , Trabecular Meshwork/drug effects , Transfection , Transforming Growth Factor beta2/pharmacology , Tyrosine , Young Adult , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
4.
Am J Physiol Cell Physiol ; 309(1): C51-9, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25948734

ABSTRACT

The role of the actin cytoskeleton in endothelial barrier function has been debated for nearly four decades. Our previous investigation revealed spontaneous local lamellipodia in confluent endothelial monolayers that appear to increase overlap at intercellular junctions. We tested the hypothesis that the barrier-disrupting agent histamine would reduce local lamellipodia protrusions and investigated the potential involvement of p38 mitogen-activated protein (MAP) kinase activation and actin stress fiber formation. Confluent monolayers of human umbilical vein endothelial cells (HUVEC) expressing green fluorescent protein-actin were studied using time-lapse fluorescence microscopy. The protrusion and withdrawal characteristics of local lamellipodia were assessed before and after addition of histamine. Changes in barrier function were determined using electrical cell-substrate impedance sensing. Histamine initially decreased barrier function, lamellipodia protrusion frequency, and lamellipodia protrusion distance. A longer time for lamellipodia withdrawal and reduced withdrawal distance and velocity accompanied barrier recovery. After barrier recovery, a significant number of cortical fibers migrated centrally, eventually resembling actin stress fibers. The p38 MAP kinase inhibitor SB203580 attenuated the histamine-induced decreases in barrier function and lamellipodia protrusion frequency. SB203580 also inhibited the histamine-induced decreases in withdrawal distance and velocity, and the subsequent actin fiber migration. These data suggest that histamine can reduce local lamellipodia protrusion activity through activation of p38 MAP kinase. The findings also suggest that local lamellipodia have a role in maintaining endothelial barrier integrity. Furthermore, we provide evidence that actin stress fiber formation may be a reaction to, rather than a cause of, reduced endothelial barrier integrity.


Subject(s)
Cell Movement/drug effects , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Pseudopodia/drug effects , Stress Fibers/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Electric Impedance , Enzyme Activation , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Microscopy, Fluorescence , Microscopy, Video , Permeability , Protein Kinase Inhibitors/pharmacology , Pseudopodia/enzymology , Signal Transduction/drug effects , Stress Fibers/enzymology , Time Factors , Time-Lapse Imaging , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
5.
Vascul Pharmacol ; 70: 45-54, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869521

ABSTRACT

Rho kinase mediates the effects of inflammatory permeability factors by increasing actomyosin-generated traction forces on endothelial adherens junctions, resulting in disassembly of intercellular junctions and increased vascular leakage. In vitro, this is accompanied by the Rho kinase-driven formation of prominent radial F-actin fibers, but the in vivo relevance of those F-actin fibers has been debated, suggesting other Rho kinase-mediated events to occur in vascular leak. Here, we delineated the contributions of the highly homologous isoforms of Rho kinase (ROCK1 and ROCK2) to vascular hyperpermeability responses. We show that ROCK2, rather than ROCK1 is the critical Rho kinase for regulation of thrombin receptor-mediated vascular permeability. Novel traction force mapping in endothelial monolayers, however, shows that ROCK2 is not required for the thrombin-induced force enhancements. Rather, ROCK2 is pivotal to baseline junctional tension as a novel mechanism by which Rho kinase primes the endothelium for hyperpermeability responses, independent from subsequent ROCK1-mediated contractile stress-fiber formation during the late phase of the permeability response.


Subject(s)
Capillary Permeability , Endothelial Cells/enzymology , Intercellular Junctions/enzymology , rho-Associated Kinases/metabolism , Animals , Capillary Permeability/drug effects , Cells, Cultured , Electric Impedance , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Intercellular Junctions/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction , Stress Fibers/enzymology , Thrombin/pharmacology , Time Factors , Transfection , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
6.
BMC Physiol ; 14: 5, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25185746

ABSTRACT

BACKGROUND: MAPK-activated protein kinase 2 (MK2) plays a pivotal role in the cell response to (inflammatory) stress. Among others, MK2 is known to be involved in the regulation of cytokine mRNA metabolism and regulation of actin cytoskeleton dynamics. Previously, MK2-deficient mice were shown to be highly resistant to LPS/d-Galactosamine-induced hepatitis. Additionally, research in various disease models has indicated the kinase as an interesting inhibitory drug target for various acute or chronic inflammatory diseases. RESULTS: We show that in striking contrast to the known resistance of MK2-deficient mice to a challenge with LPS/D-Gal, a low dose of tumor necrosis factor (TNF) causes hyperacute mortality via an oxidative stress driven mechanism. We identified in vivo defects in the stress fiber response in endothelial cells, which could have resulted in reduced resistance of the endothelial barrier to deal with exposure to oxidative stress. In addition, MK2-deficient mice were found to be more sensitive to cecal ligation and puncture-induced sepsis. CONCLUSIONS: The capacity of the endothelial barrier to deal with inflammatory and oxidative stress is imperative to allow a regulated immune response and maintain endothelial barrier integrity. Our results indicate that, considering the central role of TNF in pro-inflammatory signaling, therapeutic strategies examining pharmacological inhibition of MK2 should take potentially dangerous side effects at the level of endothelial barrier integrity into account.


Subject(s)
Inflammation/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/toxicity , Animals , Capillary Permeability , Endothelial Cells/enzymology , Inflammation/chemically induced , Inflammation/mortality , Intracellular Signaling Peptides and Proteins/genetics , Kidney/enzymology , Lipopolysaccharides , Liver/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Sepsis/enzymology , Sepsis/mortality , Stress Fibers/enzymology
7.
Microvasc Res ; 95: 94-102, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25072537

ABSTRACT

Disruption of the pulmonary endothelial barrier and subsequent vascular leak is a hallmark of acute lung injury. Dynamic rearrangements in the endothelial cell (EC) peripheral membrane and underlying cytoskeleton are critical determinants of barrier function. The cytoskeletal effector protein non-muscle myosin light chain kinase (nmMLCK) and the actin-binding regulatory protein cortactin are important regulators of the endothelial barrier. In the present study we functionally characterize a proline-rich region of nmMLCK previously identified as the possible site of interaction between nmMLCK and cortactin. A mutant nmMLCK construct deficient in proline residues at the putative sites of cortactin binding (amino acids 973, 976, 1019, 1022) was generated. Co-immunoprecipitation studies in human lung EC transfected with wild-type or mutant nmMLCK demonstrated similar levels of cortactin interaction at baseline and after stimulation with the barrier-enhancing agonist, sphingosine 1-phosphate (S1P). In contrast, binding studies utilizing recombinant nmMLCK fragments containing the wild-type or proline-deficient sequence demonstrated a two-fold increase in cortactin binding (p<0.01) to the mutant construct. Immunofluorescent microscopy revealed an increased stress fiber density in ECs expressing GFP-labeled mutant nmMLCK at baseline (p=0.02) and after thrombin (p=0.01) or S1P (p=0.02) when compared to wild-type. Mutant nmMLCK demonstrated an increase in kinase activity in response to thrombin (p<0.01). Kymographic analysis demonstrated an increased EC membrane retraction distance and velocity (p<0.01) in response to the barrier disrupting agent thrombin in cells expressing the mutant vs. the wild-type nmMLCK construct. These results provide evidence that critical prolines within nmMLCK (amino acids 973, 976, 1019, 1022) regulate cytoskeletal and membrane events associated with pulmonary endothelial barrier function.


Subject(s)
Cytoskeleton/enzymology , Endothelial Cells/enzymology , Lung/blood supply , Myosin-Light-Chain Kinase/metabolism , Antigens, CD/metabolism , Binding Sites , Cadherins/metabolism , Capillary Permeability , Cell Membrane/enzymology , Cells, Cultured , Cortactin/metabolism , Humans , Immunoprecipitation , Kymography , Lysophospholipids/metabolism , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/genetics , Proline-Rich Protein Domains , Protein Interaction Domains and Motifs , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Stress Fibers/enzymology , Thrombin/metabolism , Time Factors , Transfection
8.
Eur J Pharm Sci ; 55: 46-57, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24486483

ABSTRACT

Diacylglycerol (DAG) is a central mediator of signaling pathways that regulate cell proliferation, survival and apoptosis. Therefore, C1 domain, the DAG binding site within protein kinase C (PKC) and other DAG effector proteins, is considered a potential cancer drug target. Derivatives of 5-(hydroxymethyl)isophthalic acid are a novel group of C1 domain ligands with antiproliferative and differentiation-inducing effects. Our previous work showed that these isophthalate derivatives exhibit antiproliferative and elongation-inducing effects in HeLa human cervical cancer cells. In this study we further characterized the effects of bis(3-trifluoromethylbenzyl) 5-(hydroxymethyl)isophthalate (HMI-1a3) on HeLa cell proliferation and morphology. HMI-1a3-induced cell elongation was accompanied with loss of focal adhesions and actin stress fibers, and exposure to HMI-1a3 induced a prominent relocation of cofilin-1 into the nucleus regardless of cell phenotype. The antiproliferative and morphological responses to HMI-1a3 were not modified by pharmacological inhibition or activation of PKC, or by RNAi knock-down of specific PKC isoforms, suggesting that the effects of HMI-1a3 were not mediated by PKC. Genome-wide gene expression microarray and gene set enrichment analysis suggested that, among others, HMI-1a3 induces changes in small GTPase-mediated signaling pathways. Our experiments revealed that the isophthalates bind also to the C1 domains of ß2-chimaerin, protein kinase D (PKD) and myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK), which are potential mediators of small GTPase signaling and cytoskeletal reorganization. Pharmacological inhibition of MRCK, but not that of PKD attenuated HMI-1a3-induced cell elongation, suggesting that MRCK participates in mediating the effects of HMI-1a3 on HeLa cell morphology.


Subject(s)
Cell Shape/drug effects , Phthalic Acids/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Cell Proliferation/drug effects , Cofilin 1/metabolism , Dose-Response Relationship, Drug , Focal Adhesions/drug effects , Focal Adhesions/enzymology , HeLa Cells , Humans , Myotonin-Protein Kinase , Phenotype , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , Stress Fibers/drug effects , Stress Fibers/enzymology , Time Factors , Transfection
9.
Toxicol Lett ; 225(1): 12-9, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24291038

ABSTRACT

The exposure to particulate matter with a mean aerodynamic diameter ≤10 µm (PM10) from urban zones is considered to be a risk factor in the development of cancer. The aim of this work was to determine if PM10 exposure induces factors related to the acquisition of a neoplastic phenotype, such as cytoskeletal remodeling, changes in the subcellular localization of p21(CIP1/WAF1), an increase in ß-galactosidase activity and changes in cell cycle. To test our hypothesis, PM10 from an industrial zone (IZ) and a commercial zone (CZ) were collected, and human adenocarcinoma lung cell cultures (A549) were exposed to a sublethal PM10 concentration (10 µg/cm(2)) for 24 h and 48 h. The results showed that PM10 exposure induced an increase in F-actin stress fibers and caused the cytoplasmic stabilization of p21(CIP1/WAF1) via phosphorylation at Thr(145) and Ser(146) and the phosphorylation of ERK1/2 on Thr(202). Changes in the cell cycle or apoptosis were not observed, but an increase in ß-galactosidase activity was detected. The PM10 from CZ caused more dramatic effects in lung cells. We conclude that PM10 exposure induced cytoplasmic p21(CIP1/WAF1) retention, ERK1/2 activation, cytoskeleton remodeling and the acquisition of a senescence-like phenotype in lung cells. These alterations could have mechanistic implications regarding the carcinogenic potential of PM10.


Subject(s)
Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoskeleton/drug effects , Lung/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Particulate Matter/toxicity , Actins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytoplasm/enzymology , Cytoskeleton/enzymology , Cytoskeleton/pathology , Enzyme Activation , Humans , Lung/enzymology , Lung/pathology , Particle Size , Phenotype , Phosphorylation , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/enzymology , Stress Fibers/pathology , Time Factors , beta-Galactosidase/metabolism
10.
Circ Res ; 110(11): 1423-34, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22539766

ABSTRACT

RATIONALE: RhoA and Rho kinase contribute to pulmonary vasoconstriction and vascular remodeling in pulmonary hypertension. RhoB, a protein homologous to RhoA and activated by hypoxia, regulates neoplastic growth and vasoconstriction but its role in the regulation of pulmonary vascular function is not known. OBJECTIVE: To determine the role of RhoB in pulmonary endothelial and smooth muscle cell responses to hypoxia and in pulmonary vascular remodeling in chronic hypoxia-induced pulmonary hypertension. METHODS AND RESULTS: Hypoxia increased expression and activity of RhoB in human pulmonary artery endothelial and smooth muscle cells, coincidental with activation of RhoA. Hypoxia or adenoviral overexpression of constitutively activated RhoB increased actomyosin contractility, induced endothelial permeability, and promoted cell growth; dominant negative RhoB or manumycin, a farnesyltransferase inhibitor that targets the vascular function of RhoB, inhibited the effects of hypoxia. Coordinated activation of RhoA and RhoB maximized the hypoxia-induced stress fiber formation caused by RhoB/mammalian homolog of Drosophila diaphanous-induced actin polymerization and RhoA/Rho kinase-induced phosphorylation of myosin light chain on Ser19. Notably, RhoB was specifically required for hypoxia-induced factor-1α stabilization and for hypoxia- and platelet-derived growth factor-induced cell proliferation and migration. RhoB deficiency in mice markedly attenuated development of chronic hypoxia-induced pulmonary hypertension, despite compensatory expression of RhoA in the lung. CONCLUSIONS: RhoB mediates adaptational changes to acute hypoxia in the vasculature, but its continual activation by chronic hypoxia can accentuate vascular remodeling to promote development of pulmonary hypertension. RhoB is a potential target for novel approaches (eg, farnesyltransferase inhibitors) aimed at regulating pulmonary vascular tone and structure.


Subject(s)
Endothelial Cells/enzymology , Hypertension, Pulmonary/etiology , Hypoxia/complications , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , rhoB GTP-Binding Protein/metabolism , Actomyosin/genetics , Actomyosin/metabolism , Animals , Capillary Permeability , Cell Hypoxia , Cell Movement , Cell Proliferation , Cells, Cultured , Chronic Disease , Disease Models, Animal , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Familial Primary Pulmonary Hypertension , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/genetics , Hypoxia/enzymology , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myosin Light Chains/metabolism , Phosphorylation , Polyenes/pharmacology , Polyunsaturated Alkamides/pharmacology , Pulmonary Artery/enzymology , RNA Interference , Serine , Stress Fibers/enzymology , Time Factors , Transfection , Vasoconstriction , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/deficiency , rhoB GTP-Binding Protein/genetics
11.
Biochem Biophys Res Commun ; 420(3): 662-5, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22450311

ABSTRACT

A nanoneedle, an atomic force microscope (AFM) tip etched to 200 nm in diameter and 10 µm in length, can be inserted into cells with the aid of an AFM and has been used to introduce functional molecules into cells and to analyze intracellular information with minimal cell damage. However, some cell lines have shown low insertion efficiency of the nanoneedle. Improvement in the insertion efficiency of a nanoneedle into such cells is a significant issue for nanoneedle-based cell manipulation and analysis. Here, we have formed nanofilms composed of extracellular matrix molecules on cell surfaces and found that the formation of the nanofilms improved insertion efficiency of a nanoneedle into fibroblast and neural cells. The nanofilms were shown to improve insertion efficiency even in cells in which the formation of actin stress fibers was inhibited by the ROCK inhibitor Y27632, suggesting that the nanofilms with the mesh structure directly contributed to the improved insertion efficiency of a nanoneedle.


Subject(s)
Cell Membrane/chemistry , Cells/chemistry , Nanostructures/chemistry , Amides/pharmacology , Animals , BALB 3T3 Cells , Enzyme Inhibitors/pharmacology , Extracellular Matrix Proteins/chemistry , Mice , Microscopy, Atomic Force , Neurons/chemistry , Pyridines/pharmacology , Stress Fibers/chemistry , Stress Fibers/drug effects , Stress Fibers/enzymology , rho-Associated Kinases/antagonists & inhibitors
12.
Biochem Biophys Res Commun ; 418(3): 500-5, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22281493

ABSTRACT

Cell shape change and cytoskeletal reorganization are known to be involved in the chondrogenesis. Negative role of RhoA, a cytoskeleton-regulating protein, and its downstream target, Rho-associated protein kinase (ROCK) in the chondrogenesis has been studied in many different culture systems including primary chondrocytes, chondrogenic cell lines, dedifferentiated chondrocytes, and micromass culture of mesenchymal cells. To further investigate the role of RhoA and ROCK in the chondrogenesis, we examined the RhoA-ROCK-myosin light chains (MLC) pathway in low density culture of chick limb bud mesenchymal cells. We observed for the first time that inhibition of RhoA by C3 cell-permeable transferase, CT04, induced chondrogenesis of undifferentiated mesenchymal single cells following dissolution of actin stress fibers. Inhibition of RhoA activity by CT04 was confirmed by pull down assay using the Rho-GTP binding domain of Rhotekin. CT04 also inhibited ROCK activity. In contrast, inhibition of ROCK by Y27632 neither altered the actin stress fibers nor induced chondrogenesis. In addition, inhibition of RhoA or ROCK did not affect the phosphorylation of MLC. Inhibition of myosin light chain kinase (MLCK) by ML-7 or inhibition of myosin ATPase with blebbistatin dissolved actin stress fibers and induced chondrogenesis. ML-7 reduced the MLC phosphorylation. Taken together, our current study suggests that RhoA uses other pathway than ROCK/MLC in the modulation of actin stress fibers and chondrogenesis. Our data also imply that, irrespective of mechanisms, dissolution of actin stress fibers is crucial for chondrogenesis.


Subject(s)
Chondrogenesis , Mesoderm/enzymology , Stress Fibers/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Shape , Cells, Cultured , Chick Embryo , Extremities/embryology , Mesoderm/cytology , Myosin Light Chains/metabolism , Phosphorylation , Stress Fibers/enzymology , Transferases/metabolism , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
13.
Kidney Int ; 81(11): 1075-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22278020

ABSTRACT

Podocytes are highly differentiated cells that play an important role in maintaining glomerular filtration barrier integrity; a function regulated by small GTPase proteins of the Rho family. To investigate the role of Rho A in podocyte biology, we created transgenic mice expressing doxycycline-inducible constitutively active (V14 Rho) or dominant-negative Rho A (N19 Rho) in podocytes. Specific induction of either Rho A construct in podocytes caused albuminuria and foot process effacement along with disruption of the actin cytoskeleton as evidenced by decreased expression of the actin-associated protein synaptopodin. The mechanisms of these adverse effects, however, appeared to be different. Active V14 Rho enhanced actin polymerization, caused a reduction in nephrin mRNA and protein levels, promoted podocyte apoptosis, and decreased endogenous Rho A levels. In contrast, the dominant-negative N19 Rho caused a loss of podocyte stress fibers, did not alter the expression of either nephrin or Rho A, and did not cause podocyte apoptosis. Thus, our findings suggest that Rho A plays an important role in maintaining the integrity of the glomerular filtration barrier under basal conditions, but enhancement of Rho A activity above basal levels promotes podocyte injury.


Subject(s)
Albuminuria/etiology , Glomerular Filtration Barrier/enzymology , Podocytes/enzymology , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/enzymology , Albuminuria/enzymology , Albuminuria/genetics , Albuminuria/pathology , Animals , Apoptosis , Gene Expression Regulation , Genotype , Glomerular Filtration Barrier/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mutation , Phenotype , Podocytes/pathology , RNA, Messenger/metabolism , Stress Fibers/enzymology , Time Factors , rhoA GTP-Binding Protein/genetics
14.
Int J Oncol ; 37(4): 951-62, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20811717

ABSTRACT

Pak1 has been reported to be overexpressed in colorectal cancer, but the role of Pak1 in colorectal cancer remains unclear. In this study, Pak1 expression and activity were associated with aggressive behavior of colorectal cancer. Overexpression of Pak1 increased colorectal cancer cell motility and invasion, whereas down-regulation of Pak1 expression or activity reduced colorectal cancer cell migration and invasion. In addition, activated Pak1 inhibited stress fiber and focal adhesion complex formation in colorectal cancer cells and led to formation of motile phenotypes. Importantly, activated Pak1 elicited phosphorylation of FAK at Ser-910 via an ERK-dependent pathway in colorectal cancer cell lines and clinical samples. In conclusion, our results suggest that activated Pak1 regulates colorectal cancer metastasis requiring an ERK-dependent phosphorylation of FAK at Ser-910.


Subject(s)
Cell Movement , Colorectal Neoplasms/enzymology , Focal Adhesion Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , p21-Activated Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/secondary , Enzyme Activation , Female , Focal Adhesions/enzymology , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Phenotype , Phosphorylation , RNA Interference , Serine , Stress Fibers/enzymology , Transfection , p21-Activated Kinases/genetics
15.
Biochem Biophys Res Commun ; 401(3): 344-9, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20849825

ABSTRACT

Stress fiber realignment is an important adaptive response to cyclic stretch for nonmuscle cells, but the mechanism by which such reorganization occurs is not known. By analyzing stress fiber dynamics using live cell microscopy, we revealed that stress fiber reorientation perpendicular to the direction of cyclic uniaxial stretching at 1 Hz did not involve disassembly of the stress fiber distal ends located at focal adhesion sites. Instead, these distal ends were often used to assemble new stress fibers oriented progressively further away from the direction of stretch. Stress fiber disassembly and reorientation were not induced when the frequency of stretch was decreased to 0.01 Hz, however. Treatment with the Rho-kinase inhibitor Y27632 reduced stress fibers to thin fibers located in the cell periphery which bundled together to form thick fibers oriented parallel to the direction of stretching at 1Hz. In contrast, these thin fibers remained diffuse in cells subjected to stretch at 0.01 Hz. Cyclic stretch at 1 Hz also induced actin fiber formation parallel to the direction of stretch in cells treated with the myosin light chain kinase (MLCK) inhibitor ML-7, but these fibers were located centrally rather than peripherally. These results shed new light on the mechanism by which stress fibers reorient in response to cyclic stretch in different regions of the actin cytoskeleton.


Subject(s)
Myosin-Light-Chain Kinase/metabolism , Stress Fibers/physiology , Stress, Mechanical , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Azepines/pharmacology , Cattle , Cells, Cultured , Cytoskeleton/enzymology , Cytoskeleton/ultrastructure , Myosin-Light-Chain Kinase/antagonists & inhibitors , Naphthalenes/pharmacology , Pyridines/pharmacology , Stress Fibers/enzymology , rho-Associated Kinases/antagonists & inhibitors
16.
Cytoskeleton (Hoboken) ; 67(10): 666-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20737438

ABSTRACT

Directed cell migration requires precise spatial control of F-actin-based leading edge protrusion, focal adhesion (FA) dynamics, and actomyosin contractility. In spreading fibroblasts, the Abl family kinases, Abl and Arg, primarily localize to the nucleus and cell periphery, respectively. Here we provide evidence that Abl and Arg exert different spatial regulation on cellular contractile and adhesive structures. Loss of Abl function reduces FA, F-actin, and phosphorylated myosin light chain (pMLC) staining at the cell periphery, shifting the distribution of these elements more to the center of the cell than in wild-type (WT) and arg(-/-) cells. Conversely, loss of Arg function shifts the distribution of these contractile and adhesion elements more to the cell periphery relative to WT and abl(-/-) cells. Abl/Arg-dependent phosphorylation of p190RhoGAP (p190) promotes its binding to p120RasGAP (p120) to form a functional RhoA GTPase inhibitory complex, which attenuates RhoA activity and downstream pMLC and FA formation. p120 and p190 colocalize both in the central region and at the cell periphery in WT cells. This p120:p190 colocalization redistributes to a more peripheral distribution in abl(-/-) cells and to a more centralized distribution in arg(-/-) cells, and these altered distributions can be restored to WT patterns via re-expression of Abl or Arg, respectively. Thus, the altered p120:p190 distribution in the mutant cells correlates inversely with the redistribution in adhesions, actin, and pMLC staining in these cells. Our studies suggest that Abl and Arg exert different spatial regulation on actomyosin contractility and focal adhesions within cells.


Subject(s)
Fibroblasts/physiology , Focal Adhesions/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-abl/physiology , Stress Fibers/physiology , 3T3 Cells , Actin Cytoskeleton/metabolism , Actomyosin/metabolism , Animals , Cell Adhesion/physiology , Cytoskeleton/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesions/enzymology , Mice , Myosin Light Chains/metabolism , Stress Fibers/enzymology
17.
J Cell Sci ; 123(Pt 8): 1247-52, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20233848

ABSTRACT

Actin stress fibers are fundamental components of the actin cytoskeleton that produce contractile force in non-muscle cells. The formation of stress fibers is controlled by the small GTPase RhoA and two highly related proteins, RhoB and RhoC. Together, this subgroup of actin-regulatory proteins represents the canonical pathway of stress-fiber formation. Here, we show that the Rif GTPase is an alternative trigger of stress-fiber formation in epithelial cells. Rif is distantly related to RhoA; however, we show that the two proteins share a common downstream partner in stress-fiber formation--the Diaphanous-related formin mDia1. Rif-induced stress fibers also depend on the activity of the ROCK protein kinase. Unlike RhoA, Rif does not raise ROCK activity in cells, instead Rif appears to regulate the localization of myosin light chain phosphorylation. This study establishes Rif as a general regulator of Diaphanous-related formins and shows how non-classical Rho family members can access classical Rho pathways to create new signaling interfaces in cytoskeletal regulation.


Subject(s)
Actins/metabolism , Epithelial Cells/enzymology , Monomeric GTP-Binding Proteins/metabolism , Stress Fibers/enzymology , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Surface Extensions/enzymology , Epithelial Cells/cytology , Formins , HeLa Cells , Humans , Protein Binding , rho-Associated Kinases/metabolism
18.
J Cell Biochem ; 109(5): 906-14, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20069557

ABSTRACT

Cyclic strain is an important inducer of proliferation and migration of vascular smooth muscle cells (VSMCs) which are involved in vascular remodeling during hypertension. However, its mechanism remains to be elucidated. VSMCs of rat aorta were exposed to cyclic strains in vitro with defined parameters, the static, 5%-strain (physiological) and 15%-strain (pathological), at 1.25 Hz for 24 h respectively. Then the possible signaling molecules participated in strain-induced VSMC migration and proliferation were investigated. The results showed that 15%-strain significantly increased VSMC migration and proliferation in comparison with 5%-strain. Expression of Rho GDP dissociation inhibitor alpha (Rho-GDIalpha) was repressed by 15%-strain, but expressions of phospho-Rac1 and phospho-p38 were increased. Expressions of phospho-Akt and phospho-ERK1/2 were similar between the static, 5%-strain and 15%-strain groups. Rho-GDIalpha "knock-down" by target siRNA transfection increased migration and proliferation of VSMCs, and up-regulated phosphorylation of Rac1 and p38 in all groups. Rac1 "knock-down" repressed migration and proliferation of VSMCs, down-regulated phosphorylation of p38, but had no effect on Rho-GDIalpha expression. When siRNAs of Rho-GDIalpha and Rac1 were co-transfected to VSMCs, the expressions of Rho-GDIalpha and phospho-Rac1 were both decreased, and the effects of Rho-GDIalpha "knock-down" were blocked. Rho-GDIalpha "knock-down" promoted while Rac1 "knock-down" postponed the assembly of stress fibers and focal adhesions in static. The results demonstrate that the pathological cyclic strain might induce migration and proliferation of VSMCs via repressing expression of Rho-GDIalpha, which subsequently verified phosphorylations of Rac1 and p38.


Subject(s)
Cell Movement , Guanine Nucleotide Dissociation Inhibitors/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Stress, Mechanical , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions/enzymology , Male , Myocytes, Smooth Muscle/enzymology , Nonlinear Dynamics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Stress Fibers/enzymology , rho-Specific Guanine Nucleotide Dissociation Inhibitors
19.
Mol Cell Biol ; 29(21): 5963-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19667072

ABSTRACT

Lfc is a guanine nucleotide exchange factor (GEF) for Rho that demonstrates an unusual ability to associate with microtubules. While several phosphorylated residues have been detected in the Lfc polypeptide, the mechanism(s) by which phosphorylation regulates the exchange activity of Lfc remains unclear. We confirm that Lfc is a phosphorylated protein and demonstrate that 14-3-3 interacts directly and in a phosphorylation-dependent manner with Lfc. We identify AKAP121 as an Lfc-binding protein and show that Lfc is phosphorylated in an AKAP-dependent manner by protein kinase A (PKA). Forskolin treatment induced 14-3-3 binding to Lfc and suppressed the exchange activity of wild-type Lfc on RhoA. Importantly, a mutant of Lfc that is unable to associate with 14-3-3 proteins was resistant to inhibition by forskolin. Tctex-1, a dynein motor light chain, binds to Lfc in a competitive manner with 14-3-3.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , 14-3-3 Proteins/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Cell Line , Consensus Sequence , Dyneins , Enzyme Activation , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Rats , Rho Guanine Nucleotide Exchange Factors , Stress Fibers/enzymology , rhoA GTP-Binding Protein/metabolism , t-Complex Genome Region
20.
Cell Tissue Res ; 337(1): 167-75, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19421779

ABSTRACT

The expression pattern of diacylglycerol kinase (DGK) and the biological significance of DGKepsilon in vascular smooth muscle cells were investigated. mRNA expression for DGKalpha, DGKepsilon, and DGKzeta was detected in isolated rat aortic smooth muscle cells (RASMCs) and A7r5 cells by reverse transcription with polymerase chain reaction analysis. An immunocytochemical study revealed intense DGKepsilon in a filamentous pattern, parallel to the long axis of cell, and on actin stress fibers as shown by double-staining with fluorescent phalloidin. DGKalpha was detected sparsely in the cytoplasm and nucleus, and DGKzeta was observed as a granular pattern in the nucleus. In order to elucidate the functional significance of DGKepsilon, its immunoreactivity was examined in RASMCs incubated with serotonin, a vasoconstrictive agonist. When RASMCs were stimulated with serotonin, the cells lost their polarization and shortened, i.e., contracted. In RASMCs contracted by serotonin, DGKepsilon was detected diffusely in the cytoplasm without a filamentous stress fiber pattern. Protein and mRNA expression of DGKepsilon in RASMCs was significantly increased by stimulation with serotonin. Inhibition of Rho-associated kinases by Y-27632 or inhibition of actin polymerization by cytochalasin B resulted in a decrease in the intensity of DGKepsilon immunoreactivity on stress fibers. The results suggest that DGKepsilon interacts with actin stress fibers and is involved in their stability in vascular smooth muscle cells.


Subject(s)
Diacylglycerol Kinase/biosynthesis , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Stress Fibers/enzymology , Amides/pharmacology , Animals , Cell Culture Techniques , Cell Line , Cytochalasin B/pharmacology , Diacylglycerol Kinase/genetics , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Pyridines/pharmacology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/pharmacology , Serotonin Agents/pharmacology , Stress Fibers/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...