Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Tumour Biol ; 37(1): 763-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26245992

ABSTRACT

Ubiquitination is essential for various biological processes, such as signal transduction, intracellular trafficking, and protein degradation. Accumulating evidence has demonstrated that ubiquitination plays a crucial role in cancer development. In this report, we examine the expression and function of ubiquitin-conjugating enzyme E2S (UBE2S) in breast cancer. Immunohistochemical analysis revealed that UBE2S is highly expressed in breast cancer. The depletion of UBE2S by siRNA induced disruption of the actin cytoskeleton and focal adhesions. Interestingly, phosphorylation of FAK at Tyr397, which is important for the transduction of integrin-mediated signaling, was significantly reduced by UBE2S knockdown. We also show that UBE2S knockdown suppressed the malignant characteristics of breast cancer cells, such as migration, invasion, and anchorage-independent growth. Our results indicate that UBE2S could be a potential target for breast cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Ubiquitin-Conjugating Enzymes/metabolism , Actin Cytoskeleton/metabolism , Aged , Anoikis , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Transformation, Neoplastic , Cytoplasm/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Immunohistochemistry , Integrins/metabolism , MCF-7 Cells , Middle Aged , Neoplasm Invasiveness , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , Ubiquitination
3.
FASEB J ; 30(1): 312-23, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26381755

ABSTRACT

Proper bioriented attachment of microtubules and kinetochores is essential for the precise distribution of duplicated chromosomes to each daughter cell. An aberrant kinetochore-microtubule attachment results in chromosome instability, which leads to cellular transformation or apoptosis. In this article, we show that ubiquitin-associated protein 2-like (UBAP2L) is necessary for correct kinetochore-microtubule attachment. Depletion of UBAP2L inhibited chromosome alignment in metaphase and delayed progression to anaphase by activating spindle assembly checkpoint signaling. In addition, UBAP2L knockdown increased side-on attachment of kinetochores along the microtubules and suppressed stable kinetochore fiber formation. A proteomics analysis identified protein arginine methyltransferase (PRMT)1 as a direct interaction partner of UBAP2L. UBAP2L has an arginine- and glycine-rich motif called the RGG/RG or GAR motif in the N terminus. Biochemical analysis confirmed that arginine residues in the RGG/RG motif of UBAP2L were directly methylated by PRMT1. Finally, we demonstrated that the RGG/RG motif of UBAP2L is essential for the proper alignment of chromosomes in metaphase for the accurate distribution of chromosomes. Our results show a possible role for arginine methylation in UBAP2L for the progression of mitosis.


Subject(s)
Carrier Proteins/metabolism , Kinetochores/metabolism , Protein Processing, Post-Translational , Amino Acid Motifs , Arginine/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Methylation , Microtubules/metabolism , Protein Binding , Protein Structure, Tertiary , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism
4.
Exp Cell Res ; 332(1): 78-88, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25523619

ABSTRACT

Invadopodia are specialized actin-based microdomains of the plasma membrane that combine adhesive properties with matrix degrading activities. Proper functioning of the bone, immune, and vascular systems depend on these organelles, and their relevance in cancer cells is linked to tumor metastasis. The elucidation of the mechanisms driving invadopodia formation is a prerequisite to understanding their role and ultimately to controlling their functions. Special AT-rich sequence-binding protein 2 (SATB2) was reported to suppress tumor cell migration and metastasis. However, the mechanism of action of SATB2 is unknown. Here, we show that SATB2 inhibits invadopodia formation in HCT116 cells and that the molecular scaffold palladin is inhibited by exogenous expression of SATB2. To confirm this association, we elucidated the function of palladin in HCT116 using a knock down strategy. Palladin knock down reduced cell migration and invasion and inhibited invadopodia formation. This phenotype was confirmed by a rescue experiment. We then demonstrated that palladin expression in SATB2-expressing cells restored invasion and invadopodia formation. Our results showed that SATB2 action is mediated by palladin inhibition and the SATB2/palladin pathway is associated with invadopodia formation in colorectal cancer cells.


Subject(s)
Cell Surface Extensions/physiology , Cytoskeletal Proteins/metabolism , Matrix Attachment Region Binding Proteins/physiology , Phosphoproteins/metabolism , Transcription Factors/physiology , Cell Movement , HCT116 Cells , Humans , Protein Transport
5.
Cell Cycle ; 13(17): 2744-51, 2014.
Article in English | MEDLINE | ID: mdl-25486361

ABSTRACT

The centralspindlin complex, which is composed of MKLP1 and MgcRacGAP, is one of the crucial factors involved in cytokinesis initiation. Centralspindlin is localized at the middle of the central spindle during anaphase and then concentrates at the midbody to control abscission. A number of proteins that associate with centralspindlin have been identified. These associating factors regulate furrowing and abscission in coordination with centralspindlin. A recent study identified a novel centralspindlin partner, called Nessun Dorma, which is essential for germ cell cytokinesis in Drosophila melanogaster. SHCBP1 is a human ortholog of Nessun Dorma that associates with human centralspindlin. In this report, we analyzed the interaction of SHCBP1 with centralspindlin in detail and determined the regions that are required for the interaction. In addition, we demonstrate that the central region is necessary for the SHCBP1 dimerization. Both MgcRacGAP and MKLP1 are degraded once cells exit mitosis. Similarly, endogenous and exogenous SHCBP1 were degraded with mitosis progression. Interestingly, SHCBP1 expression was significantly reduced in the absence of centralspindlin, whereas centralspindlin expression was not affected by SHCBP1 knockdown. Finally, we demonstrate that SHCBP1 depletion promotes midbody structure disruption and inhibits abscission, a final stage of cytokinesis. Our study gives novel insight into the role of SHCBP in cytokinesis completion.


Subject(s)
Cytokinesis , Shc Signaling Adaptor Proteins/metabolism , Spindle Apparatus/metabolism , HeLa Cells , Humans , Mitosis , Models, Biological , Protein Binding , Protein Transport , Proteolysis , RNA, Small Interfering/metabolism
6.
Cancer Sci ; 105(12): 1526-32, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25250919

ABSTRACT

The striatin family of proteins, comprising STRN, STRN3 and STRN4, are multidomain-containing proteins that associate with additional proteins to form a large protein complex. We previously reported that STRN4 directly associated with protein kinases, such as MINK1, TNIK and MAP4K4, which are associated with tumor suppression or tumor progression. However, it remains unclear whether STRN4 is associated with tumor progression. In this report, we examined the role that STRN4 plays in cancer malignancy. We show that depletion of STRN4 suppresses proliferation, migration, invasion and the anchorage-independent growth of cancer cells. In addition, STRN4 knockdown increases the sensitivity of pancreatic cancer cells to gemcitabine. Finally, we show that STRN4 knockdown suppresses the proliferation and metastasis of cancer cells in mice. Our results demonstrate a possible role of STRN4 in tumor progression.


Subject(s)
Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Neoplasm Metastasis/pathology , Neoplasms, Experimental/pathology , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/pathology , Animals , Anoikis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasms, Experimental/genetics , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , RNA, Small Interfering/metabolism , Gemcitabine
7.
Carcinogenesis ; 35(9): 1993-2001, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24675530

ABSTRACT

Pleomorphic adenoma gene like-2 (PLAGL2), a member of the PLAG gene family, is a C2H2 zinc finger transcriptional factor that is involved in cellular transformation and apoptosis. In this report, we show that PLAGL2 is associated with the organization of stress fibers and with small guanosine triphosphatase (GTPase) activity. Depletion of PLAGL2 in two different ovarian cancer cell lines, ES-2 and HEY, induced activation of RhoA, whereas activity of Rac1 was suppressed. Organization of actin stress fibers and focal adhesions was significantly promoted by PLAGL2 knockdown in a RhoA-dependent manner. Conversely, exogenous expression of PLAGL2 in MDA-MB-231 cells, a breast cancer cell line, resulted in the activation of Rac1 and the inactivation of RhoA. In addition, PLAGL2 expression induced lamellipodia formation and disruption of stress fiber formation. Finally, we show that CHN1 expression is essential for Rac1 inactivation in PLAGL2-depleted cells. Our results demonstrate a crucial role of PLAGL2 in actin dynamics and give further insight into the role of PLAGL2 in cellular transformation and apoptosis.


Subject(s)
Cell Movement , DNA-Binding Proteins/physiology , RNA-Binding Proteins/physiology , Stress Fibers/metabolism , Transcription Factors/physiology , Cell Line, Tumor , Chimerin 1/metabolism , Humans , Pseudopodia/metabolism , Pseudopodia/pathology , Stress Fibers/pathology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
Mol Cell Biochem ; 389(1-2): 9-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337944

ABSTRACT

Membrane blebs are round-shaped dynamic membrane protrusions that occur under many physiological conditions. Membrane bleb production is primarily controlled by actin cytoskeletal rearrangements mediated by RhoA. Tre2-Bub2-Cdc16 (TBC) domain-containing proteins are negative regulators of the Rab family of small GTPases and contain a highly conserved TBC domain. In this report, we show that the expression of TBC1D15 is associated with the activity of RhoA and the production of membrane blebs. Depletion of TBC1D15 induced activation of RhoA and membrane blebbing, which was abolished by the addition of an inhibitor for RhoA signaling. In addition, we show that TBC1D15 is required for the accumulation of RhoA at the equatorial cortex for the ingression of the cytokinetic furrow during cytokinesis. Our results demonstrate a novel role for TBC1D15 in the regulation of RhoA during membrane blebbing and cytokinesis.


Subject(s)
GTPase-Activating Proteins/genetics , Gene Silencing/physiology , Membranes/physiology , rhoA GTP-Binding Protein/genetics , Cell Line, Tumor , Cytokinesis/genetics , Cytokinesis/physiology , GTPase-Activating Proteins/metabolism , HeLa Cells , Humans , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism
9.
J Cell Sci ; 126(Pt 16): 3627-37, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23750008

ABSTRACT

Polo-like kinase 1 (PLK1) is a widely conserved serine/threonine kinase that regulates progression of multiple stages of mitosis. Although extensive studies about PLK1 functions during cell division have been performed, it is still not known how PLK1 regulates myosin II activation at the equatorial cortex and ingression of the cleavage furrow. In this report, we show that an actin/myosin-II-binding protein, supervillin (SVIL), is a substrate of PLK1. PLK1 phosphorylates Ser238 of SVIL, which can promote the localization of SVIL to the central spindle and association with PRC1. Expression of a PLK1 phosphorylation site mutant, S238A-SVIL, inhibited myosin II activation at the equatorial cortex and induced aberrant furrowing. SVIL has both actin- and myosin-II-binding regions in the N-terminus. Expression of ΔMyo-SVIL (deleted of the myosin-II-binding region), but not of ΔAct-SVIL (deleted of actin-binding region), reduced myosin II activation and caused defects in furrowing. Our study indicates a possible role of phosphorylated SVIL as a molecular link between the central spindle and the contractile ring to coordinate the activation of myosin II for the ingression of the cleavage furrow.


Subject(s)
Cell Cycle Proteins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosin Type II/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Spindle Apparatus/metabolism , Cell Cycle Proteins/genetics , Cytokinesis/physiology , HeLa Cells , Humans , Membrane Proteins/genetics , Microfilament Proteins/genetics , Myosin Type II/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Transfection , Polo-Like Kinase 1
10.
J Cell Sci ; 126(Pt 15): 3263-70, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23704356

ABSTRACT

Centralspindlin, which is composed of MgcRacGAP and MKLP1, is essential for central spindle formation and cytokinetic furrow ingression. MgcRacGAP utilizes its GAP domain to inactivate Rac1 and induce furrow ingression in mammalian cells. In this report, we present a novel regulatory mechanism for furrowing that is mediated by the phosphorylation of SHC SH2-domain binding protein 1 (SHCBP1), a binding partner of centralspindlin, by Aurora B (AurB). AurB phosphorylates Ser634 of SHCBP1 during mitosis. We generated a phosphorylation site mutant, S634A-SHCBP1, which was prematurely recruited to the central spindle during anaphase and inhibited furrowing. An in vitro GAP assay demonstrated that SHCBP1 can suppress the MgcRacGAP-mediated inactivation of Rac1. In addition, the inhibition of Rac1 activity rescued the furrowing defect induced by S634A-SHCBP1 expression. Thus, AurB phosphorylates SHCBP1 to prevent the premature localization of SHCBP1 to the central spindle and ensures that MgcRacGAP inactivates Rac1 to promote the ingression of the cytokinetic furrow.


Subject(s)
Aurora Kinase B/metabolism , Cell Cycle/physiology , Cytokinesis/physiology , Shc Signaling Adaptor Proteins/metabolism , Spindle Apparatus/metabolism , Amino Acid Sequence , Aurora Kinase B/genetics , Cell Cycle/genetics , Cytokinesis/genetics , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Shc Signaling Adaptor Proteins/genetics , Spindle Apparatus/chemistry , Spindle Apparatus/genetics
11.
Cancer Res ; 73(5): 1581-90, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23288509

ABSTRACT

Ovarian cancer is a highly invasive and metastatic disease with a poor prognosis if diagnosed at an advanced stage, which is often the case. Recent studies argue that ovarian cancer cells that have undergone epithelial-to-mesenchymal transition (EMT) acquire aggressive malignant properties, but the relevant molecular mechanisms in this setting are not well-understood. Here, we report findings from an siRNA screen that identified the homeobox transcription factor ALX1 as a novel regulator of EMT. RNA interference-mediated attenuation of ALX1 expression restored E-cadherin expression and cell-cell junction formation in ovarian cancer cells, suppressing cell invasion, anchorage-independent growth, and tumor formation. Conversely, enforced expression of ALX1 in ovarian cancer cells or nontumorigenic epithelial cells induced EMT. We found that ALX1 upregulated expression of the key EMT regulator Snail (SNAI1) and that it mediated EMT activation and cell invasion by ALX1. Our results define the ALX1/Snail axis as a novel EMT pathway that mediates cancer invasion.


Subject(s)
Epithelial-Mesenchymal Transition , Homeodomain Proteins/physiology , Ovarian Neoplasms/genetics , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , RNA Interference , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation
12.
Mol Cell Biochem ; 374(1-2): 105-11, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23129259

ABSTRACT

Dynamic remodeling of the actin cytoskeleton is crucial for biological processes such as cell migration and cell spreading. S100A10 is a member of the S100 protein family and is involved in intracellular trafficking and cell migration. In this study, we examined the role of S100A10 in actin cytoskeletal organization and cell spreading. Depletion of S100A10 induced disruption of stress fiber formation and delay in cell spreading. Rac1 activation during spreading was suppressed by S100A10 knockdown, and exogenous expression of active Rac1 restored the ability of cells to spread in the absence of S100A10. Our results demonstrate the crucial role of S100A10 in actin dynamics promoting cell spreading via Rac1 activation.


Subject(s)
Actin Cytoskeleton/metabolism , Annexin A2/genetics , Annexin A2/metabolism , S100 Proteins/genetics , S100 Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , HEK293 Cells , HeLa Cells , Humans , Protein Transport , RNA Interference , RNA, Small Interfering , Stress Fibers
13.
J Biol Chem ; 287(30): 25019-29, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22665485

ABSTRACT

Cytokinesis is initiated by constriction of the cleavage furrow and terminated by abscission of the intercellular bridge that connects two separating daughter cells. The complicated processes of cytokinesis are coordinated by phosphorylation and dephosphorylation mediated by protein kinases and phosphatases. Mammalian Misshapen-like kinase 1 (MINK1) is a member of the germinal center kinases and is known to regulate cytoskeletal organization and oncogene-induced cell senescence. To search for novel regulators of cytokinesis, we performed a screen using a library of siRNAs and found that MINK1 was essential for cytokinesis. Time-lapse analysis revealed that MINK1-depleted cells were able to initiate furrowing but that abscission was disrupted. STRN4 (Zinedin) is a regulatory subunit of protein phosphatase 2A (PP2A) and was recently shown to be a component of a novel protein complex called striatin-interacting phosphatase and kinase (STRIPAK). Mass spectrometry analysis showed that MINK1 was a component of STRIPAK and that MINK1 directly interacted with STRN4. Similar to MINK1 depletion, STRN4-knockdown induced multinucleated cells and inhibited the completion of abscission. In addition, STRN4 reduced MINK1 activity in the presence of catalytic and structural subunits of PP2A. Our study identifies a novel regulatory network of protein kinases and phosphatases that regulate the completion of abscission.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Cytokinesis/physiology , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Calmodulin-Binding Proteins/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Multienzyme Complexes/genetics , Nerve Tissue Proteins/genetics , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/genetics
14.
Mol Biol Cell ; 22(20): 3840-52, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21865595

ABSTRACT

Rho GTPases are molecular switches that transmit biochemical signals in response to extracellular stimuli to elicit changes in the actin cytoskeleton. Rho GTPases cycle between an active, GTP-bound state and an inactive, GDP-bound state. These states are regulated by two distinct families of proteins-guanine nucleotide exchange factors and GTPase-activating proteins (GAPs). We studied the role of a previously uncharacterized GAP, ARHGAP18 (MacGAP). Overexpression of ARHGAP18 suppressed the activity of RhoA and disrupted stress fiber formation. Conversely, silencing of ARHGAP18 by small interfering RNA transfection-enhanced stress fiber formation and induced rounding of cells. We examined the role of ARHGAP18 in cell spreading and migration. Immunofluorescence analysis revealed that ARHGAP18 was localized to the leading edge during cell spreading and migration. ARHGAP18-knockdown cells showed impaired spreading, premature formation of stress fibers, and sustained activation of RhoA upon cell attachment. In addition, knockdown and overexpression of ARHGAP18 resulted in the inhibition and promotion of cell migration, respectively. Furthermore, ARHGAP18 was required for the polarization of cells for migration. Our results define ARHGAP18 as one of the crucial factors for the regulation of RhoA for the control of cell shape, spreading, and migration.


Subject(s)
Actins/metabolism , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Signal Transduction , Stress Fibers/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Polarity/genetics , Cell Shape/genetics , Fluorescent Antibody Technique , GTPase-Activating Proteins/genetics , Gene Expression , Gene Silencing , Humans , Plasmids , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stress Fibers/genetics , Transfection , rhoA GTP-Binding Protein/genetics
15.
J Sci Food Agric ; 91(12): 2259-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21547923

ABSTRACT

BACKGROUND: Medicinal plants have been used to treat various diseases since ancient times. Their specific activities, such as antioxidant, anti-inflammatory and anti-cancer, have been studied intensively. In particular, plants grown in Vietnam have attracted considerable attention among food chemists as ideal sources of natural medicinal chemicals. RESULTS: The methanol extracts from three edible Vietnamese-grown plants, Tram, Voi and Gac, tested with the DPPH assay showed antioxidant activities of 91.7 ± 0.4, 63.4 ± 0.7 and 3.7 ± 0.1% respectively. The malonaldehyde/gas chromatography assay also revealed strong antioxidant activity in Tram and Voi at a level of 25 µg mL(-1) (95.5 ± 0.3 and 78.5 ± 1.4% respectively). These results were confirmed by the thiobarbituric acid assay. The antioxidant activities correlated positively with the level of total phenolics in all plants. Tram exhibited dose response-related lipoxygenase-inhibitory activity, with values of 74.2 ± 3.1% at 5 µg mL(-1) , 62.0 ± 0% at 0.5 µg mL(-1) and 3.0 ± 1.5% at 0.05 µg mL(-1) . Conversely, Voi and Gac showed negative anti-lipoxygenase activity. CONCLUSION: The antioxidant/anti-inflammatory activities and total phenolic contents of the three edible plants grown in Vietnam revealed that they are good sources of supplements for human health.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Drug Discovery , Momordica/chemistry , Phenols/analysis , Plant Extracts/pharmacology , Syzygium/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antioxidants/chemistry , Chemical Fractionation , Flowers/chemistry , Fruit/chemistry , Lipoxygenase Inhibitors/chemistry , Lipoxygenase Inhibitors/pharmacology , Medicine, East Asian Traditional , Methanol/chemistry , Phenols/pharmacology , Plant Extracts/chemistry , Solvents/chemistry , Vegetables/chemistry , Vietnam
16.
PLoS One ; 6(12): e29338, 2011.
Article in English | MEDLINE | ID: mdl-22216253

ABSTRACT

Phosphorylation of actin-binding proteins plays a pivotal role in the remodeling of the actin cytoskeleton to regulate cell migration. Palladin is an actin-binding protein that is phosphorylated by growth factor stimulation; however, the identity of the involved protein kinases remains elusive. In this study, we report that palladin is a novel substrate of extracellular signal-regulated kinase (ERK). Suppression of ERK activation by a chemical inhibitor reduced palladin phosphorylation, and expression of active MEK alone was sufficient for phosphorylation. In addition, an in vitro kinase assay demonstrated direct palladin phosphorylation by ERK. We found that Ser77 and Ser197 are essential residues for phosphorylation. Although the phosphorylation of these residues was not required for actin cytoskeletal organization, we found that expression of non-phosphorylated palladin enhanced cell migration. Finally, we show that phosphorylation inhibits the palladin association with Abl tyrosine kinase. Taken together, our results indicate that palladin phosphorylation by ERK has an anti-migratory function, possibly by modulating interactions with molecules that regulate cell migration.


Subject(s)
Cell Movement/physiology , Cytoskeletal Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Phosphoproteins/metabolism , Base Sequence , Cell Line, Tumor , Cytoskeletal Proteins/chemistry , DNA Primers , Humans , Phosphoproteins/chemistry , Phosphorylation , Serine/metabolism
17.
Mol Biol Cell ; 21(23): 4120-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926685

ABSTRACT

Cells attach to the extracellular matrix (ECM) through integrins to form focal adhesion complexes, and this process is followed by the extension of lamellipodia to enable cell spreading. PINCH-1, an adaptor protein essential for the regulation of cell-ECM adhesion, consists of five tandem LIM domains and a small C-terminal region. PINCH-1 is known to interact with integrin-linked kinase (ILK) and Ras suppressor protein 1 (Rsu-1); however, the precise mechanism by which this complex regulates cell-ECM adhesion is not fully understood. We report here that the LIM1 domain of PINCH-1, which associates with ILK to stabilize the expression of this protein, is sufficient for cell attachment but not for cell spreading. In contrast, the C-terminal region of PINCH-1, which binds to Rsu-1, plays a pivotal role in cell spreading but not in cell attachment. We also show that PINCH-1 associates with Rsu-1 to activate Rac1 and that Rac1 activation is necessary for cell spreading. Thus, these data reveal how specific domains of PINCH-1 direct two independent pathways: one utilizing ILK to allow cell attachment, and the other recruiting Rsu-1 to activate Rac1 in order to promote cell spreading.


Subject(s)
Cell Adhesion , Cell Movement , DNA-Binding Proteins/metabolism , Extracellular Matrix/metabolism , Adaptor Proteins, Signal Transducing , Cell Line , Cell-Matrix Junctions/metabolism , DNA-Binding Proteins/genetics , Focal Adhesions/metabolism , Humans , Immunoblotting , Immunoprecipitation , Integrins/metabolism , LIM Domain Proteins , Mass Spectrometry , Membrane Proteins , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Transcription Factors/metabolism , rac1 GTP-Binding Protein/metabolism
18.
FEBS J ; 276(10): 2775-85, 2009 May.
Article in English | MEDLINE | ID: mdl-19366376

ABSTRACT

CLP36 is a member of the PDZ-LIM family of proteins, which associates with alpha-actinin and localizes to the actin cytoskeleton. CLP36 is involved in the formation of stress fibers and focal adhesions; however, the molecular mechanism of how CLP36 regulates stress fiber formation is still unknown. To investigate the physiological function of CLP36, we performed yeast two-hybrid screening, and found that CLP36 interacts with palladin. Palladin is an important structural element of the actin cytoskeleton that is ubiquitously expressed and associates with alpha-actinin. The interaction was dependent on the PDZ domain of CLP36 and the C-terminus of palladin, and silencing of palladin suppressed localization of CLP36 to stress fibers. Overexpression of the PDZ domain of CLP36 also inhibited the localization of palladin to stress fibers, suggesting that the association of CLP36 and palladin is important for the localization of both proteins to stress fibers. Our experimental results indicate that alpha-actinin, CLP36 and palladin form a protein complex and contribute to regulation of the actin cytoskeleton.


Subject(s)
Cytoskeletal Proteins/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Animals , LIM Domain Proteins , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Protein Binding , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...