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










Publication year range
1.
Nagoya J Med Sci ; 81(2): 183-192, 2019 May.
Article in English | MEDLINE | ID: mdl-31239586

ABSTRACT

In a world of increasing academic mobility, most universities seek to give their students opportunities to experience education in different countries, which is especially true for senior research students. The Nagoya University Graduate School of Medicine started a joint degree program (JDP) for PhD students with the University of Adelaide, Faculty of Health Science (Australia) in 2015 and with Lund University Faculty of Medicine (Sweden) in 2017. Furthermore, we have started a new JDP with the University of Freiburg, Faculty of Medicine (Germany) in 2018. This article reports the issues specific to counterpart medical schools, including student's recruitment, the curriculum, and the general differences between each schools. JDPs are not only important for educational collaboration, but also as a strategy to encourage international research collaboration, which is a core criterion to a university's world-ranking reputation. Acquiring knowledge about educational strategies that are implemented in different foreign medical schools represents a unique opportunity to improve our own curriculum.


Subject(s)
Schools, Medical/organization & administration , Curriculum , Germany , Universities
2.
Cancers (Basel) ; 10(10)2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30261620

ABSTRACT

Oncolytic viral therapy has been accepted as a standard immunotherapy since talimogene laherparepvec (T-VEC, Imlygic®) was approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) for melanoma treatment in 2015. Various oncolytic viruses (OVs), such as HF10 (Canerpaturev-C-REV) and CVA21 (CAVATAK), are now actively being developed in phase II as monotherapies, or in combination with immune checkpoint inhibitors against melanoma. Moreover, in glioma, several OVs have clearly demonstrated both safety and a promising efficacy in the phase I clinical trials. Additionally, the safety of several OVs, such as pelareorep (Reolysin®), proved their safety and efficacy in combination with paclitaxel in breast cancer patients, but the outcomes of OVs as monotherapy against breast cancer have not provided a clear therapeutic strategy for OVs. The clinical trials of OVs against pancreatic cancer have not yet demonstrated efficacy as either monotherapy or as part of combination therapy. However, there are several oncolytic viruses that have successfully proved their efficacy in different preclinical models. In this review, we mainly focused on the oncolytic viruses that transitioned into clinical trials against melanoma, glioma, pancreatic, and breast cancers. Hence, we described the current status and future prospects of OVs clinical trials against melanoma, glioma, pancreatic, and breast cancers.

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.
Oncol Rep ; 33(3): 1123-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25522823

ABSTRACT

Glioblastoma is a highly proliferative and invasive tumor. Despite extensive efforts to develop treatments for glioblastoma, the currently available therapies have only limited effects. To develop novel strategies for glioblastoma treatment, it is crucial to elucidate the molecular mechanisms that promote the invasive properties of glioblastoma. In the present study, we showed that the paired related homeobox 1 (PRRX1) is associated with glioblastoma cell invasion. The depletion of PRRX1 suppressed the invasion and neurosphere formation of glioblastoma cells. Conversely, the exogenous expression of PRRX1 promoted invasion. The Notch signaling pathway, which is an evolutionarily conserved pathway that is essential for developmental processes, plays an important role in the tumorigenesis of glioblastoma. The expression of PRRX1 induced the activation of Notch signaling, and the inhibition of Notch signaling suppressed PRRX1-mediated cell invasion. Our results indicate that activation of Notch signaling by PRRX1 is associated with the promotion of glioblastoma cell invasion.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Homeodomain Proteins/metabolism , Neoplasm Invasiveness/pathology , Receptors, Notch/metabolism , Animals , Cell Line, Tumor , Heterografts , Humans , Mice , Mice, Nude , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection
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.
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
7.
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
8.
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
9.
J Neurosci ; 33(22): 9408-19, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719809

ABSTRACT

DiGeorge syndrome chromosomal region 8 (Dgcr8), a candidate gene for 22q11.2 deletion-associated schizophrenia, encodes an essential component for microRNA (miRNA) biosynthesis that plays a pivotal role in hippocampal learning and memory. Adult neurogenesis is known to be important in hippocampus-dependent memory, but the role and molecular mechanisms of adult neurogenesis in schizophrenia remain unclear. Here, we show that Dgcr8 heterozygosity in mice leads to reduced cell proliferation and neurogenesis in adult hippocampus, as well as impaired hippocampus-dependent learning. Several schizophrenia-associated genes were downregulated in the hippocampus of Dgcr8(+/-) mice, and one of them, insulin-like growth factor 2 (Igf2), rescued the proliferation of adult neural stem cells both in vitro and in vivo. Furthermore, IGF2 improved the spatial working memory deficits in Dgcr8(+/-) mice. These data suggest that defective adult neurogenesis contributes to the cognitive impairment observed in 22q11.2 deletion-associated schizophrenia and could be rectified by IGF2.


Subject(s)
Hippocampus/growth & development , Hippocampus/pathology , Insulin-Like Growth Factor II/pharmacology , Memory Disorders/genetics , Memory Disorders/pathology , Memory, Short-Term/physiology , Neurogenesis/physiology , Proteins/genetics , Schizophrenia/genetics , Animals , Antimetabolites , Blotting, Western , Bromodeoxyuridine , Cell Proliferation , Female , Gene Deletion , Hippocampus/drug effects , Immunohistochemistry , Male , Maze Learning , Memory Disorders/drug therapy , Memory, Short-Term/drug effects , Mice , Mice, Knockout , Microarray Analysis , Motor Activity/physiology , Nesting Behavior/physiology , Neural Stem Cells/physiology , Neurogenesis/drug effects , RNA/biosynthesis , RNA/isolation & purification , RNA-Binding Proteins , Real-Time Polymerase Chain Reaction , Schizophrenic Psychology , Swimming/psychology
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.
PLoS One ; 7(8): e42137, 2012.
Article in English | MEDLINE | ID: mdl-22876303

ABSTRACT

Recently, miR-143 and miR-145 have been shown to belong to a subset of microRNAs whose expression is controlled by a complex of a tumor suppressor p53 and DEAD-box RNA helicase subunits p68/p72. While accumulating studies have acknowledged that both miRNAs function as tumor suppressors and are similarly regulated, evidence of their coordinated action against tumorigenesis has been poorly presented. Herein, we establish transgenic mice that express miR-143 under the control of the CAG regulatory unit. When crossbred with Apc(Min/+) mice, the development of tumors in the small intestines is significantly attenuated. In the transgenic small intestine tumors, the endogenous miR-145 is also enhanced and the expression of c-Myc and p68/p72, both of which have been reported to be pivotal for gut tumor development, is suppressed, corresponding to the downregulation of ERK5. We demonstrate that the combination of miR-143 and miR-145 inhibits the expression of c-Myc in human colon cancer cells, whereas miR-145 retards that of p72. Moreover, we show the possibilities that miR-145 modulates p72 expression through its 3' untranslated region and that c-Myc downregulation is involved in both p68 suppression and miR-145 induction. These findings suggest that forced expression of miR-143, probably interacting with endogenous miR-145, inhibits ERK5/c-Myc and p68/p72/ß-catenin signaling and hampers small intestine tumor development in Apc(Min/+) mice. This unique cascade, in turn, may prevent overproduction of a subset of tumor suppressive miRNAs by repressing their own modulators, p68/p72.


Subject(s)
DEAD-box RNA Helicases/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Order , Humans , Male , Mice , Mice, Transgenic , Models, Biological , Signal Transduction , beta Catenin/metabolism
13.
PLoS One ; 7(7): e41845, 2012.
Article in English | MEDLINE | ID: mdl-22844530

ABSTRACT

Cancer is a leading cause of death of men and women worldwide. Tumor cell motility contributes to metastatic invasion that causes the vast majority of cancer deaths. Extracellular receptors modified by α2,3-sialic acids that promote this motility can serve as ideal chemotherapeutic targets. For example, the extracellular domain of the mucin receptor podoplanin (PDPN) is highly O-glycosylated with α2,3-sialic acid linked to galactose. PDPN is activated by endogenous ligands to induce tumor cell motility and metastasis. Dietary lectins that target proteins containing α2,3-sialic acid inhibit tumor cell growth. However, anti-cancer lectins that have been examined thus far target receptors that have not been identified. We report here that a lectin from the seeds of Maackia amurensis (MASL) with affinity for O-linked carbohydrate chains containing sialic acid targets PDPN to inhibit transformed cell growth and motility at nanomolar concentrations. Interestingly, the biological activity of this lectin survives gastrointestinal proteolysis and enters the cardiovascular system to inhibit melanoma cell growth, migration, and tumorigenesis. These studies demonstrate how lectins may be used to help develop dietary agents that target specific receptors to combat malignant cell growth.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic , Membrane Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Plant Lectins/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Maackia/chemistry , Melanoma/blood supply , Melanoma/diet therapy , Melanoma/metabolism , Melanoma/pathology , Mice , Molecular Sequence Data , Necrosis/chemically induced , Neovascularization, Pathologic/diet therapy , Plant Lectins/chemistry , Plant Lectins/metabolism , src-Family Kinases/metabolism
14.
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
15.
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
16.
Cancer Res ; 71(4): 1229-34, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21169408

ABSTRACT

Src kinase dysregulation contributes to cancer progression but mechanistic understanding for this contribution remains incomplete. Signal regulatory protein α1 (SIRPα1) is a tumor suppressor that is constitutively suppressed in v-Src-transformed cells, where restoration of SIRPα1 expression inhibits anchorage-independent growth. In this study, we investigated the role of the protein tyrosine phosphatase-2 (SHP-2) in SIRPα1 activity. SHP-2 suppression resulted in a blockade of SIRPα1-mediated inhibition of anchorage-independent growth. Notably, we found that SIRPα1 did not act in v-Src-transformed cells by triggering cell growth arrest but by eliciting a suspension-selective apoptosis (anoikis), and that SHP-2 was required for this effect. Furthermore, we found that SHP-2 was crucial for recovery of stress fiber and focal contact formation by SIRPα1 in v-Src-transformed cells. Finally, we found that SIRPα1/SHP-2 signaling regulates anoikis in human breast carcinoma cells with activated c-Src. Taken together, our findings define SHP-2 as an essential component of tumor suppression and anoikis mediated by SIRPα1 in human breast carcinoma cells as well as in v-Src-transformed cells.


Subject(s)
Anoikis/genetics , Antigens, Differentiation/physiology , Breast Neoplasms/genetics , Carcinoma/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins pp60(c-src)/genetics , Receptors, Immunologic/physiology , Animals , Antigens, Differentiation/genetics , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins pp60(c-src)/physiology , Rats , Receptors, Immunologic/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
17.
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
18.
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
19.
Biochem Biophys Res Commun ; 400(2): 230-5, 2010 Sep 17.
Article in English | MEDLINE | ID: mdl-20727856

ABSTRACT

Gap junctional communication, which is mediated by the connexin protein family, is essential for the maintenance of normal tissue function and homeostasis. Loss of intercellular communication results in a failure to coordinately regulate cellular functions, and it can facilitate tumorigenesis. Expression of oncogenes and stimulation with cytokines has been shown to suppress intercellular communication; however, the exact mechanism by which intercellular communication is disrupted by these factors remains uncertain. In this report, we show that Akt is essential for the disruption of gap junctional communication in v-Src-transformed cells. In addition, inhibition of Akt restores gap junctional communication after it is suppressed by TNF-α signaling. Furthermore, we demonstrate that the expression of a constitutively active form of Akt1, but not of Akt2 or Akt3, is sufficient to suppress gap junctional communication. Our results clearly define Akt1 as one of the critical regulators of gap junctional communication.


Subject(s)
Cell Communication , Gap Junctions/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Transformed , Gap Junctions/enzymology , Mice , Oncogene Protein pp60(v-src)/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
20.
FEBS J ; 277(17): 3502-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20637038

ABSTRACT

Integrin-mediated activation of Cdc42 is essential for cell polarization, whereas the integrin adaptor protein Cas is required for cell migration during wound healing. After phosphorylation on tyrosine residues, Cas recruits the adaptor proteins Crk and Nck to execute integrin-mediated signals. However, the mechanisms leading to Cdc42 activation and its relationship with Cas, Crk and Nck have not been elucidated clearly. In the present study, we demonstrate that Cas utilizes Nck2 to activate Cdc42 and induce cell polarization in response to wounding. By contrast, Cas recruits CrkII to activate Rac1 and promote the extension of cell protrusions needed for cell motility. These results indicate that Cas utilizes Nck2 and CrkII in a coordinated set of distinct pathways leading to cell migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Crk-Associated Substrate Protein/metabolism , Oncogene Proteins/metabolism , Wound Healing , cdc42 GTP-Binding Protein/metabolism , Animals , Cell Line , Cell Movement , Mice , Mice, Knockout , Proto-Oncogene Proteins c-crk/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...