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










Publication year range
1.
J Biol Chem ; 300(6): 107327, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679330

ABSTRACT

Normal receptor tyrosine kinases (RTKs) need to reach the plasma membrane (PM) for ligand-induced activation, whereas its cancer-causing mutants can be activated before reaching the PM in organelles, such as the Golgi/trans-Golgi network (TGN). Inhibitors of protein export from the endoplasmic reticulum (ER), such as brefeldin A (BFA) and 2-methylcoprophilinamide (M-COPA), can suppress the activation of mutant RTKs in cancer cells, suggesting that RTK mutants cannot initiate signaling in the ER. BFA and M-COPA block the function of ADP-ribosylation factors (ARFs) that play a crucial role in ER-Golgi protein trafficking. However, among ARF family proteins, the specific ARFs inhibited by BFA or M-COPA, that is, the ARFs involved in RTKs transport from the ER, remain unclear. In this study, we showed that M-COPA blocked the export of not only KIT but also PDGFRA/EGFR/MET RTKs from the ER. ER-retained RTKs could not fully transduce anti-apoptotic signals, thereby leading to cancer cell apoptosis. Moreover, a single knockdown of ARF1, ARF3, ARF4, ARF5, or ARF6 could not block ER export of RTKs, indicating that BFA/M-COPA treatment cannot be mimicked by the knockdown of only one ARF member. Interestingly, simultaneous transfection of ARF1, ARF4, and ARF5 siRNAs mirrored the effect of BFA/M-COPA treatment. Consistent with these results, in vitro pulldown assays showed that BFA/M-COPA blocked the function of ARF1, ARF4, and ARF5. Taken together, these results suggest that BFA/M-COPA targets at least ARF1, ARF4, and ARF5; in other words, RTKs require the simultaneous activation of ARF1, ARF4, and ARF5 for their ER export.


Subject(s)
ADP-Ribosylation Factor 1 , ADP-Ribosylation Factors , Brefeldin A , Endoplasmic Reticulum , Protein Transport , Humans , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , Endoplasmic Reticulum/metabolism , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factor 1/genetics , Brefeldin A/pharmacology , Protein Transport/drug effects , ErbB Receptors/metabolism , ErbB Receptors/genetics , HeLa Cells
2.
Biochemistry ; 63(4): 523-532, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38264987

ABSTRACT

Globin-coupled sensors constitute an important family of heme-based gas sensors, an emerging class of heme proteins. In this study, we have identified and characterized a globin-coupled sensor phosphodiesterase containing an HD-GYP domain (GCS-HD-GYP) from the human pathogen Vibrio fluvialis, which is an emerging foodborne pathogen of increasing public health concern. The amino acid sequence encoded by the AL536_01530 gene from V. fluvialis indicated the presence of an N-terminal globin domain and a C-terminal HD-GYP domain, with HD-GYP domains shown previously to display phosphodiesterase activity toward bis(3',5')-cyclic dimeric guanosine monophosphate (c-di-GMP), a bacterial second messenger that regulates numerous important physiological functions in bacteria, including in bacterial pathogens. Optical absorption spectral properties of GCS-HD-GYP were found to be similar to those of myoglobin and hemoglobin and of other bacterial globin-coupled sensors. The binding of O2 to the Fe(II) heme iron complex of GCS-HD-GYP promoted the catalysis of the hydrolysis of c-di-GMP to its linearized product, 5'-phosphoguanylyl-(3',5')-guanosine (pGpG), whereas CO and NO binding did not enhance the catalysis, indicating a strict discrimination of these gaseous ligands. These results shed new light on the molecular mechanism of gas-selective catalytic regulation by globin-coupled sensors, with these advances apt to lead to a better understanding of the family of globin-coupled sensors, a still growing family of heme-based gas sensors. In addition, given the importance of c-di-GMP in infection and virulence, our results suggested that GCS-HD-GYP could play an important role in the ability of V. fluvialis to sense O2 and NO in the context of host-pathogen interactions.


Subject(s)
Globins , Phosphoric Diester Hydrolases , Vibrio , Humans , Phosphoric Diester Hydrolases/genetics , Globins/genetics , Bacterial Proteins/chemistry , Catalysis , Cyclic GMP/metabolism , Heme/chemistry
3.
Biochem Biophys Res Commun ; 647: 72-79, 2023 03 05.
Article in English | MEDLINE | ID: mdl-36731336

ABSTRACT

Apelin (APL), an endogenous ligand for APJ, has been reported to be upregulated in a murine model of acute colitis induced by sodium dextran sulfate, as well as inflammatory bowel diseases (IBD) in humans. However, the mechanisms and functions of APL/APJ axis in the pathogenesis of IBD are unclear. We herein analyzed CD4+ T cells to determine the functions of APL in a murine model of chronic colitis induced in Rag deficient mice (Rag-/-). In colonic tissues of wild-type mice (WT), we found that APL was expressed especially in the lamina propria lymphocytes, where CD4+ T cells are dominant, rather than the epithelial cells. Unexpectedly, the APL expression was rather downregulated in the colonic tissue of the chronic colitis group compared to the control groups (Rag-/- before colitis induction and WT). The APL expression was downregulated when naïve T cells were differentiated into effecter T cells. A lack of APL resulted in decreased naïve T cells and increased effecter T cells in secondary lymphoid organs. A synthetic APL peptide, [Pyr1]-APL-13, increased IL-10 and decreased IFN-γ productions by effecter T cells. Administration of [Pyr1]-APL-13 improved survival rate in association with lessened colitis severity and decreased pro-inflammatory cytokine production. This is the first report showing immunological function of APL specifically on T cells, and these results indicate that APL/APJ axis may be a novel therapeutic target for IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Mice , Humans , Animals , T-Lymphocytes/metabolism , Apelin/metabolism , Disease Models, Animal , Colitis/pathology , Inflammatory Bowel Diseases/metabolism , Dextran Sulfate , Mice, Inbred C57BL , CD4-Positive T-Lymphocytes
4.
J Neurochem ; 163(5): 375-390, 2022 12.
Article in English | MEDLINE | ID: mdl-36227633

ABSTRACT

Phosphacan, a chondroitin sulfate proteoglycan, is a repulsive cue of cerebellar granule cells. This study aims to explore the molecular mechanism. The glycosylphosphatidylinositol-anchored neural adhesion molecule TAG-1 is a binding partner of phosphacan, suggesting that the repulsive effect of phosphacan is possibly because of its interaction with TAG-1. The repulsive effect was greatly reduced on primary cerebellar granule cells of TAG-1-deficient mice. Surface plasmon resonance analysis confirmed the direct interaction of TAG-1 with chondroitin sulfate C. On postnatal days 1, 4, 7, 11, 15, and 20 and in adulthood, phosphacan was present in the molecular layer and internal granular layer, but not in the external granular layer. In contrast, transient TAG-1 expression was observed exclusively within the premigratory zone of the external granular layer on postnatal days 1, 4, 7, and 11. Boyden chamber cell migration assay demonstrated that phosphacan exerted its repulsive effect on the spontaneous and brain-derived neurotrophic factor (BDNF)-induced migration of cerebellar granule cells. The BDNF-induced migration was inhibited by MK-2206, an Akt inhibitor. The pre-treatment with a raft-disrupting agent, methyl-ß-cyclodextrin, also inhibited the BDNF-induced migration, suggesting that lipid rafts are involved in the migration of cerebellar granule cells. In primary cerebellar granule cells obtained on postnatal day 7 and cultured for 7 days, the ganglioside GD3 and TAG-1 preferentially localized in the cell body, whereas the ganglioside GD1b and NB-3 localized in not only the cell body but also neurites. Pre-treatment with the anti-GD3 antibody R24, but not the anti-GD1b antibody GGR12, inhibited the spontaneous and BDNF-induced migration, and attenuated BDNF-induced Akt activation. These findings suggest that phosphacan is responsible for the repulsion of TAG-1-expressing cerebellar granule cells via GD3 rafts to attenuate BDNF-induced migration signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Animals , Mice , Rats , Brain-Derived Neurotrophic Factor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum/metabolism , Membrane Microdomains/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
5.
ACS Omega ; 6(50): 34912-34919, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34963974

ABSTRACT

Heme-based gas sensors are an emerging class of heme proteins. AfGcHK, a globin-coupled histidine kinase from Anaeromyxobacter sp. Fw109-5, is an oxygen sensor enzyme in which oxygen binding to Fe(II) heme in the globin sensor domain substantially enhances its autophosphorylation activity. Here, we reconstituted AfGcHK with cobalt protoporphyrin IX (Co-AfGcHK) in place of heme (Fe-AfGcHK) and characterized the spectral and catalytic properties of the full-length proteins. Spectroscopic analyses indicated that Co(III) and Co(II)-O2 complexes were in a 6-coordinated low-spin state in Co-AfGcHK, like Fe(III) and Fe(II)-O2 complexes of Fe-AfGcHK. Although both Fe(II) and Co(II) complexes were in a 5-coordinated state, Fe(II) and Co(II) complexes were in high-spin and low-spin states, respectively. The autophosphorylation activity of Co(III) and Co(II)-O2 complexes of Co-AfGcHK was fully active, whereas that of the Co(II) complex was moderately active. This contrasts with Fe-AfGcHK, where Fe(III) and Fe(II)-O2 complexes were fully active and the Fe(II) complex was inactive. Collectively, activity data and coordination structures of Fe-AfGcHK and Co-AfGcHK indicate that all fully active forms were in a 6-coordinated low-spin state, whereas the inactive form was in a 5-coordinated high-spin state. The 5-coordinated low-spin complex was moderately active-a novel finding of this study. These results suggest that the catalytic activity of AfGcHK is regulated by its heme coordination structure, especially the spin state of its heme iron. Our study presents the first successful preparation and characterization of a cobalt-substituted globin-coupled oxygen sensor enzyme and may lead to a better understanding of the molecular mechanisms of catalytic regulation in this family.

6.
Sci Rep ; 11(1): 22678, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811450

ABSTRACT

FMS-like tyrosine kinase 3 (FLT3) in hematopoietic cells binds to its ligand at the plasma membrane (PM), then transduces growth signals. FLT3 gene alterations that lead the kinase to assume its permanently active form, such as internal tandem duplication (ITD) and D835Y substitution, are found in 30-40% of acute myelogenous leukemia (AML) patients. Thus, drugs for molecular targeting of FLT3 mutants have been developed for the treatment of AML. Several groups have reported that compared with wild-type FLT3 (FLT3-wt), FLT3 mutants are retained in organelles, resulting in low levels of PM localization of the receptor. However, the precise subcellular localization of mutant FLT3 remains unclear, and the relationship between oncogenic signaling and the mislocalization is not completely understood. In this study, we show that in cell lines established from leukemia patients, endogenous FLT3-ITD but not FLT3-wt clearly accumulates in the perinuclear region. Our co-immunofluorescence assays demonstrate that Golgi markers are co-localized with the perinuclear region, indicating that FLT3-ITD mainly localizes to the Golgi region in AML cells. FLT3-ITD biosynthetically traffics to the Golgi apparatus and remains there in a manner dependent on its tyrosine kinase activity. Tyrosine kinase inhibitors, such as quizartinib (AC220) and midostaurin (PKC412), markedly decrease FLT3-ITD retention and increase PM levels of the mutant. FLT3-ITD activates downstream in the endoplasmic reticulum (ER) and the Golgi apparatus during its biosynthetic trafficking. Results of our trafficking inhibitor treatment assays show that FLT3-ITD in the ER activates STAT5, whereas that in the Golgi can cause the activation of AKT and ERK. We provide evidence that FLT3-ITD signals from the early secretory compartments before reaching the PM in AML cells.


Subject(s)
Cell Proliferation/genetics , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System/genetics , Mutation , Tandem Repeat Sequences/genetics , fms-Like Tyrosine Kinase 3/biosynthesis , fms-Like Tyrosine Kinase 3/genetics , Benzothiazoles/pharmacology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Endoplasmic Reticulum/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Golgi Apparatus/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , MAP Kinase Signaling System/drug effects , Oncogenes , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , THP-1 Cells , Tumor Suppressor Proteins/metabolism , fms-Like Tyrosine Kinase 3/antagonists & inhibitors
7.
Macromol Rapid Commun ; 42(16): e2100274, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34292631

ABSTRACT

Copper complexes act as catalysts for redox reactions to generate reactive oxygen species that destroy biomolecules and, therefore, are utilized to design drugs including antitumor and antibacterial medicines. Especially, catalytic reaction for hydrogen peroxide decomposition is important because it includes the process for generating highly toxic hydroxyl radical, i.e., Fenton-like reaction. Considering that multicoppers/hydrogen peroxide species are the important intermediates for the redox reaction, herein a polymer having copper complexes in the side chains is designed to facilitate the formation of the intermediates by building locally concentrated state of the copper complexes. The polymer increases their catalytic activities for hydrogen peroxide decomposition and promotes reactive oxygen species' generation, eventually leading to higher antibacterial activity. This reveals the virtue of building a locally concentrated state of catalysts on polymers toward drug design with low amounts of transition metals.


Subject(s)
Copper , Hydrogen Peroxide , Catalysis , Oxidation-Reduction , Polymers
8.
J Nat Prod ; 81(11): 2364-2370, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30375869

ABSTRACT

The first total synthesis of violaceoid A, a cytotoxic agent, and the asymmetric total synthesis of (-)- and (+)-violaceoid B are reported. The precursor was accessed by desymmetrization of a substituted quinol moiety, and the racemic secondary alcohol was kinetically resolved using a chiral nucleophilic catalyst. The asymmetric synthesis of (-)- and (+)-violaceoid B elucidated the absolute configuration of the naturally occurring violaceoid B. Synthetic violaceoid A inhibited the growth of human breast cancer cell lines MCF-7 and Hs 578T at concentrations of less than 100 µM, while ( S)- and ( R)-violaceoid B were inactive.


Subject(s)
Hydroquinones/chemical synthesis , Catalysis , Cell Line, Tumor , Humans , Hydroquinones/chemistry , Hydroquinones/pharmacology , Stereoisomerism
9.
Int J Oncol ; 53(5): 2157-2166, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30226592

ABSTRACT

Tamoxifen is an estrogen receptor (ER) antagonist used as first-line chemotherapy in breast cancer. Recent studies suggest that tamoxifen may be effective not only for ER­positive but also for ER­negative cancer cases. The aim of the present study was to investigate the antiproliferative effect of tamoxifen against human non­melanoma skin cancer cells. Tamoxifen inhibited the proliferation of the skin squamous cell carcinoma (SCC) cell lines A431, DJM­1 and HSC­1. A431 cells did not express ER­α or -ß, suggesting that tamoxifen may exert antiproliferative effects on skin SCC cells via a non­ER­mediated pathway. Tamoxifen increased the intracellular calcium concentration of skin SCC cells, and this increase in intracellular calcium concentration by calcium ionophore A23187 suppressed the proliferation of skin SCC cells. These data indicate that tamoxifen inhibited the proliferation of human skin SCC cells via increasing intracellular calcium concentration. Voltage-gated calcium channels and non­selective cation channels are involved in the increase in intracellular calcium concentration induced by tamoxifen. The broad-spectrum protein kinase C (PKC) inhibitor phloretin significantly attenuated the antiproliferative effect of tamoxifen on skin SCC cells. From these data, it may be concluded that tamoxifen inhibits the proliferation of skin SCC cells by induction of extracellular calcium influx via calcium channels in the plasma membrane and by subsequent activation of PKC.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Calcium/metabolism , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Skin Neoplasms/drug therapy , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Calcimycin/pharmacology , Calcium/analysis , Calcium Channels/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Phloretin/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/pathology , Tamoxifen/therapeutic use
10.
Biomed Rep ; 9(2): 175-180, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30013780

ABSTRACT

Ridaifens (RIDs), a novel series of tamoxifen derivatives, exhibit a potent growth-inhibitory effect against numerous tumor cells regardless of the expression of estrogen receptors, and are thus promising candidates as novel anti-tumor drugs. RID-B is a first generation RIDs, and inhibits the proliferation of several tumor cell lines. However, the potentially growth inhibitory effect of RID-B against hepatoma cells, and the detailed mechanism underlying RID-B-mediated tumor cell death remain to be elucidated. The purpose of the current study was to evaluate the anti-proliferative effect of RID-B against hepatoma cells. The anti-proliferative effect of RID-B against human hepatoma Huh-7 cells was investigated by cell proliferation assay using WST-1 reagent, and caspase-3 activity was evaluated by using specific fluorescent substrate. In addition, DNA fragmentation in Huh-7 cells induced by RID-B was estimated by terminal deoxynucleotidyl transferase dUTP nick-end labelling assay, and binding of RID-B to double-stranded DNA was confirmed by mass spectrometry. RID-B (0.5, 1 and 2 µM) inhibited the growth of Huh-7 cells, seemingly dose-dependently, but did not inhibit the growth of normal primary rat hepatocytes in the same concentration range. Furthermore, the caspase-3 activity of Huh-7 cells was increased by RID-B (0.5 and 5 µM), and the anti-proliferative effect of RID-B (1 µM) on Huh-7 cells was partially suppressed by the addition of the caspase inhibitor, Z-VAD-FMK. Additionally, RID-B (10 µM) directly bound to double-stranded DNA, and the addition of DNA suppressed RID-B-mediated cell growth inhibition and DNA fragmentation in Huh-7 cells. From these data, it may be concluded that RID-B inhibited cell growth and induced apoptosis via activating caspase-3 and binding to DNA directly, leading to DNA fragmentation in hepatoma cells.

11.
PLoS One ; 12(1): e0169609, 2017.
Article in English | MEDLINE | ID: mdl-28072855

ABSTRACT

Stromal cell-derived factor-1α (SDF-1α)-induced platelet aggregation is mediated through its G protein-coupled receptor CXCR4 and phosphatidylinositol 3 kinase (PI3K). Here, we demonstrate that SDF-1α induces phosphorylation of Akt at Thr308 and Ser473 in human platelets. SDF-1α-induced platelet aggregation and Akt phosphorylation are inhibited by pretreatment with the CXCR4 antagonist AMD3100 or the PI3K inhibitor LY294002. SDF-1α also induces the phosphorylation of PDK1 at Ser241 (an upstream activator of Akt), GSK3ß at Ser9 (a downstream substrate of Akt), and myosin light chain at Ser19 (a downstream element of the Akt signaling pathway). SDF-1α-induced platelet aggregation is inhibited by pretreatment with the Akt inhibitor MK-2206 in a dose-dependent manner. Furthermore, SDF-1α-induced platelet aggregation and Akt phosphorylation are inhibited by pretreatment with the raft-disrupting agent methyl-ß-cyclodextrin. Sucrose density gradient analysis shows that 35% of CXCR4, 93% of the heterotrimeric G proteins Gαi-1, 91% of Gαi-2, 50% of Gß and 4.0% of PI3Kß, and 4.5% of Akt2 are localized in the detergent-resistant membrane raft fraction. These findings suggest that SDF-1α/CXCR4 signaling in lipid rafts induces platelet aggregation via PI3K-dependent Akt phosphorylation.


Subject(s)
Blood Platelets/metabolism , Chemokine CXCL12/metabolism , Membrane Microdomains/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Humans , Phosphorylation
12.
PLoS One ; 9(2): e88369, 2014.
Article in English | MEDLINE | ID: mdl-24520376

ABSTRACT

It has been suggested that prolonged inflammatory bowel diseases (IBD) may lead to colitis-associated carcinogenesis (CAC). We previously observed that the NF-κB activation in colonic epithelial cells is associated with increased tumor necrosis factor receptor 2 (TNFR2) expression in CAC development. However, the mechanism by which epithelial NF-κB activation leading to CAC is still unclear. Myosin light chain kinase (MLCK) has been reported to be responsible for the epithelial permeability associated with TNF signaling. Therefore we focused on the role of MLCK expression via TNFR2 signaling on CAC development. Pro-tumorigenic cytokines such as IL-1ß, IL-6 and MIP-2 production as well as INF-γ and TNF production at the lamina propria were increased in the setting of colitis, and further in tumor tissues in associations with up-regulated TNFR2 and MLCK expressions in the epithelial cells of a CAC model. The up-regulated MLCK expression was observed in TNF-stimulated colonic epithelial cells in a dose-dependent fashion in association with up-regulation of TNFR2. Silencing TNFR2, but not TNFR1, resulted in restoration of epithelial tight junction (TJ) associated with decreased MLCK expression. Antibody-mediated blockade of TNF signaling also resulted in restoration of TJ in association with suppressed MLCK expression, and interestingly, similar results were observed with suppressing TNFR2 and MLCK expressions by inhibiting MLCK in the epithelial cells. Silencing of MLCK also resulted in suppressed TNFR2, but not TNFR1, expression, suggesting that the restored TJ leads to reduced TNFR2 signaling. Such suppression of MLCK as well as blockade of TNFR2 signaling resulted in restored TJ, decreased pro-tumorigenic cytokines and reduced CAC development. These results suggest that MLCK may be a potential target for the prevention of IBD-associated tumor development.


Subject(s)
Carcinogenesis/pathology , Colitis/pathology , Epithelial Cells/enzymology , Myosin-Light-Chain Kinase/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line , Cell Proliferation/drug effects , Colitis/metabolism , Colon/drug effects , Colon/pathology , Colon/ultrastructure , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Female , Inflammation/pathology , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
13.
Blood ; 122(19): 3340-8, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24002447

ABSTRACT

Membrane rafts are spatially and functionally heterogenous in the cell membrane. We observed that lysenin-positive sphingomyelin (SM)-rich rafts are identified histochemically in the central region of adhered platelets where fibrin and myosin are colocalized on activation by thrombin. The clot retraction of SM-depleted platelets from SM synthase knockout mouse was delayed significantly, suggesting that platelet SM-rich rafts are involved in clot retraction. We found that fibrin converted by thrombin translocated immediately in platelet detergent-resistant membrane (DRM) rafts but that from Glanzmann's thrombasthenic platelets failed. The fibrinogen γ-chain C-terminal (residues 144-411) fusion protein translocated to platelet DRM rafts on thrombin activation, but its mutant that was replaced by A398A399 at factor XIII crosslinking sites (Q398Q399) was inhibited. Furthermore, fibrin translocation to DRM rafts was impaired in factor XIII A subunit-deficient mouse platelets, which show impaired clot retraction. In the cytoplasm, myosin translocated concomitantly with fibrin translocation into the DRM raft of thrombin-stimulated platelets. Furthermore, the disruption of SM-rich rafts by methyl-ß-cyclodextrin impaired myosin activation and clot retraction. Thus, we propose that clot retraction takes place in SM-rich rafts where a fibrin-αIIbß3-myosin complex is formed as a primary axis to promote platelet contraction.


Subject(s)
Blood Platelets/metabolism , Clot Retraction/genetics , Factor XIII/metabolism , Fibrin/metabolism , Myosins/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelins/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Blood Platelets/cytology , Blood Platelets/drug effects , Clot Retraction/drug effects , Factor XIII/genetics , Fibrin/genetics , Gene Expression , Humans , Membrane Microdomains/chemistry , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Myosins/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Transport , Signal Transduction , Thrombin/pharmacology , Transferases (Other Substituted Phosphate Groups)/deficiency , Transferases (Other Substituted Phosphate Groups)/genetics
14.
J Appl Toxicol ; 32(1): 72-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22162085

ABSTRACT

Nanoparticles of rare metal compounds are used in various products. However, their carcinogenicity and genotoxicity have not been sufficiently evaluated. The tumor-initiating and -promoting potentials of four rare metals, indium oxide (In2O3), dysprosium oxide (Dy2O3), tungsten oxide (WO3) and molybdenum (Mo), with a well-defined particle diameter were evaluated. The mutagenicity of these rare metals was investigated by Ames test using five bacteria strains, and transformability of these rare metals was investigated by cell-transformation assay using v-Ha-ras-transfected BALB/c 3T3 cells (Bhas 42 cells). Nano-sized Dy2O3 showed strong mutagenesis in all five bacteria strains tested with and without metabolic activation, while micro-sized particles showed weak mutagenesis in two bacterial strains. Dy2O3 induced transformation colonies of Bhas 42 cell dose-dependently, although there was no difference in the number of transformed foci between nano-sized and micro-sized particles. Nano-sized In2O3 and WO3 showed positive mutagenic response in TA1537 and TA98, respectively, whereas the micro-sized metal oxide particles showed no mutagenesis in the test bacterial strains. Both nano-sized and micro-sized In2O3 showed similar levels of transformability. However, nano-sized and micro-sized WO3 did not show any transformability. Both nano-sized and micro-sized Mo particles showed neither mutagenesis nor transformability. These results suggest that mutagenicity of rare metals depends on their particle size, although transformability depends on their chemical components but not on their particle size.


Subject(s)
Carcinogens/toxicity , Metals, Rare Earth/toxicity , Mutagens/toxicity , Nanoparticles/toxicity , Oxides/toxicity , Analysis of Variance , Animals , BALB 3T3 Cells , Carcinogenicity Tests , Carcinogens/chemistry , Cell Transformation, Neoplastic/drug effects , Escherichia coli/drug effects , Escherichia coli/genetics , Metals, Rare Earth/chemistry , Mice , Mutagenicity Tests , Mutagens/chemistry , Nanoparticles/chemistry , Oxides/chemistry , Particle Size , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics
15.
J Biol Chem ; 284(30): 19817-25, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19460753

ABSTRACT

Fibronectin plays important roles in erythropoiesis through the fibronectin receptors VLA-4 and VLA-5. However, the substantial role of these fibronectin receptors and their functional assignment in erythroid differentiation are not yet fully understood. Here, we investigated the effects of cell adhesion to fibronectin on erythroid differentiation using K562 human erythroid progenitor cells. Erythroid differentiation could be induced in K562 cells in suspension by stimulating with hemin. This hemin-stimulated erythroid differentiation was highly accelerated when cells were induced to adhere to fibronectin by treatment with TNIIIA2, a peptide derived from tenascin-C, which has recently been found to induce beta1-integrin activation. Another integrin activator, Mn(2+), also accelerated hemin-stimulated erythroid differentiation. Adhesive interaction with fibronectin via VLA-4 as well as VLA-5 was responsible for acceleration of the hemin-stimulated erythroid differentiation in response to TNIIIA2, although K562 cells should have been lacking in VLA-4. Adhesion to fibronectin forced by TNIIIA2 causally induced VLA-4 expression in K562 cells, and this was blocked by the RGD peptide, an antagonist for VLA-5. The resulting adhesive interaction with fibronectin via VLA-4 strongly enhanced the hemin-stimulated activation of p38 mitogen-activated protein kinase, which was shown to serve as a signaling molecule crucial for erythroid differentiation. Suppression of VLA-4 expression by RNA interference abrogated acceleration of hemin-stimulated erythroid differentiation in response to TNIIIA2. Thus, VLA-4 and VLA-5 may contribute to erythropoiesis at different stages of erythroid differentiation.


Subject(s)
Erythroid Precursor Cells/cytology , Erythropoiesis , Fibronectins/metabolism , Hemin/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Amino Acid Sequence , Cell Adhesion/drug effects , Cell Line, Tumor , Erythroid Precursor Cells/metabolism , Gene Expression Regulation , Humans , Integrin alpha4beta1/genetics , Manganese/pharmacology , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , Tenascin/chemistry , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Endocr J ; 56(3): 509-19, 2009.
Article in English | MEDLINE | ID: mdl-19261995

ABSTRACT

Thyroglobulin is the precursor of the thyroid hormones, triiodothyronine and thyroxine. Because the molecular size of thyroglobulin is relatively large (660 kDa), it could have other additional functions besides serving as the precursor of the thyroid hormones. In this report, we examined the proliferative effects of thyroglobulins purified from bovine and porcine thyroid tissues on the growth of a rat thyroid follicular cell line, FRTL-5, as well as the primary culture of porcine thyroid epithelial cells. Bovine and porcine thyroglobulins stimulated the proliferation of not only FRTL-5 cells but also porcine thyroid epithelial cells in a dose-dependent manner. The proliferative effect of thyroglobulin was neutralized by an anti-thyroglobulin monoclonal antibody but not by two different anti-fibroblast growth factor antibodies. The stimulatory signal of thyroglobulin was transmitted via the phosphatidylinositol 3-kinase pathway. Also, removal of the N-linked oligosaccharides on thyroglobulin reduced the proliferative activity of porcine thyroglobulin, suggesting that the proliferative effect of thyroglobulin is in part exerted by its carbohydrate moiety. Taken together, we have demonstrated for the first time that thyroglobulin possesses proliferative effect on thyroid epithelial cells in addition to being the precursor of the thyroid hormones.


Subject(s)
Epithelial Cells/drug effects , Thyroglobulin/physiology , Androstadienes/pharmacology , Animals , Cattle , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction/physiology , Swine , Thyroid Gland/cytology , Wortmannin
17.
Mol Cell Biochem ; 322(1-2): 1-13, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18979187

ABSTRACT

Vascular reorganization in wound healing is a complex process, which involves coagulation, endothelial cell proliferation and migration, basement membrane regeneration, and fibrinolysis. During this healing process, the hemostatic system and the angiogenic system are intimately interconnected. To elucidate the contribution of plasminogen in the process of wound healing, we have established a perfusion cell culture system. Using this novel cell culture system, we found that addition of plasminogen in the perfusion medium allowed the "scratch-wounded" endothelial cells to recover completely, while mini-plasminogen only affected the migration but not the proliferation of the endothelial cells. In the process of recovery with the addition of plasminogen, significant plasmin activity could only be detected when the growth of the endothelial cells have almost reached confluence. This finding indicates that wound healing is triggered and promoted during the absence of the proteolytic activity of plasmin. In addition, we could not detect any matrix metalloproteinase activity in the perfusion culture medium throughout the whole culture period. However, we did found that the circulating medium collected from the perfusion system at the early phase of the healing process has stimulatory activity on the growth of endothelial cells, but the proliferative activity decreased back to the basal level when the cells reached confluence. Thus, by using the perfusion cell culture system, we found that proliferation of endothelial cells is regulated by plasminogen and the wound healing process is controlled by a temporal interaction between the endothelial cells and plasminogen.


Subject(s)
Cell Culture Techniques , Plasminogen/metabolism , Wound Healing/physiology , Animals , Cattle , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Fibrinolysin/metabolism , Kringles/physiology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism
18.
Biochem Biophys Res Commun ; 369(2): 635-40, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18294956

ABSTRACT

Angiostatin, a potent inhibitor of angiogenesis, is derived from the fibrinolytic proenzyme, plasminogen, by enzymatic processing. Plasminogen N-terminal activation peptide (PAP) is one of the products concomitantly released aside from angiostatin (kringles 1-4) and mini-plasminogen (kringle 5 plus the catalytic domain) when plasminogen is processed. To determine whether PAP alone or together with the angiostatin-related peptides derived from the processing of plasminogen modulate the proliferation and motility of endothelial cells, we have generated a recombinant PAP and used it to study its effects on endothelial cells in the presence and absence of the angiostatin-related peptides. Our results showed that PAP alone slightly increased the migration but not the proliferation of endothelial cells. However, in the presence of the angiostatin-related peptides, PAP attenuated the inhibitory activity of the angiostatin-related peptides on the proliferation and migration of endothelial cells. The inhibitory effect of PAP on the angiostatin-related peptides could be due to its binding to the kringle domains of the latter peptides.


Subject(s)
Angiostatic Proteins/administration & dosage , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Plasminogen/administration & dosage , Cells, Cultured , Drug Interactions , Endothelial Cells/cytology , Humans , Kringles/physiology
19.
Cancer Sci ; 98(10): 1577-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17645579

ABSTRACT

The presence of a chondroitin sulfate (CS) chain on human thyroglobulin (Tg) distinguishes it from Tg of other species; the role played by this chain in normal thyroid function is unclear. In the present study, we determined the structure of the CS oligosaccharides in human thyroid-derived Tg. Q-Sepharose anion exchange column chromatography of thyroid extracts indicated that the negative charge of human Tg was primarily due to the presence of the CS chain. Interestingly, the Tg of papillary carcinomas was less negatively charged, suggesting that its CS side chain was less sulfated. Structural analysis of the CS in Tg revealed that its most abundant disaccharide is the DeltaDi-0S unit (50.2 +/- 18.3%), which is not sulfated. The DeltaDi-0S, DeltaDi-6S (31.7 +/- 13.7%) and DeltaDi-diSD (12.8 +/- 4.3%) units comprise more than 90% of the disaccharides in normal Tg. However, the DeltaDi-6S (0.0-21.2%) and DeltaDi-diSD (0.0-7.7%) units were significantly reduced in Tg extracted from papillary thyroid carcinomas, whereas DeltaDi-0S (86.0 +/- 21.3%) was increased. These results suggest that the Tg in papillary carcinomas has a less sulfated CS side chain and, by virtue of that fact, is less negatively charged. What role this change in carcinoma cells has in their transformation and spread remains to be determined.


Subject(s)
Carcinoma, Papillary/metabolism , Chondroitin Sulfates/metabolism , Thyroglobulin/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Carcinoma, Papillary/pathology , Disaccharides/metabolism , Humans , Immunoblotting , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...