Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 463(4): 800-5, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26056940

ABSTRACT

Osteosarcoma is a common metastatic bone cancer that predominantly develops in children and adolescents. Metastatic osteosarcoma remains associated with a poor prognosis; therefore, more effective anti-metastatic drugs are needed. Differentiation-inducing factor-1 (DIF-1), -2, and -3 are novel lead anti-tumor agents that were originally isolated from the cellular slime mold Dictyostelium discoideum. Here we investigated the effects of a panel of DIF derivatives on lysophosphatidic acid (LPA)-induced migration of mouse osteosarcoma LM8 cells by using a Boyden chamber assay. Some DIF derivatives such as Br-DIF-1, DIF-3(+2), and Bu-DIF-3 (5-20 µM) dose-dependently suppressed LPA-induced cell migration with associated IC50 values of 5.5, 4.6, and 4.2 µM, respectively. On the other hand, the IC50 values of Br-DIF-1, DIF-3(+2), and Bu-DIF-3 versus cell proliferation were 18.5, 7.2, and 2.0 µM, respectively, in LM8 cells, and >20, 14.8, and 4.3 µM, respectively, in mouse 3T3-L1 fibroblasts (non-transformed). Together, our results demonstrate that Br-DIF-1 in particular may be a valuable tool for the analysis of cancer cell migration, and that DIF derivatives such as DIF-3(+2) and Bu-DIF-3 are promising lead anti-tumor agents for the development of therapies that suppress osteosarcoma cell proliferation, migration, and metastasis.


Subject(s)
Cell Movement/drug effects , Dictyostelium/metabolism , Hexanones/pharmacology , Lysophospholipids/pharmacology , Osteosarcoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Mitochondria/drug effects , Osteosarcoma/pathology , Oxygen Consumption/drug effects , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
2.
J Radiat Res ; 56(4): 663-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25887043

ABSTRACT

The mammalian target of rapamycin (mTOR) correlates with cell survival under hypoxia and regulates hypoxia-inducible factor-1α (HIF-1α), a key protein in hypoxia-related events. However, the role of mTOR in radio-resistance has not been fully investigated. Therefore, the effect of mTOR on the radio-resistance of cancer cells under hypoxia was evaluated using the mTOR inhibitor temsirolimus. Clonogenic survival was examined in the A549 human lung adenocarcinoma cell line under normoxia or hypoxia, with or without temsirolimus. An oxygen enhancement ratio (OER) was calculated using the D(10) values, the doses giving 10% survival. Western blotting was performed to investigate the effect of temsirolimus on mTOR and the HIF-1α pathway under normoxia and hypoxia. A549 cells showed a radio-resistance of 5.1 and 14.2 Gy, as indicated by D(10) values under normoxia and hypoxia, respectively; the OER was 2.8. The cell survival rates under hypoxia and with temsirolimus remarkably decreased compared with those under normoxia. The D(10) values of the cells under normoxia and hypoxia were 4.8 and 5.4 Gy, respectively (OER = 1.1). mTOR expression was suppressed by temsirolimus under both normoxia and hypoxia. HIF-1α expression decreased under hypoxia in the presence of temsirolimus. These results suggest that temsirolimus can overcome the radio-resistance induced by hypoxia. When the fact that mTOR acts upstream of HIF-1α is considered, our data suggest that the restoration of radiation sensitivity by temsirolimus under hypoxia may be associated with the suppression of the HIF-1α pathway. Temsirolimus could therefore be used as a hypoxic cell radio-sensitizer.


Subject(s)
Cell Hypoxia/radiation effects , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Radiation-Sensitizing Agents/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Hypoxia/drug effects , Cell Line, Tumor , Dose-Response Relationship, Radiation , Humans , Radiation Tolerance , Radiotherapy Dosage , Sirolimus/administration & dosage , Treatment Outcome
3.
Cancer Sci ; 105(10): 1236-44, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25154680

ABSTRACT

Recently, we reported that human leukocyte antigen (HLA) class I expression is predominantly regulated by the mitogen-activated protein kinase (MAPK) pathway as one of the oncogenic regulations of HLA class I expression. In the present study, we examined mechanisms of how HLA class I and PD-L1 are regulated by MAPK inhibitors and interferon-γ (IFN-γ). Furthermore, we evaluated the expression of major signal transduction molecules by Western blot and anti-tumor CTL activity by a cytotoxic assay when HLA class I and PD-L1 were modulated by MAPK inhibitors and/or IFN-γ. As a result, we confirmed, as a more general phenomenon, that the inhibition of MAPK could upregulate HLA class I expression in a panel of human solid tumors (n = 26). Of note, we showed that MAPK inhibitors act on the upregulation of HLA class I expression through a different pathway from IFN-γ; there was an additive effect in the upregulation of HLA class I when treated with the combination of MAPK inhibitors and IFN-γ, and there was no overlapping activation of JAK2/STAT1 and Erk1/2 molecules when treated with either IFN-γ or MAPK inhibitors. Furthermore, we showed that IFN-γ-treatment impaired the tumor-specific CTL activity due to the upregulation of PD-L1 in spite of the upregulation of HLA class I, while MAPK inhibitors can augment the tumor-specific CTL activity due to the upregulated HLA class I without PD-L1 alterations. In conclusion, in addition to the original anti-proliferative activity, MAPK inhibitors may work toward the enhancement of T-cell-mediated anti-tumor immunity through the upregulation of HLA class I without the upregulation of PD-L1.


Subject(s)
B7-H1 Antigen/physiology , Histocompatibility Antigens Class I/physiology , Interferon-gamma/pharmacology , MAP Kinase Signaling System/physiology , B7-H1 Antigen/analysis , Cell Line, Tumor , Flavonoids/pharmacology , Humans , Killer Cells, Natural/immunology , MAP Kinase Signaling System/drug effects , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation
4.
Radiother Oncol ; 111(2): 222-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24746574

ABSTRACT

BACKGROUND AND PURPOSE: Chromatin remodeling through histone modifications, including acetylation, plays an important role in the appropriate response to DNA damage induced by ionizing radiation (IR). Here we investigated the radiosensitizing effect of C646, a selective small molecule inhibitor of p300 histone acetyltransferase, and explored the underlying mechanisms. MATERIALS AND METHODS: A549, H157 and H460 human non-small cell lung carcinoma (NSCLC) cells, and HFL-III human lung fibroblasts were assessed by clonogenic survival assay. Apoptosis and necrosis were assessed by annexin V staining. Senescence was assessed by Senescence-associated ß-galactosidase staining. Mitotic catastrophe was assessed by evaluating nuclear morphology with DAPI staining. Cell cycle profiles were analyzed by flow cytometry. Protein expression was analyzed by immunoblotting. RESULTS: C646 sensitized A549, H460 and H157 cells to IR with a dose enhancement ratio at 10% surviving fraction of 1.4, 1.2 and 1.2, respectively. C646 did not radiosensitize HFL-III cells. In A549 cells, but not in HFL-III cells, C646 (i) enhanced mitotic catastrophe but not apoptosis, necrosis, or senescence after IR; (ii) increased the hyperploid cell population after IR; and (iii) suppressed the phosphorylation of CHK1 after IR. CONCLUSIONS: C646 radiosensitizes NSCLC cells by enhancing mitotic catastrophe through the abrogation of G2 checkpoint maintenance.


Subject(s)
Benzoates/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Histone Acetyltransferases/antagonists & inhibitors , Lung Neoplasms/pathology , Mitosis/drug effects , Pyrazoles/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Checkpoint Kinase 1 , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Lung Neoplasms/drug therapy , Nitrobenzenes , Protein Kinases/metabolism , Pyrazolones , Radiation, Ionizing
5.
J Radiat Res ; 55(4): 665-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24566720

ABSTRACT

Radiation therapy (RT) has become particularly important recently for treatment of liver tumors, but there are few experimental investigations pertaining to radiation-induced liver injuries over long-term follow-up periods. Thus, the present study examined pathological liver features over a 10-month period using an intraoperative whole-liver irradiation model. Liver function tests were performed in blood samples, whereas cell death, cell proliferation, and fibrotic changes were evaluated pathologically in liver tissues, which were collected from irradiated rats 24 h, 1, 2, 4 and 40 weeks following administration of single irradiation doses of 0 (control), 15 or 30 Gy. The impaired liver function, increased hepatocyte number, and decreased apoptotic cell proportion observed in the 15 Gy group, but not the 30 Gy group, returned to control group levels after 40 weeks; however, the Ki-67 indexes in the 15 Gy group were still higher than those in the control group after 40 weeks. Azan staining showed a fibrotic pattern in the irradiated liver in the 30 Gy group only, but the expression levels of alpha smooth muscle actin (α-SMA) and transforming growth factor-beta 1 (TGF-ß1) in both the 15 and 30 Gy groups were significantly higher than those in the control group (P < 0.05). There were differences in the pathological features of the irradiated livers between the 15 Gy and 30 Gy groups, but TGF-ß1 and α-SMA expression patterns supported the gradual progression of radiation-induced liver fibrosis in both groups. These findings will be useful in the future development of protective drugs for radiation-induced liver injury.


Subject(s)
Liver/radiation effects , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Actins/metabolism , Animals , Apoptosis/radiation effects , Cell Proliferation/radiation effects , Disease Progression , Dose-Response Relationship, Radiation , Immunohistochemistry , Intraoperative Period , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Rats , Rats, Wistar , Transforming Growth Factor beta1/metabolism
6.
Cancer Res ; 73(17): 5508-18, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23872584

ABSTRACT

The occurrence of inactivating mutations in SWI/SNF chromatin-remodeling genes in common cancers has attracted a great deal of interest. However, mechanistic strategies to target tumor cells carrying such mutations are yet to be developed. This study proposes a synthetic-lethality therapy for treating cancers deficient in the SWI/SNF catalytic (ATPase) subunit, BRG1/SMARCA4. The strategy relies upon inhibition of BRM/SMARCA2, another catalytic SWI/SNF subunit with a BRG1-related activity. Immunohistochemical analysis of a cohort of non-small-cell lung carcinomas (NSCLC) indicated that 15.5% (16 of 103) of the cohort, corresponding to preferentially undifferentiated tumors, was deficient in BRG1 expression. All BRG1-deficient cases were negative for alterations in known therapeutic target genes, for example, EGFR and DDR2 gene mutations, ALK gene fusions, or FGFR1 gene amplifications. RNA interference (RNAi)-mediated silencing of BRM suppressed the growth of BRG1-deficient cancer cells relative to BRG1-proficient cancer cells, inducing senescence via activation of p21/CDKN1A. This growth suppression was reversed by transduction of wild-type but not ATPase-deficient BRG1. In support of these in vitro results, a conditional RNAi study conducted in vivo revealed that BRM depletion suppressed the growth of BRG1-deficient tumor xenografts. Our results offer a rationale to develop BRM-ATPase inhibitors as a strategy to treat BRG1/SMARCA4-deficient cancers, including NSCLCs that lack mutations in presently known therapeutic target genes.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , DNA Helicases/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Cell Cycle , Cell Differentiation , Cell Proliferation , Cellular Senescence , DNA Helicases/antagonists & inhibitors , DNA Helicases/metabolism , Discoidin Domain Receptors , Female , Fluorescent Antibody Technique , Genes, Lethal , Humans , Immunoenzyme Techniques , Kinesins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Middle Aged , Mutation/genetics , Neoplasm Staging , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
7.
Cancer Sci ; 103(6): 1099-104, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22348348

ABSTRACT

Pancreatic cancer is highly metastatic and has a poor prognosis. However, there is no established treatment for pancreatic cancer. Lysophosphatidic acid (LPA) has been shown to be present in effluents of cancers and involved in migration and proliferation in a variety of cancer cells, including pancreatic cancer cells, in vitro. In the current study, we examined whether an orally active LPA antagonist is effective for pancreatic cancer tumorigenesis and metastasis in vivo. Oral administration of Ki16198, which is effective for LPA(1) and LPA(3), into YAPC-PD pancreatic cancer cell-inoculated nude mice significantly inhibited tumor weight and remarkably attenuated invasion and metastasis to lung, liver, and brain, in association with inhibition of matrix metalloproteinase (MMP) accumulation in ascites in vivo. Ki16198 inhibited LPA-induced migration and invasion in several pancreatic cancer cells in vitro, which was associated with the inhibition of LPA-induced MMP production. In conclusion, Ki16198 is a promising orally active LPA antagonist for inhibiting the invasion and metastasis of pancreatic cancer cells. The inhibitory effects of the antagonist on invasion and metastasis in vivo may be partially explained by the inhibition of motility activity and MMP production in cancer cells.


Subject(s)
Isoxazoles/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Propionates/pharmacology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Animals , Ascites/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Humans , Isoxazoles/administration & dosage , Isoxazoles/therapeutic use , Lysophospholipids/pharmacology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Pancreatic Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/prevention & control , Peritoneal Neoplasms/secondary , Propionates/administration & dosage , Propionates/therapeutic use , Xenograft Model Antitumor Assays
8.
Biochem Biophys Res Commun ; 415(4): 627-31, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22074830

ABSTRACT

Dexamethasone (DEX), a potent glucocorticoid, increased the expression of T-cell death associated gene 8 (TDAG8), a proton-sensing G protein-coupled receptor, which is associated with the enhancement of acidic pH-induced cAMP accumulation, in peritoneal macrophages. We explored the role of increased TDAG8 expression in the anti-inflammatory actions of DEX. The treatment of macrophages with either DEX or acidic pH induced the cell death of macrophages; however, the cell death was not affected by TDAG8 deficiency. While DEX inhibited lipopolysaccharide-induced production of tumor necrosis factor-α, an inflammatory cytokine, which was independent of TDAG8, at neutral pH, the glucocorticoid enhanced the acidic pH-induced inhibition of tumor necrosis factor-α production in a manner dependent on TDAG8. In conclusion, the DEX-induced increase in TDAG8 expression is in part involved in the glucocorticoid-induced anti-inflammatory actions through the inhibition of inflammatory cytokine production under the acidic pH environment. On the other hand, the role of TDAG8 in the DEX-induced cell death is questionable.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Macrophages, Peritoneal/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Hydrogen-Ion Concentration , Macrophages, Peritoneal/metabolism , Mice , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
9.
Biochem Biophys Res Commun ; 413(4): 499-503, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21907704

ABSTRACT

Asthma is characterized by airway inflammation, hyper-responsiveness and remodeling. Extracellular acidification is known to be associated with severe asthma; however, the role of extracellular acidification in airway remodeling remains elusive. In the present study, the effects of acidification on the expression of connective tissue growth factor (CTGF), a critical factor involved in the formation of extracellular matrix proteins and hence airway remodeling, were examined in human airway smooth muscle cells (ASMCs). Acidic pH alone induced a substantial production of CTGF, and enhanced transforming growth factor (TGF)-ß-induced CTGF mRNA and protein expression. The extracellular acidic pH-induced effects were inhibited by knockdown of a proton-sensing ovarian cancer G-protein-coupled receptor (OGR1) with its specific small interfering RNA and by addition of the G(q/11) protein-specific inhibitor, YM-254890, or the inositol-1,4,5-trisphosphate (IP(3)) receptor antagonist, 2-APB. In conclusion, extracellular acidification induces CTGF production through the OGR1/G(q/11) protein and inositol-1,4,5-trisphosphate-induced Ca(2+) mobilization in human ASMCs.


Subject(s)
Airway Remodeling , Connective Tissue Growth Factor/biosynthesis , Lung/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/metabolism , Acids/metabolism , Calcium/metabolism , Humans , Hydrogen-Ion Concentration , Inositol 1,4,5-Trisphosphate/pharmacology , Lung/cytology , Peptides, Cyclic/pharmacology , Protons , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics
10.
J Neurochem ; 117(1): 164-74, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21244430

ABSTRACT

Action mechanism of lipopolysaccharide (LPS), interleukin-1ß (IL-1ß), and lysophosphatidic acid (LPA) to regulate motility, an important process of astrogliosis, was investigated in rat astrocytes. While LPA exerted no significant effect on the cell migration, the prior treatment of the cells with LPS or IL-1ß resulted in the appearance of migration activity in response to LPA. The LPS induction of the migration response to LPA was associated with the production of IL-1ß precursor protein and inhibited by the IL-1 receptor antagonist. The IL-1ß treatment also allowed LPA to activate Rac1. The LPA-induced Rac1 activation and migration were inhibited by pertussis toxin, a small interfering RNA specific to LPA(1) receptors, and LPA(1) receptor antagonists, including Ki16425. However, the IL-1ß treatment had no appreciable effect on LPA(1) receptor mRNA expression and LPA-induced activation of ERK, Akt, and proliferation. The induction of the migration response to LPA by IL-1ß was inhibited by a constitutively active RhoA. Moreover, LPA significantly activated RhoA through the LPA(1) receptor in the control cells but not in the IL-1ß-treated cells. These results suggest that IL-1ß inhibits the LPA(1) receptor-mediated Rho signaling through the IL-1 receptor, thereby disclosing the LPA(1) receptor-mediated G(i) protein/Rac/migration pathway.


Subject(s)
Astrocytes/metabolism , Cell Movement/physiology , Interleukin-1beta/physiology , Lysophospholipids/pharmacology , Receptors, Lysophosphatidic Acid/physiology , rhoA GTP-Binding Protein/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/enzymology , Cell Movement/drug effects , Cells, Cultured , Interleukin-1beta/antagonists & inhibitors , Rats , Rats, Wistar , Receptors, Interleukin-1/physiology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , rac1 GTP-Binding Protein/physiology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
11.
J Immunol ; 185(8): 4863-72, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20861350

ABSTRACT

Lysophosphatidic acid (LPA) is a phospholipid mediator that exerts a variety of biological responses through specific G-protein-coupled receptors (LPA(1)-LPA(5) and P2Y5). LPA is thought to be involved in airway inflammation by regulating the expression of anti-inflammatory and proinflammatory genes. Chemokines such as CCL5/RANTES are secreted from airway epithelium and play a key role in allergic airway inflammation. CCL5/RANTES is a chemoattractant for eosinophils, T lymphocytes, and monocytes and seems to exacerbate asthma. We stimulated CCL5/RANTES production in a human bronchial epithelial cell line, BEAS-2B, with IFN-γ and TNF-α. When LPA was added, CCL5/RANTES mRNA expression and protein secretion were inhibited, despite the presence of IFN-γ and TNF-α. The LPA effect was attenuated by Ki16425, a LPA(1)/LPA(3) antagonist, but not by dioctylglycerol pyrophosphate 8:0, an LPA(3) antagonist. Pertussis toxin, the inhibitors for PI3K and Akt also attenuated the inhibitory effect of LPA on CCL5/RANTES secretion. We also identify the transcription factor IFN regulatory factor-1 (IRF-1) as being essential for CCL5/RANTES production. Interestingly, LPA inhibited IFN-γ and TNF-α-induced IRF-1 activation by blocking the binding of IRF-1 to its DNA consensus sequence without changing IRF-1 induction and its nuclear translocation. Ki16425, pertussis toxin, and PI3K inhibitors attenuated the inhibitory effect of LPA on IRF-1 activation. Our results suggest that LPA inhibits IFN-γ- and TNF-α-induced CCL5/RANTES production in BEAS-2B cells by blocking the binding of IRF-1 to the CCL5/RANTES promoter. LPA(1) coupled to G(i) and activation of PI3K is required for this unique effect.


Subject(s)
Chemokine CCL5/metabolism , Gene Expression Regulation/immunology , Interferon Regulatory Factor-1/metabolism , Lysophospholipids/metabolism , Respiratory Mucosa/metabolism , Blotting, Western , Bronchi/immunology , Bronchi/metabolism , Cell Line , Chemokine CCL5/immunology , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression , Humans , Interferon Regulatory Factor-1/immunology , Lysophospholipids/immunology , Promoter Regions, Genetic , Respiratory Mucosa/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Transcription, Genetic
12.
Am J Physiol Heart Circ Physiol ; 299(3): H731-42, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20622109

ABSTRACT

Atherosclerosis is a chronic inflammation disease characterized by acidic micromilieu and the accumulation of numerous bioactive lipid mediators, such as lysophosphatidic acid (LPA) and prostaglandins, in the atherosclerotic lesion. Chronic acidification induced various effects on vascular smooth muscle cells, but the molecular mechanisms underlying these effects remain unknown. In this study, we examine the role of proton-sensing ovarian cancer G protein-coupled receptor 1 (OGR1) in extracellular acidification-induced regulation of cyclooxygenase (COX)-2 induction, PGI(2) production, MAPK phosphatase (MKP)-1 expression, and plasminogen activator inhibitor (PAI)-1 expression and proliferation in human aortic smooth muscle cells (AoSMCs). Experiments with knockdown with small interfering RNA specific to OGR1 and specific inhibitors for G proteins showed that acidification-induced COX-2 expression, PGI(2) production, and MKP-1 expression, but not PAI-1 expression and inhibition of proliferation, were dependent on OGR1 and mainly mediated by G(q/11) protein. LPA remarkably enhanced, through the LPA(1) receptor/G(i) protein, the OGR1-mediated vascular actions to acidic pH. In conclusion, acidic pH-induced vascular actions of AoSMCs can be dissected to OGR1-dependent and -independent pathways: COX-2 expression, PGI(2) production, and MKP-1 expression are mediated by OGR1, but PAI-1 expression and inhibition of proliferation are not. LPA, which is usually thought to be a proatherogenic lipid mediator, may exert antiatherogenic actions under acidic micromilieu through cross-talk between LPA(1)/G(i) protein and OGR1/G(q/11) protein.


Subject(s)
Aorta/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/metabolism , Analysis of Variance , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Cyclooxygenase 2/metabolism , Dual Specificity Phosphatase 1/metabolism , Epoprostenol/metabolism , Humans , Hydrogen-Ion Concentration , Muscle, Smooth, Vascular/cytology , Plasminogen Activator Inhibitor 1/metabolism , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
Pharmacol Res ; 61(6): 499-505, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20211729

ABSTRACT

GPR4, previously proposed as the receptor for sphingosylphosphorylcholine, has recently been identified as the proton-sensing G-protein-coupled receptor coupling to multiple intracellular signaling pathways, including the G(s)-protein/cAMP, G(12/13)-protein/Rho, and G(q)-protein/phospholipase C pathways. In the present study, we examined whether extracellularly located histidine residues of GPR4 sense extracellular protons and, if so, whether a certain histidine residue is critical for coupling to the single or multiple signaling pathway(s). We found that the mutation of histidine residue at 79, 165, or 269 from the N-terminal of GPR4 to phenylalanine shifted the half-maximal effective concentration (EC(50)) of proton-induced signaling activities to the right, including cAMP accumulation, SRE promoter activity reflecting Rho activity, and NFAT promoter activity reflecting phospholipase C signaling activity, without an appreciable change in the maximal activities. These results suggest that the protonation of each one of histidine residues at 79, 165, and 269 in GPR4 may be critical for conformational change of the receptor for coupling to multiple intracellular signaling pathways through G-proteins.


Subject(s)
Histidine/genetics , Point Mutation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Cell Line , Humans , Protons
14.
Vascul Pharmacol ; 50(5-6): 178-84, 2009.
Article in English | MEDLINE | ID: mdl-19563736

ABSTRACT

Low-density lipoprotein (LDL) and lysophosphatidic acid (LPA), one of the lipid components of lipoprotein, induced the DNA synthesis of coronary artery smooth muscle cells (CASMCs). The LDL- and LPA-induced DNA synthesis was markedly inhibited by the LPA receptor antagonist Ki16425, pertussis toxin, small interfering RNAs targeted for LPA1 receptors, and a potent calcineurin inhibitor cyclosporine A. It has been reported that LDL and LPA induced a migration response in a manner sensitive to Ki16425, pertussis toxin, and a LPA1 receptor-specific small interfering RNA. However, cyclosporine A was ineffective in inhibiting the migration response. Instead, an epidermal growth factor (EGF) receptor tyrosine kinase inhibitor markedly suppressed the migration response to LDL and LPA without having any significant effect on DNA synthesis. Thus, the LDL-induced stimulation of DNA synthesis and migration in CASMCs is mediated by its component LPA through LPA1 receptors and G(i/o)-proteins. Ca2+/calcineurin pathways and transactivation of EGF receptors mediate LPA1-receptor-induced DNA synthesis and migration, respectively.


Subject(s)
Cell Movement/physiology , Coronary Vessels/metabolism , Coronary Vessels/physiology , DNA/biosynthesis , Lipoproteins, LDL/physiology , Lysophospholipids/physiology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Signal Transduction/physiology , Calcineurin/physiology , Cells, Cultured , ErbB Receptors/physiology , Humans , Receptors, Lysophosphatidic Acid/agonists , Transcriptional Activation/physiology
15.
Carcinogenesis ; 30(3): 457-65, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129242

ABSTRACT

Malignant ascites from pancreatic cancer patients has been reported to stimulate migration of pancreatic cancer cells through lysophosphatidic acid (LPA) and LPA(1) receptors. Indeed, ascites- and LPA-induced migration was inhibited by Ki16425, an LPA(1) and LPA(3) antagonist, in Panc-1 cells. Unexpectedly, however, in the presence of Ki16425, ascites and LPA inhibited cell migration in response to epidermal growth factor (EGF). The inhibitory migratory response to ascites and LPA was also observed in the cells treated with pertussis toxin (PTX), a G(i) protein inhibitor, and attenuated by a small interfering RNA (siRNA) specific to the LPA(2) receptor. The inhibitory LPA action was reversed by the regulators of G-protein signaling domain of p115RhoGEF, dominant-negative RhoA or C3 toxin. Indeed, LPA activated RhoA, which was attenuated by the siRNA against the LPA(2) receptor. Moreover, LP-105, an LPA(2) agonist, also inhibited EGF-induced migration in the PTX-treated cells. A similar inhibitory migration response through LPA(2) receptors was also observed in YAPC-PD, BxPC-3, CFPAC-1 and PK-1 pancreatic cancer cell lines. LPA also inhibited the invasion of Panc-1 cells in the PTX-treated cells in the in vitro Matrigel invasion assay. We conclude that LPA(2) receptors are coupled to the G(12/13) protein/Rho-signaling pathway, leading to the inhibition of EGF-induced migration and invasion of pancreatic cancer cells.


Subject(s)
Ascites/metabolism , Cell Movement/drug effects , Lysophospholipids/pharmacology , Pancreatic Neoplasms/metabolism , Receptors, Lysophosphatidic Acid/physiology , Ascites/pathology , Cell Line, Tumor , Collagen , Drug Combinations , Epidermal Growth Factor/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Humans , Isoxazoles/pharmacology , Laminin , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Pertussis Toxin/pharmacology , Propionates/pharmacology , Proteoglycans , RNA, Small Interfering/genetics , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/genetics , rhoA GTP-Binding Protein/metabolism
16.
J Bone Miner Res ; 23(7): 1129-39, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18302504

ABSTRACT

Acidosis has been shown to induce depletion of bone calcium from the body. This calcium release process is thought to be partially cell mediated. In an organ culture of bone, acidic pH has been shown to induce cyclooxygenase-2 (COX-2) induction and prostaglandin E(2) (PGE(2)) production, resulting in stimulation of bone calcium release. However, the molecular mechanisms whereby osteoblasts sense acidic circumstances and thereby induce COX-2 induction and PGE(2) production remain unknown. In this study, we used a human osteoblastic cell line (NHOst) to characterize cellular activities, including inositol phosphate production, intracellular Ca(2+) concentration ([Ca(2+)](i)), PGE(2) production, and COX-2 mRNA and protein expression, in response to extracellular acidification. Small interfering RNA (siRNA) specific to the OGR1 receptor and specific inhibitors for intracellular signaling pathways were used to characterize acidification-induced cellular activities. We found that extracellular acidic pH induced a transient increase in [Ca(2+)](i) and inositol phosphate production in the cells. Acidification also induced COX-2 induction, resulting in PGE(2) production. These proton-induced actions were markedly inhibited by siRNA targeted for the OGR1 receptor and the inhibitors for G(q/11) protein, phospholipase C, and protein kinase C. We conclude that the OGR1/G(q/11)/phospholipase C/protein kinase C pathway regulates osteoblastic COX-2 induction and subsequent PGE(2) production in response to acidic circumstances.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Hydrogen-Ion Concentration , Osteoblasts/metabolism , Receptors, G-Protein-Coupled/physiology , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Humans , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
17.
Cell Signal ; 19(8): 1745-53, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17462861

ABSTRACT

GPR4 was initially identified as a receptor for sphingosylphosphorylcholine and lysophosphatidylcholine; however, lipid actions have not always been confirmed. Instead, ligand-independent actions have sometimes been observed in GPR4- and other OGR1 family receptor-expressing cells. Here, we examined the possible involvement of extracellular protons, which have recently been proposed as another ligand for GPR4. At pH 7.4, the epidermal growth factor-induced extracellular signal-regulated kinase activity was lower in GPR4-transfected RH7777 cells, in association with increased cAMP accumulation, than in vector-transfected cells. The serum response element (SRE)-driven transcriptional activity was also clearly higher in GPR4-expressing HEK293 cells than in vector-transfected cells at pH 7.4. These apparent ligand-independent actions were very small at alkalinic 7.8. The SRE activity was further increased by extracellular acidification in a manner dependent on the G13 protein/Rho signaling pathway in HEK293 cells expressing GPR4 or other OGR1 receptor family members. GPR4-expressing cells also showed a calcineurin-dependent nuclear factor of activated T cell (NFAT) promoter activation at pH 7.4, and this activity was further increased by pH below 7.2 in association with inositol phosphate production. In contrast to the cAMP and SRE responses, however, alkalinization to pH 7.8 hardly affected the high basal activity. Finally, the expression of GPR4 hardly modulated the sphingosylphosphorylcholine- or lysophosphatidylcholine-induced action. These results suggest that an extracellular proton play a role as a ligand in some of previously postulated ligand-independent actions through GPR4 receptors. Moreover, GPR4 may be a multi-functional receptor coupling to Gs, G13, and Gq/11 proteins in response to extracellular acidification.


Subject(s)
Protons , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Cell Line , Cell Line, Tumor , Cyclic AMP/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Hydrogen-Ion Concentration , Ligands , Lysophosphatidylcholines/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/metabolism , Serum Response Element/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism
18.
Am J Physiol Heart Circ Physiol ; 292(5): H2513-22, 2007 May.
Article in English | MEDLINE | ID: mdl-17237247

ABSTRACT

The migration of vascular smooth muscle cells (SMCs) is a hallmark of the pathogenesis of atherosclerosis and restenosis after angioplasty. Plasma low-density lipoprotein (LDL), but not high-density lipoprotein (HDL), induced the migration of human coronary artery SMCs (CASMCs). Among bioactive lipids postulated to be present in LDL, lysophosphatidic acid (LPA) appreciably mimicked the LDL action. In fact, the LDL-induced migration was markedly inhibited by pertussis toxin, an LPA receptor antagonist Ki-16425, and a small interfering RNA (siRNA) targeted for LPA(1) receptors. Moreover, LDL contains a higher amount of LPA than HDL does. HDL markedly inhibited LPA- and platelet-derived growth factor (PDGF)-induced migration, and sphingosine 1-phosphate (S1P), the content of which is about fourfold higher in HDL than in LDL, mimicked the HDL action. The inhibitory actions of HDL and S1P were suppressed by S1P(2) receptor-specific siRNA. On the other hand, the degradation of the LPA component of LDL by monoglyceride lipase or the antagonism of LPA receptors by Ki-16425 allowed LDL to inhibit the PDGF-induced migration. The inhibitory effect of LDL was again suppressed by S1P(2) receptor-specific siRNA. In conclusion, LPA/LPA(1) receptors and S1P/S1P(2) receptors mediate the stimulatory and inhibitory migration response to LDL and HDL, respectively. The balance of not only the content of LPA and S1P in lipoproteins but also the signaling activity between LPA(1) and S1P(2) receptors in the cells may be critical in determining whether the lipoprotein is a positive or negative regulator of CASMC migration.


Subject(s)
Coronary Vessels/physiology , Lipoproteins/metabolism , Lysophospholipids/metabolism , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Adult , Cell Movement/physiology , Cells, Cultured , Humans , Male , Middle Aged
19.
J Pharmacol Sci ; 99(2): 160-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16210776

ABSTRACT

Ovarian cancer G-protein-coupled receptor 1 (OGR1), previously proposed as a receptor for sphingosylphosphorylcholine (SPC), has recently been identified as a proton-sensing or extracellular pH-responsive G-protein-coupled receptor stimulating inositol phosphate production, reflecting the activation of phospholipase C. In the present study, we found that acidic pH stimulated cAMP accumulation, reflecting the activation of adenylyl cyclase, in addition to inositol phosphate production in OGR1-expressing cells. The cAMP response was hardly affected by the inhibition of phospholipase C. SPC inhibited the acidification-induced actions in a pH-dependent manner, while no OGR1-dependent agonistic action of SPC was observed. Thus, the dose-response curves of the proton-induced actions were shifted to the right in the presence of SPC regardless of stereoisoform. The antagonistic property was also observed for psychosine and glucosylsphingosine. In conclusion, OGR1 stimulation may lead to the activation of adenylyl cyclase in addition to phospholipase C in response to extracellular acidification but not to SPC. However, SPC and related lysolipids antagonize the proton-induced and OGR1-mediated actions.


Subject(s)
Cyclic AMP/metabolism , Inositol Phosphates/metabolism , Phosphorylcholine/analogs & derivatives , Receptors, G-Protein-Coupled/antagonists & inhibitors , Second Messenger Systems/drug effects , Sphingosine/analogs & derivatives , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Lysophosphatidylcholines/pharmacology , Phosphorylcholine/pharmacology , Psychosine/analogs & derivatives , Psychosine/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Second Messenger Systems/physiology , Sphingosine/pharmacology , Transfection , Type C Phospholipases/metabolism
20.
J Biol Chem ; 280(41): 34458-64, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16087674

ABSTRACT

Ovarian cancer G-protein-coupled receptor 1 (OGR1) and GPR4 have recently been identified as proton-sensing or extracellular pH-responsive G-protein-coupled receptors stimulating inositol phosphate production and cAMP accumulation, respectively. In the present study, we found that OGR1 and GPR4 mRNAs were expressed in human aortic smooth muscle cells (AoSMCs). Acidic extracellular pH induced inositol phosphate production, a transient increase in intracellular Ca(2+) concentration ([Ca(2+)](i)), and cAMP accumulation in these cells. When small interfering RNAs (siRNAs) targeted for OGR1 and GPR4 were transfected to the cells, the acid-induced inositol phosphate production and [Ca(2+)](i) increase were markedly inhibited by the OGR1 siRNA but not by the GPR4 siRNA. Unexpectedly, the acid-induced cAMP accumulation was also largely inhibited by OGR1 siRNA but only slightly by GPR4 siRNA. Acidic extracellular pH also stimulated prostaglandin I2 (PGI(2)) production, which was again inhibited by OGR1 siRNA. The specific inhibitors for extracellular signal-regulated kinase kinase and cyclooxygenase attenuated the acid-induced PGI(2) production and cAMP accumulation without changes in the inositol phosphate production. A specific inhibitor of phospholipase C also inhibited the acid-induced cAMP accumulation. In conclusion, OGR1 is a major receptor involved in the extracellular acid-induced stimulation of PGI(2) production and cAMP accumulation in AoSMCs. The cAMP accumulation may occur through OGR1-mediated stimulation of the phospholipase C/cyclooxygenase/PGI(2) pathway.


Subject(s)
Aorta/metabolism , Cyclic AMP/metabolism , Epoprostenol/biosynthesis , Myocytes, Smooth Muscle/cytology , Receptors, G-Protein-Coupled/physiology , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Epoprostenol/chemistry , Humans , Hydrogen-Ion Concentration , Inositol Phosphates/metabolism , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transfection , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...