Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Chem Pharm Bull (Tokyo) ; 70(12): 876-884, 2022.
Article in English | MEDLINE | ID: mdl-36450586

ABSTRACT

To verify the interaction between sodium polystyrene sulfonate (SPS) and its concomitant drugs, we elucidated the capability of potassium ions and concomitant drugs to adsorb onto SPS and the effect of their coexistence on the amount adsorbed. We identified 14 drugs used concomitantly with SPS from 2017-2019 in our investigation, and 5 drug preparations used in the clinical setting were used for the experiments. In the artificial intestinal juice, SPS adsorbed 39.05-69.77 mEq/g of potassium ions. Amlodipine besylate and nifedipine were well-adsorbed, while azosemide and febuxostat were did not adsorb well onto SPS. Our results and the results of a previous study suggest that additives in drug preparations affect the adsorption of drugs onto SPS. The adsorption kinetics onto SPS of drugs conformed to the pseudo-second order model. However, the adsorption of amlodipine besylate completely may not be fitted to the pseudo-second order model. The amount of amlodipine besylate adsorbed under the coexistence of potassium ions decreased compared to when potassium ions were absent. The amount of nifedipine and potassium ions adsorbed remained constant, regardless of whether potassium ions were present or not. These results might be due to the differences in their mechanisms of adsorption onto SPS and to the characteristics of the drugs. In a clinical setting, SPS is used concomitantly with various oral drugs. The interaction between SPS and its other concomitant drugs need to be elucidated more to obtain enough evidence for pharmacists to propose the appropriate prescription.


Subject(s)
Nifedipine , Potassium , Adsorption , Ions , Gastrointestinal Tract , Amlodipine
2.
J Bone Oncol ; 8: 18-22, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28884071

ABSTRACT

BACKGROUND: Bone represents one of the most common sites to which breast cancer cells metastasize. Patients experience skeletal related adverse events (pathological fractures, spinal cord compressions, and irradiation for deteriorated pain on bone) even during treatment with zoledronic acid (ZA). Therefore, we conducted a retrospective cohort study to investigate the predictive factors for symptomatic skeletal events (SSEs) in bone-metastasized breast cancer (b-MBC) patients. METHODS: We retrospectively collected data on b-MBC patients treated with ZA. Patient characteristics, including age, subtype, the presence of non-bone lesions, the presence of multiple bone metastases at the commencement of ZA therapy, duration of ZA therapy, the time interval between breast cancer diagnosis and the initiation of ZA therapy, and type of systemic therapy, presence of previous SSE were analyzed using multivariable logistic regression analysis. RESULTS: The medical records of 183 patients were reviewed and 176 eligible patients were analyzed. The median age was 59 (range, 30-87) years. Eighty-seven patients were aged ≥60 years and 89 patients were aged < 60 years. The proportions of patients with estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2-positive disease were 81.8%, 63.1%, and 17.6%, respectively. Fifty-three patients had bone-only MBC at the commencement of ZA therapy. SSEs were observed in 42 patients. In the multivariable logistic regression analysis, bone-only MBC but not a breast cancer subtype was an independent risk factor for an SSE during ZA therapy (odds ratio: 3.878, 95% confidence interval: 1.647-9.481; p = 0.002). CONCLUSIONS: Bone-only MBC patients are more likely to experience an SSE even after treatment with ZA.

3.
Gan To Kagaku Ryoho ; 40(8): 1031-6, 2013 Aug.
Article in Japanese | MEDLINE | ID: mdl-23986046

ABSTRACT

Hiccups are often observed in patients treated with cisplatin(CDDP)-based chemotherapy. It has been reported that gender and specific dosages of CDDP and antiemetic drugs(e.g., dexamethasone and 5-HT3 receptor antagonist)using standard therapy are major risk factors in the onset of hiccups. Recently, aprepitant has been added to the antiemetic therapy in CDDP-based chemotherapy. However, it is not known how the onset of hiccups takes place in antiemetic therapy including aprepitant according to the guideline. In this study, we used cluster analysis to classify 229 patients treated with CDDP-based chemotherapy, to investigate the effect of antiemetic therapy on the onset of hiccups and chemotherapy-induced nausea and vomiting(CINV). Our analysis indicated that aprepitant was not a major risk factor for the onset of hiccups in the high CDDP dose group(≥70 mg/m(2)). However, an effect of antiemesis was confirmed in the standard therapy with aprepitant. In conclusion, we suggest that aprepitant is effective for CINV, without causing the onset of hiccups in patients treated with high-dose CDDP-based chemotherapy.


Subject(s)
Antiemetics/therapeutic use , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Dexamethasone/therapeutic use , Hiccup/drug therapy , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Cluster Analysis , Female , Hiccup/chemically induced , Hiccup/epidemiology , Humans , Male , Middle Aged , Receptors, Serotonin, 5-HT3/metabolism , Risk Factors
4.
Gan To Kagaku Ryoho ; 39(7): 1093-8, 2012 Jul.
Article in Japanese | MEDLINE | ID: mdl-22790045

ABSTRACT

Zoledronic acid(ZA)dosage should be adjusted according to the risk it poses for renal impairment. The recommended dosage for patients with creatinine clearance(Ccr)of less than 60mL/min was established on the basis of an area under the curve analysis, but is doubted because it was calculated without performing a clinical trial. Creatinine secretion from the renal tubule affects Ccr; therefore, using Ccr as the basis for dosage adjustment may be inappropriate since this can cause an overestimation of the glomerular filtration rate(GFR). The Japanese Society of Nephrology recommends using the estimated GFR(eGFR)for evaluating renal function. Therefore, this study investigated the relationship between renal function before and adverse events(AEs)after ZA administration. The dosage of only 3 of the 47 patients with Ccr less than 60mL/min could be adjusted on the basis of Ccr. During ZA therapy(3 courses), the blood urea nitrogen level and occurrence of hypokalemia were higher in the non-adjusted group than in the adjusted group, but the total number of AEs was equivalent for both groups. For all the patients, Ccr and eGFR were used as parameters for investigating AEs; the total number of AEs was equivalent for patients with differing levels of renal function. Therefore, we suggest that AEs observed during ZA therapy did not depend on the renal function level before ZA administration.


Subject(s)
Bone Density Conservation Agents/adverse effects , Diphosphonates/adverse effects , Imidazoles/adverse effects , Kidney/drug effects , Kidney/physiology , Aged , Bone Density Conservation Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Creatine/blood , Diphosphonates/therapeutic use , Female , Humans , Imidazoles/therapeutic use , Male , Middle Aged , Zoledronic Acid
5.
Leuk Res ; 36(10): 1315-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22819074

ABSTRACT

Multidrug resistance represents a major obstacle for the chemotherapy of a wide variety of human tumors. To investigate the underlying mechanisms associated with resistance to anti-cancer drugs, we established anti-cancer drug-resistant multiple myeloma (MM) cell lines RPMI8226/ADM, RPMI8226/VCR, RPMI8226/DEX, and RPMI8226/L-PAM, the 50% inhibitory concentration values of which were 77-, 58-, 79-, and 30-fold higher than their parental cell lines, respectively. The resistant cell lines overexpressed MDR1 and survivin, or showed decreased Bim expression. These results indicated that regulating these factors with inhibitors might be a viable approach to increasing the susceptibility of quiescent MM cells to chemotherapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis Regulatory Proteins/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Inhibitor of Apoptosis Proteins/metabolism , Membrane Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proto-Oncogene Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Blotting, Western , Calcium Channel Blockers/pharmacology , Caspases/metabolism , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Membrane Proteins/genetics , Multiple Myeloma/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Survivin , Tumor Cells, Cultured , Verapamil/pharmacology
6.
Mol Cell Endocrinol ; 361(1-2): 219-31, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22579611

ABSTRACT

Osteoclast differentiation is influenced by receptor activator of the NF-κB ligand (RANKL), macrophage colony-stimulating factor (M-CSF), and CD9, which are expressed on bone marrow stromal cells and osteoblasts. In addition, osteoprotegerin (OPG) is known as an osteoclastogenesis inhibitory factor. In this study, we investigated whether bisphosphonates and statins increase OPG expression and inhibit the expression of CD9, M-CSF, and RANKL in the bone marrow-derived stromal cell line ST2. We found that bisphosphonates and statins enhanced OPG mRNA expression and inhibited the expression of CD9, M-CSF, and RANKL mRNA. Futhermore, bisphosphonates and statins decreased the membrane localization of Ras and phosphorylated ERK1/2, and activated the p38MAPK. This indicates that bisphosphonates and statins enhanced OPG expression, and inhibited the expression of CD9, M-CSF, and RANKL through blocking the Ras/ERK pathway and activating p38MAPK. Accordingly, we believe that its clinical applications will be investigated in the future for the development of osteoporosis therapy.


Subject(s)
Bone Marrow Cells/enzymology , Diphosphonates/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Macrophage Colony-Stimulating Factor/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Tetraspanin 29/metabolism , Animals , Bone Marrow Cells/drug effects , Cell Death/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Macrophage Colony-Stimulating Factor/genetics , Mice , Osteoprotegerin/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Prenylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stromal Cells/drug effects , Stromal Cells/enzymology , Tetraspanin 29/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism
7.
Toxicol Appl Pharmacol ; 259(3): 402-10, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22326785

ABSTRACT

Osteosarcoma is one of the most common primary malignant bone tumors in children and adolescents. Some patients continue to have a poor prognosis, because of the metastatic disease. YM529/ONO-5920 is a nitrogen-containing bisphosphonate that has been used for the treatment of osteoporosis. YM529/ONO-5920 has recently been reported to induce apoptosis in various tumors including osteosarcoma. However, the mode of metastasis suppression in osteosarcoma by YM529/ONO-5920 is unclear. In the present study, we investigated whether YM529/ONO-5920 inhibited tumor cell migration, invasion, adhesion, or metastasis in the LM8 mouse osteosarcoma cell line. We found that YM529/ONO-5920 significantly inhibited metastasis, cell migration, invasion, and adhesion at concentrations that did not have antiproliferative effects on LM8 cells. YM529/ONO-5920 also inhibited the mRNA expression and protein activities of matrix metalloproteinases (MMPs). In addition, YM529/ONO-5920 suppressed phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and the serine/threonine protein kinase B (Akt) by the inhibition of Ras prenylation. Moreover, U0126, a mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor, and LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, also inhibited LM8 cell migration, invasion, adhesion, and metastasis, as well as the mRNA expression and protein activities of MMP-1, MMP-2, MMP-9, and MT1-MMP. The results indicated that YM529/ONO-5920 suppressed the Ras/MEK/ERK and Ras/PI3K/Akt pathways, thereby inhibiting LM8 cell migration, invasion, adhesion, and metastasis. These findings suggest that YM529/ONO-5920 has potential clinical applications for the treatment of tumor cell metastasis in osteosarcoma.


Subject(s)
Diphosphonates/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Imidazoles/pharmacology , Neoplasm Metastasis/prevention & control , Osteosarcoma/drug therapy , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C3H , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasm Invasiveness/prevention & control , Osteosarcoma/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA, Messenger/metabolism , ras Proteins/antagonists & inhibitors
8.
J Exp Clin Cancer Res ; 30: 74, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21831290

ABSTRACT

BACKGROUND: Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, the rate-limiting enzyme in cholesterol synthesis. The inhibition of this key enzyme in the mevalonate pathway leads to suppression of cell proliferation and induction of apoptosis. However, the molecular mechanism of apoptosis induction by statins is not well understood in glioblastoma. In the present study, we attempted to elucidate the mechanism by which statins induce apoptosis in C6 glioma cells. METHODS: The cytotoxicity of statins toward the C6 glioma cells were evaluated using a cell viability assay. The enzyme activity of caspase-3 was determined using activity assay kits. The effects of statins on signal transduction molecules were determined by western blot analyses. RESULTS: We found that statins inhibited cell proliferation and induced apoptosis in these cells. We also observed an increase in caspase-3 activity. The apoptosis induced by statins was not inhibited by the addition of farnesyl pyrophosphate, squalene, ubiquinone, and isopentenyladenine, but by geranylgeranyl-pyrophosphate (GGPP). Furthermore, statins decreased the levels of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) and Akt. CONCLUSIONS: These results suggest that statins induce apoptosis when GGPP biosynthesis is inhibited and consequently decreases the level of phosphorylated ERK1/2 and Akt. The results of this study also indicate that statins could be used as anticancer agents in glioblastoma.


Subject(s)
Apoptosis/drug effects , Glioblastoma/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polyisoprenyl Phosphates/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Acyl Coenzyme A/metabolism , Cell Growth Processes/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Glioblastoma/enzymology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mevalonic Acid/metabolism , Phosphorylation , Polyisoprenyl Phosphates/metabolism
9.
Gan To Kagaku Ryoho ; 38(7): 1143-8, 2011 Jul.
Article in Japanese | MEDLINE | ID: mdl-21772099

ABSTRACT

Glomerular filtration rate (GFR) is an important factor when considering carboplatin dosage adjustment. The Japanese equation for estimating GFR (eGFR) was recommended as a guideline for evaluating GFR in 2009 by The Japanese Society of Nephrology. However, benefits in the field of cancer chemotherapy with the use of eGFR have not yet been shown. To clarify the clinical benefits of eGFR, we investigated the renal function of 100 patients with gynecologic cancer who were treated with carboplatin from 2003 through 2009, and the carboplatin dosage was calculated by the Calvert formula in which eGFR was substituted for GFR. To predict the clinical benefit on the basis of carboplatin dosage using eGFR, we retrospectively divided the patients into two groups so that carboplatin dosage was within dosage in using eGFR and one was not. We compared response rates and adverse effects of the two groups. Renal function using eGFR was lower than renal function calculated by using the other formulae. Carboplatin dosage using eGFR was significantly lower than the dosage calculated with the other formulae (p<0.01). Moreover, the patients group actually, administered the dosage calculated by eGFR showed less side effects than the group of patients not treated this way, but the efficacy did not change. Thus, using eGFR in planning carboplatin dosage suggested clinical application to patients with Japanese gynecologic cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Genital Neoplasms, Female/drug therapy , Glomerular Filtration Rate/drug effects , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Asian People , Carboplatin/adverse effects , Carboplatin/therapeutic use , Female , Humans , Middle Aged , Retrospective Studies
10.
Arch Pharm Res ; 34(3): 469-75, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21547680

ABSTRACT

Mangiferin, 1,3,6,7-tetrahydroxyxanthone-C2-ß-D: -glucoside (C-glucosylxanthone), is a xanthone derivative that is widely distributed in higher plants. Recently, mangiferin was found to exhibit potential antitumor effects. However, the molecular mechanisms of this effect have not been elucidated. In the present study, we attempt to clarify the mechanism of mangiferin-induced apoptosis in the human acute myeloid leukemia cell line HL-60; mangiferin was found to induce apoptosis. We also observed a concurrent increase in caspase-3 activity and DNA fragmentation. Furthermore, on examining the survival signals expressed during apoptotic induction, we observed that mangiferin caused a remarkable decrease in the nuclear entry of NF-κB p65. However, there were no changes in the expression of other survival signals, such as extracellular signal-regulated kinase 1/2, protein kinase B, and p38 mitogenactivated protein kinase. In addition, mangiferin suppressed the expressions of Bcl-xL and XIAP; however, we did not note any changes in the levels of Bcl-2, Bax, and Bim. These results indicate that mangiferin induces apoptosis by suppressing NF-κB activation and expressions of Bcl-xL and XAIP. These findings suggest that mangiferin may be useful as an anticancer agent and can be used in combination therapy with other anticancer drugs for the treatment of acute myeloid leukemia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Nucleus/drug effects , NF-kappa B/metabolism , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Xanthones/pharmacology , bcl-X Protein/antagonists & inhibitors , Blotting, Western , Caspase 3/metabolism , Cell Nucleus/metabolism , Cell Survival/drug effects , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Protein Transport , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , X-Linked Inhibitor of Apoptosis Protein/biosynthesis , bcl-X Protein/biosynthesis
11.
Cytokine ; 54(1): 100-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21292498

ABSTRACT

The tumor microenvironment plays a critical role in modulating malignant behavior and can dramatically influence cancer treatment strategies. We investigated whether statins inhibit the expression of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), and transforming growth factor-ß (TGF-ß) mRNA in the mouse osteosarcoma cell line LM8. We found that statins significantly inhibited mRNA expressions of bFGF, HGF, and TGF-ß, and bFGF, HGF, and TGF-ß secretions at concentrations that did not have antiproliferative effects on LM8 cells, but had no effect on the mRNA expression and secretion of VEGF. The inhibition of bFGF, HGF, and TGF-ß mRNA expression, and bFGF, HGF, TGF-ß secretions was reversed when geranylgeranyl pyrophosphate (GGPP), an intermediate in the mevalonate pathway, was used in combination with statins. Furthermore, statins reduced the membrane localization of K-Ras, phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated Akt. Our research indicates that statins inhibit GGPP biosynthesis in the mevalonate pathway, and then inhibit signal transduction in the Ras/ERK and Ras/Akt pathways, thereby inhibiting bFGF, HGF, TGF-ß expression in LM8 cells. These results suggest that statins are potentially useful as anti-angiogenic agents for the treatment of osteosarcoma.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation, Enzymologic , Hepatocyte Growth Factor/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neovascularization, Pathologic , Osteosarcoma/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , ras Proteins/antagonists & inhibitors , Animals , Cell Line, Tumor , Mevalonic Acid/metabolism , Mice , Phosphorylation , Polyisoprenyl Phosphates/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
J Cell Biochem ; 111(6): 1661-72, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21053363

ABSTRACT

Multiple myeloma (MM) is a bone disease that affects many individuals. It was recently reported that macrophage inflammatory protein (MIP)-1α is constitutively secreted by MM cells. MIP-1α causes bone destruction through the formation of osteoclasts (OCs). However, the molecular mechanism underlying MIP-1α-induced OC formation is not well understood. In the present study, we attempted to clarify the mechanism whereby MIP-1α induces OC formation in a mouse macrophage-like cell line comprising C7 cells. We found that MIP-1α augmented OC formation in a concentration-dependent manner; moreover, it inhibited IFN-ß and ISGF3γ mRNA expression, and IFN-ß secretion. MIP-1α increased the expressions of phosphorylated ERK1/2 and c-Fos and decreased those of phosphorylated p38MAPK and IRF-3. We found that the MEK1/2 inhibitor U0126 inhibited OC formation by suppressing the MEK/ERK/c-Fos pathway. SB203580 induced OC formation by upregulating c-fos mRNA expression, and SB203580 was found to inhibit IFN-ß and IRF-3 mRNA expressions. The results indicate that MIP-1α induces OC formation by activating and inhibiting the MEK/ERK/c-Fos and p38MAPK/IRF-3 pathways, respectively, and suppressing IFN-ß expression. These findings may be useful in the development of an OC inhibitor that targets intracellular signaling factors.


Subject(s)
Chemokine CCL3/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Imidazoles/pharmacology , Mice , Nitriles/pharmacology , Osteoclasts/cytology , Osteoclasts/metabolism , Pyridines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics
13.
J Exp Clin Cancer Res ; 29: 127, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20843370

ABSTRACT

BACKGROUND: Melanomas are highly malignant and have high metastatic potential; hence, there is a need for new therapeutic strategies to prevent cell metastasis. In the present study, we investigated whether statins inhibit tumor cell migration, invasion, adhesion, and metastasis in the B16BL6 mouse melanoma cell line. METHODS: The cytotoxicity of statins toward the B16BL6 cells were evaluated using a cell viability assay. As an experimental model, B16BL6 cells were intravenously injected into C57BL/6 mice. Cell migration and invasion were assessed using Boyden chamber assays. Cell adhesion analysis was performed using type I collagen-, type IV collagen-, fibronectin-, and laminin-coated plates. The mRNA levels, enzyme activities and protein levels of matrix metalloproteinases (MMPs) were determined using RT-PCR, activity assay kits, and Western blot analysis, respectively; the mRNA and protein levels of vary late antigens (VLAs) were also determined. The effects of statins on signal transduction molecules were determined by western blot analyses. RESULTS: We found that statins significantly inhibited lung metastasis, cell migration, invasion, and adhesion at concentrations that did not have cytotoxic effects on B16BL6 cells. Statins also inhibited the mRNA expressions and enzymatic activities of matrix metalloproteinases (MMPs). Moreover, they suppressed the mRNA and protein expressions of integrin α2, integrin α4, and integrin α5 and decreased the membrane localization of Rho, and phosphorylated LIM kinase (LIMK) and myosin light chain (MLC). CONCLUSIONS: The results indicated that statins suppressed the Rho/Rho-associated coiled-coil-containing protein kinase (ROCK) pathways, thereby inhibiting B16BL6 cell migration, invasion, adhesion, and metastasis. Furthermore, they markedly inhibited clinically evident metastasis. Thus, these findings suggest that statins have potential clinical applications for the treatment of tumor cell metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lung Neoplasms/prevention & control , Melanoma, Experimental/drug therapy , Signal Transduction/drug effects , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Blotting, Western , Cell Survival/drug effects , Extracellular Matrix Proteins/metabolism , Fatty Acids, Monounsaturated/pharmacology , Female , Fluvastatin , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Indoles/pharmacology , Integrins/metabolism , Lim Kinases/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Melanoma, Experimental/enzymology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Neoplasm Invasiveness , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Simvastatin/pharmacology , Time Factors , rho-Associated Kinases/antagonists & inhibitors
14.
Biochem Biophys Res Commun ; 328(1): 91-7, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15670755

ABSTRACT

Increase in bone resorption by osteoclasts can cause metabolic bone diseases, such as osteoporosis. Recent attention has been paid to the receptor activator of the NF-kappaB ligand (RANKL), an accelerator of osteoclast differentiation. RANKL is expressed on the bone marrow-derived stromal cell membrane and induces the differentiation of osteoclasts by binding to RANK expressed on the osteoclast precursor cell membrane. Since the inhibition of RANKL expression can lead to the inhibition of osteoclastic bone resorption, the clinical application of RANKL inhibition could be expected to have a major effect on metabolic bone disease therapy. In this study, we investigated whether or not YM529/ONO-5920, a nitrogen-containing bisphosphonate (a novel minodronic acid), inhibits RANKL expression in a bone marrow-derived stromal cell line (ST2 cells). Reverse transcription-polymerase chain reaction revealed that the administration of YM529/ONO-5920 to ST2 cells inhibited RANKL mRNA expression and reduced RANKL proteins as assessed by Western blot analysis. The inhibition of RANKL mRNA expression was reversed when geranylgeranyl pyrophosphate (GGPP), an intermediate in the mevalonate pathway, was used in combination. Furthermore, YM529/ONO-5920 reduced phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), and similarly, U0126, a mitogen-activated protein kinase kinase 1/2 inhibitor, inhibited RANKL expression. Pretreatment with GGPP reversed the YM529/ONO-5920-induced decrease in phosphorylation of ERK. Furthermore, YM529/ONO-5920 decreased TRAP-positive cells in co-culture of ST2 cells and an osteoclast cell line, C7 cells, and this decrease was inhibited by pretreatment with GGPP. This indicates that YM529/ONO-5920 inhibits GGPP biosynthesis in the mevalonate pathway and then signal transduction in the Ras-mitogen-activated protein kinase pathway, thereby inhibiting RANKL expression on ST2 cells. These results suggest a newly elucidated action of bisphosphonates in the inhibition of bone resorption.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Carrier Proteins/metabolism , Diphosphonates/administration & dosage , Imidazoles/administration & dosage , Membrane Glycoproteins/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/ultrastructure , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Diphosphonates/chemistry , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Mice , Nitrogen/administration & dosage , Nitrogen/chemistry , Osteoclasts/drug effects , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Stromal Cells/drug effects , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL