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










Publication year range
1.
Bioorg Med Chem Lett ; 24(16): 3961-3, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25001485

ABSTRACT

The 2,4,5-substituted-1,3,4-thiadiazoline derivative 1a has been identified as a new class of mitotic kinesin Eg5 inhibitor. With the aim of enhancement of the mitotic phase accumulation activity, structure optimization of side chains at the 2-, 4-, and 5-positions of the 1,3,4-thiadiazoline ring of 1a was performed. The introduction of sulfonylamino group at the side chain at the 5-position and bulky acyl group at the 2- and 4-position contributed to a significant increase in the mitotic phase accumulation activity and Eg5 inhibitory activity. As a result, a series of optically active compounds exhibited an increased antitumor activity in a human ovarian cancer xenograft mouse model that was induced by oral administration.


Subject(s)
Enzyme Inhibitors/pharmacology , Kinesins/antagonists & inhibitors , Thiazolidines/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Kinesins/metabolism , Molecular Structure , Structure-Activity Relationship , Thiazolidines/chemical synthesis , Thiazolidines/chemistry
2.
J Pharmacol Sci ; 124(2): 201-7, 2014.
Article in English | MEDLINE | ID: mdl-24451995

ABSTRACT

The mouse embryonic stem cell test (mEST) is used to assess the embryotoxicity of drug candidates by evaluating the effects on the cardiac differentiation of stem cells. However, thalidomide embryotoxicity has not yet been reported using the mEST. To detect the effects of thalidomide, we used human induced pluripotent stem cells (hiPSCs) instead of mouse embryonic stem cells, and assessed three endpoints: the inhibition of cardiac differentiation, the cytotoxicity to hiPSCs, and the cytotoxicity to human dermal fibroblasts, according to the mEST. From these data (IC50 values), the embryotoxicity was classified into one of three different classes based on the mEST and our criteria. Valproate was used as a positive control and ascorbic acid was used as a negative control, and their effects were assessed. Similar to valproate, thalidomide was classified as a Class 2 agent, with weak embryotoxicity, by the mEST criteria, and was classified as Category 3 embryotoxic based on our criteria. Ascorbic acid was classified as a Class 1 / Category 1, non-embryotoxic agent, based on both criteria. Thalidomide embryotoxicity was detected in the embryonic stem cell test based on hiPSCs. This test system is thus considered to have a much greater predictive ability than the mEST.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Teratogens/toxicity , Thalidomide/toxicity , Toxicity Tests/methods , Ascorbic Acid/pharmacology , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Skin/cytology , Valproic Acid/pharmacology
3.
Life Sci ; 88(19-20): 898-907, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21466810

ABSTRACT

AIMS: Benidipine, a dihydropyridine Ca(2+) channel blocker, has been reported to block T-type Ca(2+) channels; however, the mechanism underlying this effect was unclear. In this study, we characterized the mechanism responsible for this blocking activity. Furthermore, the blocking activity was compared between two enantiomers of benidipine, (S, S)- and (R, R)-benidipine. MAIN METHODS: Human Ca(v)3.2 (hCa(v)3.2) T-type Ca(2+) channels stably expressed in the human embryonic kidney cell line, HEK-293, were studied in whole-cell patch-clamp recordings and Ca(2+) mobilization assay. KEY FINDINGS: In whole-cell patch-clamp recordings, benidipine blocked hCa(v)3.2 T-type Ca(2+) currents elicited by depolarization to a comparable extent as efonidipine. The block was dependent on stimulation frequency and holding potential, but not test potential. Benidipine significantly shifted the steady-state inactivation curve to the hyperpolarizing direction, but had no effect on the activation curve. Benidipine prolonged the recovery from inactivation of hCa(v)3.2 T-type Ca(2+) channels without any effect on the kinetics of activation, inactivation, or deactivation. In the Ca(2+) mobilization assay, benidipine was more potent than efonidipine in blocking Ca(2+) influx through hCa(v)3.2 T-type Ca(2+) channels. (S, S)-Benidipine was more potent than (R, R)-benidipine in blocking hCa(v)3.2 T-type Ca(2+) currents, but there was no difference in blocking the Ca(2+) influx. SIGNIFICANCE: We have characterized the blocking activity of benidipine against hCa(v)3.2 Ca(2+) channels and revealed the difference between the two enantiomers of benidipine. The blocking action of benidipine could be mediated by stabilizing hCa(v)3.2 Ca(2+) channels in an inactivated state.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Dihydropyridines/pharmacology , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology
4.
Eur J Pharmacol ; 635(1-3): 49-55, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20307534

ABSTRACT

Aldosterone-induced activation of mineralocorticoid receptor, a member of the nuclear receptor family, results in increased tissue damage such as vascular inflammation and cardiac and perivascular fibrosis. Benidipine, a long-lasting dihydropyridine calcium channel blocker, is used for hypertension and angina. Benidipine exhibits pleiotropic pharmacological features such as renoprotective and cardioprotective effects through triple blockade of L-, N-, and T-type calcium channels. However, the mechanism of additional beneficial effects on end-organ damage is poorly understood. Here, we examined the effects of benidipine and other calcium channel blockers on aldosterone-induced mineralocorticoid receptor activation using luciferase reporter assay system. Benidipine showed more potent activity than efonidipine, amlodipine, or azelnidipine. Benidipine depressed the response to higher concentrations of aldosterone, whereas pretreatment of eplerenone, a steroidal mineralocorticoid receptor antagonist, did not. Binding studies using [(3)H] aldosterone indicated that benidipine and other calcium channel blockers competed for binding to mineralocorticoid receptor. Benidipine and other calcium channel blockers showed antagonistic activity on Ser810 to Leu mutant mineralocorticoid receptor, which is identified in patients with early-onset hypertension. On the other hand, eplerenone partially activated the mutant. Results of analysis using optical isomers of benidipine indicated that inhibitory effect of aldosterone-induced mineralocorticoid receptor activation was independent of its primary blockade of calcium channels. These results suggested that benidipine directly inhibits aldosterone-induced mineralocorticoid receptor activation, and the antagonistic activity might contribute to the drug's pleiotropic pharmacological features.


Subject(s)
Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Mineralocorticoid Receptor Antagonists , Aldosterone/pharmacology , Binding Sites , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Calcium Channels, T-Type/metabolism , Cell Line , Dihydropyridines/chemistry , Dihydropyridines/metabolism , Eplerenone , Humans , Models, Molecular , Molecular Conformation , Mutation , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Mineralocorticoid/chemistry , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Spironolactone/pharmacology , Stereoisomerism
5.
Cancer Res ; 69(9): 3901-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19351824

ABSTRACT

The aim of this study was to investigate the mechanism of inhibition of Eg5 (kinesin spindle protein), a mitotic kinesin that plays an essential role in establishing mitotic spindle bipolarity, by the novel small molecule inhibitor K858. K858 was selected in a phenotype-based forward chemical genetics screen as an antimitotic agent, and subsequently characterized as an inhibitor of Eg5. K858 blocked centrosome separation, activated the spindle checkpoint, and induced mitotic arrest in cells accompanied by the formation of monopolar spindles. Long-term continuous treatment of cancer cells with K858 resulted in antiproliferative effects through the induction of mitotic cell death, and polyploidization followed by senescence. In contrast, treatment of nontransformed cells with K858 resulted in mitotic slippage without cell death, and cell cycle arrest in G(1) phase in a tetraploid state. In contrast to paclitaxel, K858 did not induce the formation of micronuclei in either cancer or nontransformed cells, suggesting that K858 has minimal effects on abnormalities in the number and structure of chromosomes. K858 exhibited potent antitumor activity in xenograft models of cancer, and induced the accumulation of mitotic cells with monopolar spindles in tumor tissues. Importantly, K858, unlike antimicrotubule agents, had no effect on microtubule polymerization in cell-free and cell-based assays, and was not neurotoxic in a motor coordination test in mice. Taken together, the Eg5 inhibitor K858 represents an important compound for further investigation as a novel anticancer therapeutic.


Subject(s)
Colorectal Neoplasms/drug therapy , Kinesins/antagonists & inhibitors , Thiadiazoles/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Mad2 Proteins , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Paclitaxel/pharmacology , Polyploidy , Repressor Proteins/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Thiadiazoles/adverse effects , Xenograft Model Antitumor Assays
6.
Eur J Pharmacol ; 584(2-3): 424-34, 2008 Apr 28.
Article in English | MEDLINE | ID: mdl-18331727

ABSTRACT

Benidipine, a long-lasting dihydropyridine calcium channel blocker, is used for treatment of hypertension and angina. Benidipine exerts pleiotropic pharmacological features, such as renoprotective and cardioprotective effects. In pathophysiological conditions, the antidiuretic hormone aldosterone causes development of renal and cardiovascular diseases. In adrenal glomerulosa cells, aldosterone is produced in response to extracellular potassium, which is mainly mediated by T-type voltage-dependent Ca2+ channels. More recently, it has been demonstrated that benidipine inhibits T-type Ca2+ channels in addition to L-type Ca2+ channels. Therefore, effect of calcium channel blockers, including benidipine, on aldosterone production and T-type Ca2+ channels using human adrenocortical cell line NCI-H295R was investigated. Benidipine efficiently inhibited KCl-induced aldosterone production at low concentration (3 and 10 nM), with inhibitory activity more potent than other calcium channel blockers. Patch clamp analysis indicated that benidipine concentration-dependently inhibited T-type Ca2+ currents at 10, 100 and 1000 nM. As for examined calcium channel blockers, inhibitory activity for T-type Ca2+ currents was well correlated with aldosterone production. L-type specific calcium channel blockers calciseptine and nifedipine showed no effect in both assays. These results indicate that inhibition of T-type Ca2+ channels is responsible for inhibition of aldosterone production in NCI-H295R cells. Benidipine efficiently inhibited KCl-induced upregulation of 11-beta-hydroxylase mRNA and aldosterone synthase mRNA as well as KCl-induced Ca2+ influx, indicating it as the most likely inhibition mechanism. Benidipine partially inhibited angiotensin II-induced aldosterone production, plus showed additive effects when used in combination with the angiotensin II type I receptor blocker valsartan. Benidipine also partially inhibited angiotensin II-induced upregulation of the above mRNAs and Ca2+ influx inhibitory activities of benidipine for aldosterone production. T-type Ca2+ channels may contribute to additional benefits of this drug for treating renal and cardiovascular diseases, beyond its primary anti-hypertensive effects from blocking L-type Ca2+ channels.


Subject(s)
Adrenal Cortex/drug effects , Aldosterone/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Dihydropyridines/pharmacology , Adrenal Cortex/enzymology , Adrenal Cortex/metabolism , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Cell Line , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Humans , Membrane Potentials , Patch-Clamp Techniques , Potassium Chloride/pharmacology , RNA, Messenger/metabolism , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Tetrazoles/pharmacology , Valine/analogs & derivatives , Valine/pharmacology , Valsartan
7.
Prostate ; 68(6): 640-50, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18213631

ABSTRACT

BACKGROUND: Fibroblast growth factor 8-isoform b (FGF8b) has been detected in human clinical sex-organ related cancers including hormone-refractory prostate cancer. There are, however, few relevant experimental models. A murine monoclonal anti-FGF8 antibody, KM1334, has been shown to neutralize FGF8b and inhibit the growth of androgen-dependent mouse mammary SC-3 cells in vitro and in vivo. In the present study, we evaluated the anti-tumor activity of KM1334 against androgen-dependent and -independent progression of FGF8b-expressing human prostate cancer xenografts. METHODS: FGF8b cDNA was transfected into androgen-dependent human prostate cancer cell line LNCaP, and its xenograft tumors were established subcutaneously in SCID mice with or without castration. KM1334 at the dose of 400 microg/head was injected twice weekly. RESULTS: FGF8b-expressing LNCaP cells secreted FGF8b, showed enhanced level of Erk1/2 phosphorylation, and showed more potent growth properties than mock-expressing cells in vitro and in vivo. KM1334 reduced these properties in vitro, inhibited tumorigenecity in vivo (T/C=0.33), and showed anti-tumor activity against established tumors (T/C=0.47) of FGF8b-expressing cells. FGF8b-expressing LNCaP tumors were androgen-dependent. However, they recurred as androgen-independent FGF8b positive tumors after castration. KM1334 also inhibited the growth of established FGF8b-expressing tumors in the androgen-independent states (T/C=0.47). CONCLUSIONS: These results indicate that humanized monoclonal antibodies, conserving the paratope of KM1334, are a promising candidate for therapy of FGF8b-expressing clinical prostate cancers. Follow-up studies using xenograft models with clinical FGF8b-expressing tumors are required to validate these early findings.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Fibroblast Growth Factor 8/immunology , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Immunologic , Female , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, SCID , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 6/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transfection , Xenograft Model Antitumor Assays
8.
Int J Cancer ; 118(10): 2602-8, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16353147

ABSTRACT

Multiple myeloma (MM) is a fatal disease that affects plasma cells. Patients with MM have 1 or more osteolytic lesions in their bone tissues, where insulin-like growth factors (IGFs; IGF-I and IGF-II) are mainly stored. The role of bone-derived IGFs in the development of MM has not been extensively studied because reliable animal models are lacking. We established an animal model using a human MM cell line, RPMI8226, in nonobese diabetic/severe-combined immunodeficient (NOD/SCID) mice implanted with human adult bone (HAB) fragments. Treatment with an anti-human IGF-neutralizing monoclonal antibody, KM1468, inhibited the IGF-I-stimulated phosphorylation of type-I IGF receptors (IGF-IR) in RPMI8226 cells and the activation of the downstream PI3-K/Akt signaling pathway in vitro. KM1468 inhibited IGF-I-mediated RPMI8226 cell growth in a dose-dependent manner. In the NOD/SCID-HAB model, treatment with KM1468 significantly inhibited the growth of RPMI8226 cells (p<0.02). These results indicated that the growth of MM cells was predominantly stimulated not by serum-derived IGFs, but by bone-derived IGFs. Furthermore, the targeting of bone-derived IGFs, using a neutralizing antibody, may offer a new therapeutic strategy for MM.


Subject(s)
Multiple Myeloma/immunology , Multiple Myeloma/physiopathology , Somatomedins/biosynthesis , Somatomedins/immunology , Animals , Antibodies, Monoclonal/immunology , Bone and Bones/physiology , Cell Proliferation , Disease Models, Animal , Humans , Ligands , Male , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Signal Transduction , Somatomedins/metabolism , Transplantation, Heterologous
9.
Eur J Pharmacol ; 528(1-3): 176-82, 2005 Dec 28.
Article in English | MEDLINE | ID: mdl-16316644

ABSTRACT

The insulinotropic activity of KCP256 [(R)-8-benzyl-2-cyclopentyl-7, 8-dihydro-4-propyl-1H-imidazo[2,1-i]purin-5(4H)-one hydrochloride] was examined using MIN6 cells (a pancreatic beta-cell line) and pancreatic islets isolated from rats. Unlike sulfonylurea anti-diabetic drugs, KCP256 dose-dependently (0.1-10 microM) enhanced insulin secretion from MIN6 cells and its insulinotropic effect was exerted only at high concentrations of glucose (8.3-22 mM) but not at low concentrations of glucose (3.3-5.5 mM). Furthermore, the action mechanism of KCP256 was different because, unlike sulfonylurea drugs, KCP256 did not displace the binding of [3H]glibenclamide, and did not inhibit the 86Rb+ efflux nor K(ATP) channel activity. In isolated islets, KCP256 also enhanced insulin secretion in a dose- and a glucose-concentration-dependent manner. Plasma levels of insulin after glucose challenge in KCP256-administrated rats were higher than those in vehicle-administrated animals, indicating that KCP256 can enhance insulin secretion in vivo. Since the insulinotropic activity of KCP256 only occurs at high concentrations of glucose, this novel drug may exhibit a decreased risk of drug-induced hypoglycemia compared with sulfonylurea drugs when treating patients with diabetes.


Subject(s)
Glucose/pharmacology , Hypoglycemic Agents/pharmacology , Imidazoles/pharmacology , Insulin/metabolism , Purines/pharmacology , Purinones/pharmacology , Animals , Blood Glucose/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Glucose Tolerance Test , Hypoglycemic Agents/administration & dosage , In Vitro Techniques , Insulin/blood , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Purinones/administration & dosage , Rats , Rats, Wistar
10.
Clin Cancer Res ; 11(10): 3897-904, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897591

ABSTRACT

PURPOSE: Fibroblast growth factor 8b (FGF8b) has been implicated in oncogenesis of sex hormone-related malignancies. A murine monoclonal anti-FGF8 antibody, KM1334, has been raised against a FGF8b-derived peptide and shown to neutralize FGF8b activity in an androgen-dependent mouse mammary cell line (SC-3) in vitro growth. The purpose of this study was to evaluate KM1334 as a therapeutic agent for FGF8-dependent cancer. EXPERIMENTAL DESIGN: Specificity and neutralizing activity of KM1334 were examined in vitro. In vivo therapeutic studies were done in nude mice bearing SC-3 tumors s.c. RESULTS: KM1334 recognized FGF8b and FGF8f specifically out of four human FGF8 isoforms and showed little binding to other members of FGF family. Neutralizing activity of KM1334 was confirmed by both blocking of FGF8b binding to its three receptors (FGFR2IIIc, FGFR3IIIc, and FGFR4) and FGF8b-induced phosphorylation of FGFR substrate 2alpha and extracellular signal-regulated kinase 1/2 in SC-3 cells. The in vitro inhibitory effect could be extended to in vivo tumor models, where KM1334 caused rapid regression of established SC-3 tumors in nude mice. This rapid regression of tumors after KM1334 treatment was explained by two independent mechanisms: (a) decreased DNA synthesis, as evidenced by a decrease in uptake of 5-bromo-2'-deoxyuridine, and (b) induction of apoptosis as shown by the terminal deoxynucleotidyl transferase-mediated nick end labeling assay. CONCLUSIONS: KM1334 possesses strong blocking activity in vitro and antitumor activity in vivo and therefore may be an effective therapeutic candidate for the treatment of cancers that are dependent on FGF8b signaling for growth and survival.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity/immunology , Fibroblast Growth Factors/immunology , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Animals , Cell Proliferation , DNA/biosynthesis , Fibroblast Growth Factor 8 , Humans , Immunohistochemistry , Mice , Mice, Nude , Protein Isoforms , Signal Transduction , Tumor Cells, Cultured
11.
Eur J Pharmacol ; 495(1): 1-16, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15219815

ABSTRACT

Adenosine is a purine nucleoside and modulates a variety of physiological functions by interacting with cell-surface adenosine receptors. Under several adverse conditions, including ischemia, trauma, stress, seizures and inflammation, extracellular levels of adenosine are increased due to increased energy demands and ATP metabolism. Increased adenosine could protect against excessive cellular damage and organ dysfunction. Indeed, several protective effects of adenosine have been widely reported (e.g., amelioration of ischemic heart and brain injury, seizures and inflammation). However, the effects of adenosine itself are insufficient because extracellular adenosine is rapidly taken up into adjacent cells and subsequently metabolized. Adenosine uptake inhibitors (nucleoside transport inhibitors) could retard the disappearance of adenosine from the extracellular space by blocking adenosine uptake into cells. Therefore, it is expected that adenosine uptake inhibitors will have protective effects in various diseases, by elevating extracellular adenosine levels. Protective or ameliorating effects of adenosine uptake inhibitors in ischemic cardiac and cerebral injury, organ transplantation, seizures, thrombosis, insomnia, pain, and inflammatory diseases have been reported. Preclinical and clinical results indicate the possibility of therapeutic application of adenosine uptake inhibitors.


Subject(s)
Adenosine/antagonists & inhibitors , Adenosine/pharmacology , Nucleoside Transport Proteins/antagonists & inhibitors , Animals , Brain Ischemia/drug therapy , Forecasting , Humans , Models, Biological , Molecular Structure , Myocardial Reperfusion Injury/drug therapy , Nucleoside Transport Proteins/physiology , Nucleoside Transport Proteins/therapeutic use
12.
Nihon Yakurigaku Zasshi ; 122(2): 121-34, 2003 Aug.
Article in Japanese | MEDLINE | ID: mdl-12890898

ABSTRACT

Adenosine protects against cellular damage and dysfunction under several adverse conditions, including inflammation. We examined the effects of KF24345, a novel adenosine uptake inhibitor, on inflammatory diseases to investigate whether the adenosine uptake inhibition is useful for the treatment of inflammation. KF24345 inhibited adenosine uptake into washed erythrocytes (in vitro) and sampled blood cells from mice after its oral administration (in vivo). KF24345 significantly suppressed lipopolysaccharide-induced tumor necrosis factor-alpha production and leukopenia in mice, and the effects of KF24345 were abolished by the treatment with a non-selective or an A(2A)-selective adenosine receptor antagonist. In the experimental glomerulonephritis induced in mice by anti-glomerular basement membrane antiserum, KF24345 significantly inhibited proteinuria and glomerular damage without exhibiting the side effects observed following the treatment with prednisolone and cyclophosphamide. In addition, KF24345 ameliorated the severity of experimental acute pancreatitis induced by cerulein or choline-deficient and ethionine-supplemented diet in mice, and it decreased mortality accompanying severe acute pancreatitis. The anti-pancreatitis effects of KF24345 were abolished by the treatment with a non-selective or an A(2A)-selective adenosine receptor antagonist. These results suggest that KF24345 and adenosine uptake inhibitors can be a new therapeutic approach for various inflammatory diseases, including glomerulonephritis and acute pancreatitis.


Subject(s)
Adenosine/metabolism , Glomerulonephritis/drug therapy , Inflammation/drug therapy , Neurotransmitter Uptake Inhibitors/pharmacology , Neurotransmitter Uptake Inhibitors/therapeutic use , Pancreatitis/drug therapy , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Acute Disease , Adenosine/physiology , Animals , Depression, Chemical , Erythrocytes/metabolism , Humans , Leukopenia/drug therapy , Lipopolysaccharides/antagonists & inhibitors , Mice , Tumor Necrosis Factor-alpha/biosynthesis
13.
Cancer Res ; 63(14): 3931-9, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12873987

ABSTRACT

Telomerase, the enzyme responsible for proliferative immortality, is expressed in essentially all cancer cells, but not in most normal human cells. Thus, specific telomerase inhibition is potentially a universal anticancer therapy with few side effects. We designed N3'-->P5' thio-phosphoramidate (NPS) oligonucleotides as telomerase template antagonists and found that their ability to form stable duplexes with the telomerase RNA subunit was the key factor for antitelomerase activity. In biochemical assays 11-13-mer NPS oligonucleotides demonstrated sequence- and dose-dependent inhibition of telomerase with IC(50) values <1 nM. Optimization of the sequence, length, and bioavailability resulted in the selection of a 13-mer NPS oligonucleotide, GRN163, as a drug development candidate. GRN163 inhibited telomerase in a cell-free assay at 45 +/- 7 pM, and in various tumor cell lines at approximately 1 nM and approximately 0.3-1.0 micro M in the presence and absence of carriers, respectively. GRN163 was competitive with telomeric primer binding, primarily because of hybridization to human telomerase RNA (hTR) component. Tumor cells treated with GRN163 in culture underwent telomere shortening, followed by cellular senescence or apoptosis after a period of time that generally correlated with initial telomere length. In a flank DU145 (prostate cancer) xenograft model, parenterally administered GRN163 caused suppression of tumor growth in the absence of gross toxicity. These data demonstrate that GRN163 has significant potential for additional development as an anticancer agent.


Subject(s)
Oligonucleotides/pharmacology , Telomerase/antagonists & inhibitors , Amides/metabolism , Amides/pharmacology , Biological Availability , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Oligonucleotides/genetics , Oligonucleotides/metabolism , Oligonucleotides/pharmacokinetics , Phosphoric Acids/metabolism , Phosphoric Acids/pharmacology , RNA/genetics , RNA/metabolism , Telomerase/genetics , Telomerase/metabolism
14.
Arthritis Rheum ; 48(3): 668-74, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12632419

ABSTRACT

OBJECTIVE: Adenosine deaminase (ADA; EC 3.5.4.4) activity is elevated in the synovial fluid (SF) of patients with rheumatoid arthritis (RA). Since the antiinflammatory effect of methotrexate is reportedly associated with increased levels of extracellular adenosine, the present study was undertaken to clarify the role of 2 ADA isozymes, ADA1 and ADA2, in the pathogenesis of RA. METHODS: The activities of ADA1 and ADA2 were measured in SF from RA and osteoarthritis (OA) patients, in sera from RA patients, and in lysates prepared from mononuclear and polymorphonuclear cells from SF from RA patients, peripheral blood from RA patients, and fibroblast-like synoviocytes (FLS) from RA and OA patients. Also measured were the effects of proinflammatory cytokines on ADA1 activity and ADA messenger RNA (mRNA) expression in RA FLS, as determined using real-time polymerase chain reaction. The adenosine concentration in RA SF was measured by radioimmunoassay. RESULTS: The adenosine concentration in RA SF ranged from 0.027 microM to 0.508 microM (mean +/- SD 0.156 +/- 0.132 microM). At those concentrations, ADA1 would be expected to be functionally dominant due to its higher affinity for adenosine. ADA1 activity in RA SF (mean +/- SD 14.4 +/- 8.5 IU/liter) was significantly higher than that in OA SF (3.0 +/- 1.1 IU/liter) or RA sera (3.0 +/- 0.6 IU/liter); moreover, ADA1 activity in RA FLS lysate was the highest among the cell lysates tested. Proinflammatory cytokines did not affect ADA1 activity or ADA mRNA expression in RA FLS. CONCLUSION: Elevated ADA1 activity is an intrinsic characteristic of RA FLS, which likely contributes to the pathogenesis of RA by neutralizing the antirheumatic properties of endogenous adenosine.


Subject(s)
Adenosine Deaminase/biosynthesis , Arthritis, Rheumatoid/enzymology , Fibroblasts/enzymology , Osteoarthritis/enzymology , Synovial Fluid/enzymology , Adenosine/metabolism , Adenosine Deaminase/blood , Adenosine Deaminase/genetics , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Cells, Cultured , Fibroblasts/cytology , Humans , Isoenzymes , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/enzymology , Monokines/pharmacology , Neutrophils/cytology , Neutrophils/enzymology , Osteoarthritis/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synovial Fluid/cytology
15.
J Clin Endocrinol Metab ; 87(12): 5760-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466383

ABSTRACT

Estrogens play a key role in various target tissues. Enzymes involved in the biosynthesis and metabolism of these sex steroids also regulate estrogenic actions in these tissues. Estrone sulfate (E1S) is a major circulating plasma estrogen that is converted into the biologically active estrogen, estrone (E1), by steroid sulfatase (STS). E1 is also sulfated and reverted into E1S by estrogen sulfotransferase (EST). These two enzymes have recently been shown to play important roles in the in situ estrogen actions of various sex steroid-dependent human tumors. However, the distribution of STS and EST in normal adult and fetal human tissues remains largely unknown. Therefore, in this study, in addition to examining the tissue distribution of both STS and EST mRNA in human adult and fetal tissues using RT followed by quantitative PCR, we studied the activity of these enzymes using (3)H-labeled E1/E1S as substrates in the homogenates of various human adult tissues. We also examined the localization of STS and EST protein in human adult and fetal tissues using immunohistochemistry, and that of EST mRNA in the adult kidney using laser dissection microscopy and PCR. STS mRNA, enzyme activity, and immunoreactivity were either absent or detected at very low levels in all adult and fetal tissues examined in this study. EST mRNA expression, however, was detected in all of the tissues examined, except for adult spleen and pancreas. EST enzyme activities were consistent with those of mRNA expression in the great majority of the tissues examined. Marked EST immunoreactivity was detected in hepatocytes, adrenal gland (adult, zona fasciculate to the reticularis; fetus, fetal zone), and epithelial cells of the gastrointestinal tract, smooth muscle cells of the tunica media in aorta, Leydig cells of the testis, and syncytiotrophoblast of the placenta. Patterns of EST immunolocalization were similar between adult and fetal human tissues, but EST immunoreactivity was detected in the urinary tubules of adult kidney, whereas in the fetal kidney, it was localized in the interstitial cells surrounding the urinary tubules. In the adult kidney, the presence of EST mRNA was also confirmed in the cells of urinary tubules using laser dissection microscopy and RT-PCR. Although the number of human tissues available for examination in this study was limited, our results suggest that between the enzymes involved in estrogen activation or inactivation, EST and not STS is the more widely expressed enzyme in various peripheral tissues in humans. We speculate that EST may play an important role in protecting peripheral tissues from possible excessive estrogenic effects.


Subject(s)
Aging/metabolism , Arylsulfatases/metabolism , Fetus/metabolism , Sulfotransferases/metabolism , Adult , Aged , Aged, 80 and over , Arylsulfatases/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/metabolism , Steryl-Sulfatase , Sulfotransferases/genetics , Tissue Distribution
16.
Eur J Pharmacol ; 454(1): 85-93, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12409009

ABSTRACT

Adenosine protects against cellular damage and dysfunction under several adverse conditions including inflammation and ischemia. In this study, we examined the effects of 3-[1-(6,7-diethoxy-2-morpholinoquinazolin-4-yl)piperidin-4-yl]-1,6-dimethyl-2,4(1H,3H)-quinazolinedione hydrochloride (KF24345), an adenosine uptake inhibitor, on experimental acute pancreatitis induced by choline-deficient and ethionine-supplemented diet in mice. KF24345, administered with the diet onset and every 24 h thereafter, prevented hyperamylasemia, acinar cell injury and serum tumor necrosis factor-alpha elevation and ultimately decreased mortality. Therapeutic treatment with KF24345, which started 32 h after the diet onset, also decreased mortality. The beneficial effect of KF24345 on mortality was abolished by the pretreatment with 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385), a selective adenosine A(2A) receptor antagonist. An intravenous injection of KF24345 at 48 h after the diet onset increased plasma adenosine concentrations in mice with acute pancreatitis. These results suggest that KF24345 shows anti-pancreatitis effects via endogenous adenosine and adenosine A(2A) receptors. The adenosine uptake inhibition could be a new therapeutic approach for acute pancreatitis.


Subject(s)
Adenosine/antagonists & inhibitors , Neurotransmitter Uptake Inhibitors/pharmacology , Pancreatitis/drug therapy , Pyrimidinones/pharmacology , Quinazolines/pharmacology , Acute Disease , Adenosine/blood , Amylases/blood , Animals , Choline Deficiency/complications , Diet , Female , L-Lactate Dehydrogenase/blood , Mice , Organ Size , Pancreas/drug effects , Pancreatitis/etiology , Pancreatitis/mortality , Time Factors , Tumor Necrosis Factor-alpha/metabolism
17.
Pancreas ; 25(4): 387-92, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12409834

ABSTRACT

INTRODUCTION AND AIMS: Adenosine shows protective effects against cellular damage and dysfunction under several adverse conditions such as inflammation and ischemia. In the current study, we examined the effects of 3-[1-(6,7-diethoxy-2-morpholinoquinazolin-4-yl)piperidin-4-yl]-1,6-dimethyl-2,4(1,3 )-quinazolinedione hydrochloride (KF24345), an adenosine uptake inhibitor, on cerulein-induced acute pancreatitis in mice to investigate whether inhibition of adenosine uptake could ameliorate the severity of acute pancreatitis. METHODOLOGY: Acute pancreatitis was induced in mice with six intraperitoneal injections of cerulein (50 microg/kg each) at hourly intervals. RESULTS: The cerulein injection increased activities of serum amylase and lipase and caused pathologic changes such as interstitial edema, polymorphonuclear cell infiltration, and acinar cell necrosis in the pancreas. KF24345 (10 mg/kg p.o.) ameliorated all these changes observed in mice with acute pancreatitis, and the suppressing effect of KF24345 on the elevation in serum amylase activity was abolished by the treatment with 8-(p-sulfophenyl)theophylline, an adenosine receptor antagonist. In addition, 2-(aminocarbonyl)- -(4-amino-2,6-dichlorophenyl)-4-[5,5-bis-(4-fluorophenyl)pentyl]-1-piperazineacetamide (R75231) and dipyridamole, other adenosine uptake inhibitors, also decreased the elevated serum amylase activity. CONCLUSIONS: These are the first demonstrations that the adenosine uptake inhibitors ameliorate cerulein-induced acute pancreatitis in mice, and these data suggest that adenosine uptake inhibition could ameliorate the severity of acute pancreatitis in vivo.


Subject(s)
Adenosine/antagonists & inhibitors , Pancreatitis/drug therapy , Pyrimidinones/therapeutic use , Quinazolines/therapeutic use , Theophylline/analogs & derivatives , Acute Disease , Adenosine/metabolism , Amylases/blood , Animals , Biological Transport/drug effects , Ceruletide , Dipyridamole/pharmacology , Female , Lipase/blood , Mice , Mice, Inbred BALB C , Models, Chemical , Pancreatitis/chemically induced , Pancreatitis/enzymology , Pancreatitis/pathology , Piperazines/pharmacology , Pyrimidinones/antagonists & inhibitors , Pyrimidinones/chemistry , Quinazolines/antagonists & inhibitors , Quinazolines/chemistry , Theophylline/pharmacology
18.
Eur J Pharmacol ; 449(3): 293-300, 2002 Aug 09.
Article in English | MEDLINE | ID: mdl-12167472

ABSTRACT

This study evaluated the effects of KF24345 (3-[1-(6,7-diethoxy-2-morpholinoquinazolin-4-yl)piperidin-4-yl]-1,6-dimethyl-2,4(1H,3H)-quinazolinedione hydrochloride), a novel adenosine uptake inhibitor, on experimental glomerulonephritis induced in mice by two intravenous injections of rabbit anti-mouse glomerular basement membrane antiserum. Mice with glomerulonephritis showed continuous proteinuria and the histological evaluation revealed glomerular and tubular damage at 7 weeks after the first antiserum injection. KF24345 as well as prednisolone and cyclophosphamide significantly inhibited proteinuria and glomerular damage when it was orally administered once a day from 2 to 7 weeks. Prednisolone elevated plasma bilirubin and glutamic-pyruvic transaminase levels, and cyclophosphamide decreased erythrocytes. Moreover, both prednisolone and cyclophosphamide decreased spleen and thymus weights. KF24345 did not show this kind of side effects. These results demonstrate that KF24345 ameliorates glomerulonephritis with minimal side effects in mice, suggesting that the adenosine uptake inhibitor may be useful for the treatment of glomerulonephritis.


Subject(s)
Adenosine/metabolism , Glomerulonephritis/drug therapy , Neurotransmitter Uptake Inhibitors/therapeutic use , Pyrimidinones/therapeutic use , Quinazolines/therapeutic use , Alanine Transaminase/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Bilirubin/blood , Cyclophosphamide/therapeutic use , Erythrocyte Count , Glomerular Mesangium/metabolism , Glomerulonephritis/pathology , Immunosuppressive Agents/therapeutic use , Kidney Function Tests , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice , Neurotransmitter Uptake Inhibitors/adverse effects , Organ Size/drug effects , Periodic Acid-Schiff Reaction , Prednisolone/therapeutic use , Pyrimidinones/adverse effects , Quinazolines/adverse effects , Rabbits , Spleen/drug effects , Thymus Gland/drug effects
19.
J Pharmacol Exp Ther ; 300(1): 200-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752117

ABSTRACT

3-[1-(6,7-Diethoxy-2-morpholinoquinazolin-4-yl)piperidin-4-yl]-1,6-dimethyl-2,4(1H,3H)-quinazolinedione hydrochloride (KF24345) is a novel potent adenosine uptake inhibitor. KF24345 inhibited [(3)H]adenosine uptake into erythrocytes from human, mouse, rabbit, and hamster with IC(50) values of 59.5, 130.1, 104.2, and 30.9 nM, respectively. In mice, oral administration of KF24345 at 10 mg/kg almost completely inhibited the [(3)H]adenosine uptake into sampled blood cells at least up to 10 h of the administration. In this study, to examine whether the adenosine uptake inhibition exhibits anti-inflammatory effects, we determined the effects of KF24345 on lipopolysaccharide (LPS)-induced tumor necrosis factor-alpha (TNF-alpha) production and leukopenia in mice. KF24345 (10 mg/kg p.o.) significantly suppressed the elevation of serum TNF-alpha concentration after the LPS injection, and the suppressing effect of KF24345 was abolished by the treatment with 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol, a selective adenosine A(2) receptor antagonist, but not with 8-(noradamantan-3-yl)-1,3-dipropylxanthine, a selective adenosine A(1) receptor antagonist. KF24345 (10 mg/kg p.o.) also inhibited the decrease of leukocytes after the LPS injection, and 8-(p-sulfophenyl)theophylline, a nonselective adenosine receptor antagonist, completely reversed the inhibitory effect of KF24345. These results demonstrate that KF24345 inhibits LPS-induced TNF-alpha production and leukopenia via enhancing the effect of endogenous adenosine. It is thus suggested that the adenosine uptake inhibitor has anti-inflammatory effects in vivo and represents a novel therapeutic approach to the treatment of various inflammatory diseases.


Subject(s)
Adenosine/metabolism , Leukopenia/chemically induced , Lipopolysaccharides/antagonists & inhibitors , Neurotransmitter Uptake Inhibitors/pharmacology , Pyrimidinones/pharmacology , Quinazolines/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cricetinae , Erythrocytes/drug effects , Erythrocytes/metabolism , Humans , Lipopolysaccharides/pharmacology , Mesocricetus , Mice , Rabbits , Receptors, Purinergic P1/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...