Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Pharmacogenomics J ; 9(2): 90-102, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19188929

ABSTRACT

AZD1152 is a highly selective Aurora B kinase inhibitor currently undergoing Phase I and II clinical evaluation in patients with acute myelogenous leukemia and advanced solid malignancies. We have established two AZD1152-resistant cell lines from SW620 colon and MiaPaCa pancreatic carcinoma lines, which are >100-fold resistant to the active metabolite of AZD1152, AZD1152 HQPA and interestingly, cross-resistant to the pan-Aurora kinase inhibitor, VX-680/MK0457. Using whole-genome microarray analysis and comparative genomic hybridization, we were able to identify MDR1 and BCRP as the causative genes that underlie AZD1152 HQPA-resistance in these models. Furthermore, the upregulation of either of these genes is sufficient to render in vivo tumor growth insensitive to AZD1152. Finally, the upregulation of MDR1 or BCRP is predictive of tumor cell sensitivity to this agent, both in vitro and in vivo. The data provide a genetic basis for resistance to Aurora kinase inhibitors, which could be utilized to predict clinical response to therapy.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Organophosphates/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Cell Survival/drug effects , Comparative Genomic Hybridization , Dose-Response Relationship, Drug , Gene Expression Profiling/methods , Humans , Inhibitory Concentration 50 , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Piperazines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays
2.
Cell Death Differ ; 16(3): 498-511, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19079141

ABSTRACT

Small-molecule inhibitors of the Aurora A and B kinases interfere with mitotic centrosome function and disrupt the mitotic spindle assembly checkpoint resulting in polyploidization and apoptosis of proliferating cells. As such, several Aurora kinase inhibitors are at various stages of clinical development as anticancer agents. To identify candidate apoptosis-sensitizing genes that could be exploited in combination with Aurora kinase inhibitors in malignant glioma, we have carried out global gene expression analysis in a D54MG glioma cell derivative treated with three Aurora kinase inhibitors chosen for their distinctive selectivities: MLN8054 (Aurora A-selective), AZD1152 (Aurora B-selective), and VX-680 (Aurora A/B). The modulation of apoptotic gene expression by p53 under these conditions was ascertained, as p53 expression can be toggled on and off in this D54MG derivative by virtue of a stable, inducible, p53-targeting short hairpin RNA (D54MG(shp53)). This analysis identified the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) death receptor, TRAIL receptor 2 (TRAIL-R2), as an apoptosis-sensitizing gene induced selectively following inhibition of Aurora B. In glioma cell lines where TRAIL-R2 was induced following polyploidization, the sensitivity, kinetics, and magnitude of TRAIL-mediated apoptosis were enhanced. Our data shed light on the apoptotic program induced during polyploidization and suggest that TRAIL-R2 activation is a putative point of therapeutic intervention in combination with inhibitors of Aurora B.


Subject(s)
Brain Neoplasms , Gene Expression Regulation, Neoplastic , Glioma , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Aurora Kinase B , Aurora Kinases , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/physiology , Cell Line, Tumor , Gene Expression Profiling , Glioma/metabolism , Glioma/pathology , Humans , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Leukemia ; 22(1): 138-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17943175

ABSTRACT

Internal tandem duplications (ITDs) of fms-like tyrosine kinase 3 (FLT3) receptor play an important role in the pathogenesis of acute myeloid leukemia (AML) and represent an attractive therapeutic target. ABT-869 has demonstrated potent effects in AML cells with FLT3-ITDs. Here, we provide further evidence that ABT-869 treatment significantly downregulates cyclins D and E but increases the expression of p21 and p27. ABT-869 induces apoptosis through downregulation of Bcl-xL and upregulation of BAK, BID and BAD. We also evaluate the combinations of ABT-869 and chemotherapy. ABT-869 demonstrates significant sequence-dependent synergism with cytarabine and doxorubicin in cell lines and primary leukemia samples. The optimal combination was validated in MV4-11 xenografts. Low-density array analysis revealed the synergistic interaction involved in downregulation of cell cycle and mitogen-activated protein kinase pathway genes. CCND1 and c-Mos were the most significantly inhibited targets on both transcriptional and translational levels. Treatment with short hairpin RNAs targeting either CCND1 or c-Mos further sensitized MV4-11 cells to ABT-869. These findings suggest that specific pathway genes were further targeted by adding chemotherapy and support the rationale of combination therapy. Thus, a clinical trial using sequence-dependent combination therapy with ABT-869 in AML is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Cycle Proteins/metabolism , Indazoles/therapeutic use , Leukemia, Myeloid/drug therapy , Mitogen-Activated Protein Kinases/metabolism , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins c-mos/metabolism , Signal Transduction/drug effects , Acute Disease , Animals , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cytarabine/administration & dosage , Down-Regulation , Doxorubicin/administration & dosage , Drug Synergism , Drug Therapy, Combination , Gene Expression Profiling , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, SCID , Proto-Oncogene Proteins c-mos/genetics , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Transplantation, Heterologous
4.
Biochem Biophys Res Commun ; 325(3): 683-90, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15541343

ABSTRACT

Histone deacetylase (HDAC) inhibitors induce the hyperacetylation of nucleosomal histones in carcinoma cells resulting in the expression of repressed genes that cause growth arrest, terminal differentiation, and/or apoptosis. In vitro selectivity of several novel hydroxamate HDAC inhibitors including succinimide macrocyclic hydroxamates and the non-hydroxamate alpha-ketoamide inhibitors was investigated using isolated enzyme preparations and cellular assays. In vitro selectivity for the HDAC isozymes (HDAC1/2, 3, 4/3, and 6) was not observed for these HDAC inhibitors or the reference HDAC inhibitors, MS-275 and SAHA. In T24 and HCT116 cells these compounds caused the accumulation of acetylated histones H3 and H4; however, the succinimide macrocyclic hydroxamates and the alpha-ketoamides did not cause the accumulation of acetylated alpha-tubulin. These data suggest "selectivity" can be observed at the cellular level with HDAC inhibitors and that the nature of the zinc-chelating moiety is an important determinant of activity against tubulin deacetylase.


Subject(s)
Amides/pharmacology , Breast Neoplasms/enzymology , Fibrosarcoma/enzymology , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Amides/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors , Fibrosarcoma/pathology , Histone Deacetylases/chemistry , Humans , Hydroxamic Acids/chemistry
5.
Shock ; 11(3): 180-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10188770

ABSTRACT

The effects of lipopolysaccharide (LPS) on the central nervous system, one of the first organs to be affected by sepsis, are still incompletely understood. Rat microglia (BMphi) constitute the main leukocyte-dependent source of reactive oxygen species in the central nervous system. The in vitro effect of LPS on agonist-stimulated superoxide (O2-) generation from BMphi appears controversial. Our purpose was to determine the time- and concentration-dependent effect of Escherichia coil LPS on phorbol-12 myristate 13-acetate-stimulated O2- generation from BMphi. Our results demonstrate that BMphi O2- generation in vitro peaked 17 h after stimulation of with .3 ng/mL LPS. Furthermore, stimulation of BMphi with LPS for 17 h resulted in the following concentration-dependent responses: .1-1 ng/mL LPS induced no prior mediator generation but potently enhanced subsequent phorbol-12 myristate 13-acetate-stimulated O2- generation; 3-10 ng/mL LPS caused nitric oxide, tumor necrosis factor-alpha (TNF-alpha), thromboxane B2 and matrix metalloproteinase-9 release although partially inhibiting ensuing phorbol-12 myristate 13-acetate-stimulated O2- generation; 30-100 ng/mL LPS, maximized nitric oxide, TNF-alpha, thromboxane B2, matrix metalloproteinase-9 generation with concomitant lactic dehydrogenase release although strongly deactivating successive phorbol-12 myristate 13-acetate-stimulated O2 production. Our in vitro studies suggest that enhanced release of these four mediators (nitric oxide, TNF-alpha, thromboxane B2, and matrix metalloproteinase-9) during stimulation of BMphi with LPS might play a critical role in the subsequent ability of BMphi to generate O2- in vivo. Potential clinical implications of our findings are suggested by the fact that LPS levels similar to the ones used in this study have been observed in cerebrospinal fluid both in Gram-negative meningitis and sepsis.


Subject(s)
Escherichia coli/metabolism , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides/pharmacology , Microglia/metabolism , Superoxides/metabolism , Animals , Animals, Newborn , Anions/metabolism , Collagenases/drug effects , Collagenases/metabolism , Dose-Response Relationship, Drug , L-Lactate Dehydrogenase/drug effects , Matrix Metalloproteinase 9 , Metalloendopeptidases/drug effects , Metalloendopeptidases/metabolism , Microglia/drug effects , Microglia/microbiology , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate/pharmacology , Thromboxane B2/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Biochem Pharmacol ; 57(3): 291-302, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9890556

ABSTRACT

The monocytic cell line THP-1 can be induced to express and release tumor necrosis factor alpha (TNFalpha) and both TNFalpha receptors (p55 and p75) upon exposure to bacterial lipopolysaccharide (LPS). The broad-spectrum matrix metalloprotease (MMP) inhibitors [4-(N-hydroxyamino)-2R-isobutyl-3S-(phenylthiomethyl)succinyl]-L-p henylalanine-N-methylamide (GI-129471) and marimastat [2S-[N4(R*),2R*,3S*]]-N4[2,2-dimethyl-1-[(methylamino)carbonyl]propyl]-N 1,2-dihydroxy-3-(2-methylpropyl)butanediamide (BB-2516) were effective inhibitors of LPS-induced TNFalpha (soluble) release with IC50 values of 0.2 and 4.0 microM, respectively. Upon LPS stimulation, the expression of pro-TNFalpha (membrane associated) on the cell surface (FACS analysis) could not be observed. However, in the presence of GI-129471, a concentration-dependent increase in TNFalpha surface expression was observed. Peak expression (percentage of cells expressing pro-TNFalpha and mean fluorescence units) in the presence of GI-129471 was at 2 hr, and steadily declined to return to near control levels by 8 hr. This time course was similar to TNFalpha release, which also peaked at 2-4 hr after LPS exposure and then declined. Stimulation of THP-1 cells with LPS + phorbol myristate acetate increased the percentage of cells expressing pro-TNFalpha by 10-fold. In the presence of GI-129471, these increases were augmented further and peaked between 2 and 4 hr, but also returned to near control levels of expression by 24 hr. This was in contrast to the release of soluble TNFalpha, which continued to accumulate over a 24-hr time course. TNFalpha receptor I (p55, TNFRI) and II (p75, TNFRII) shedding was also inhibited by GI-129471 (IC50 = 1.5 and 3.1 microM, respectively) and BB-2516 (IC50 = 14 and 15 microM, respectively). Unlike pro-TNFalpha surface expression, surface expression of both TNFalpha receptors steadily increased over 72 hr. In contrast to pro-TNFalpha surface expression, TNFRI surface expression was not augmented by these MMP inhibitors in THP-1 cells after LPS stimulation. Surface expression of TNFRII was augmented by these MMP inhibitors. These results suggest that even in the continued presence of LPS stimulation and an inhibitor of TNFalpha processing, the augmented surface expression of TNFalpha is transient. The potential "deleterious" implications of high levels of surface pro-TNFalpha expression in the presence of these inhibitors may be lessened by its transient nature.


Subject(s)
Metalloendopeptidases/antagonists & inhibitors , Monocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Cell Line , Cell Membrane/metabolism , Cytokines/biosynthesis , Humans , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology
7.
Inflamm Res ; 47(8): 339-44, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9754868

ABSTRACT

OBJECTIVE AND DESIGN: To determine whole blood levels of sirolimus, a macrolide antibiotic in the rat developing adjuvant arthritis (AA) model after dosing orally with two different vehicles, and whether combinational doses of sirolimus and cyclosporin A (CsA) produced additive or synergistic inhibitory effects in this model. MATERIAL: Male Lewis rats (150-180g). TREATMENT: Arthritis was induced by the injection (0.5 mg/ rat) of heat-killed Mycobacterium butyricum suspended in light mineral oil. Drugs were administered orally either in fine suspension (0.5% Tween 80) or in emulsion (phosal 50 PG in 1% Tween 80) at doses of 0.1 to 5 mg/kg in a 7 day, MWF or daily regimen. METHOD: Paw volumes (ml) were measured by automated mercury plethysmograph and sirolimus concentrations in whole blood were quantitated by liquid chromatography/ mass spectroscopy. RESULTS: At 72h (7 days after adjuvant) after receiving the third oral dose (4.5 mg/kg p.o.), the phosal vehicle resulted in higher sirolimus blood levels (2.5 ng/ml) than in Tween 80 (1.6 ng/ml). After the rats received the last oral dose on day 14, (7 total doses of sirolimus at 4.5 mg/kg) the sirolimus blood levels (2h after the last dose) were about 2 times higher for the phosal dosed rats (9.8 ng/ml) compared to Tween 80 dosed rats (4.6ng/ml). Even 24h after the last dose, sirolimus blood levels were still elevated in the phosal dosed rats (0.8 ng/ml) relative to 0.5% Tween 80 dosed rats (0.5 ng/ml). At day 16 in the rat developing model, sirolimus, when given in phosal vehicle, produced an ED50 of 0.28 mg/ kg (i.e. inhibition of uninjected paw edema) that was about 5.5 times lower than using 0.5% Tween 80 as the suspending agent (ED50 = 1.6mg/kg). When combining sirolimus and CsA using precalculated doses for producing an additive effect in this adjuvant model, an additive inhibitory effect on uninjected paw edema was observed at equal combinational doses of 0.5 and 2 mg/kg, respectively. CONCLUSIONS: The phosal vehicle used in administering sirolimus increases the absorption and whole blood levels in the rat and the elevated blood levels correlated positively with the therapeutic effect in the rat developing AA model. In addition, combination therapy using sirolimus and CsA produced an additive effect in rat developing AA.


Subject(s)
Arthritis, Experimental/metabolism , Cyclosporine/pharmacokinetics , Drug Therapy, Combination , Sirolimus/pharmacokinetics , Administration, Oral , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/microbiology , Cyclosporine/administration & dosage , Cyclosporine/therapeutic use , Disease Models, Animal , Dosage Forms , Male , Mycobacterium/immunology , Rats , Rats, Inbred Lew/metabolism , Rats, Inbred Lew/microbiology , Sirolimus/administration & dosage , Sirolimus/therapeutic use
8.
Life Sci ; 62(26): PL401-7, 1998.
Article in English | MEDLINE | ID: mdl-9651113

ABSTRACT

Pseudopterosin E (PSE), a C-10 linked fucose glycoside and pseudopterosin A (PSA), a C-9 xylose glycoside isolated from the marine gorgonian Pseudopterogorgia elisabethae were both effective in reducing PMA-induced mouse ear edema when administered topically (ED50 (microg/ear) PSE(38), PSA(8)) or systemically (ED50 (mg/kg, i.p.) PSE (14), PSA (32)). Both compounds exhibited in vivo analgesic activity in phenyl-p-benzoquinone-induced writhing (ED50 (mg/kg, i.p.) PSE(14), PSA(4). PSE inhibited zymosan-induced writhing (ED50 = 6 mg/kg, i.p.), with a concomitant dose-dependent inhibition of peritoneal exudate 6-keto-prostaglandin F1alpha (ED50 = 24 mg/kg) and leukotriene C4 (ED50 = 24 mg/kg). In vitro, the pseudopterosins were inactive as inhibitors of phospholipase A2, cyclooxygenase, cytokine release, or as regulators of adhesion molecule expression. PSA inhibited prostaglandin E2 and leukotriene C4 production in zymosan-stimulated murine peritoneal macrophages (IC50 = 4 microM and 1 microM, respectively); however, PSE was much less effective. These data suggest that the pseudopterosins may mediate their anti-inflammatory effects by inhibiting eicosanoid release from inflammatory cells in a concentration and dose-dependent manner.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cnidaria/chemistry , Diterpenes/pharmacology , Glycosides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Cell Line , Cytokines/metabolism , Diterpenes/isolation & purification , Edema/chemically induced , Edema/prevention & control , Eicosanoids/metabolism , Glycosides/isolation & purification , Humans , Macrophages, Peritoneal/drug effects , Mice , Neutrophils/drug effects , Neutrophils/metabolism , Pain Measurement/drug effects
10.
J Pharmacol Exp Ther ; 280(2): 1065-74, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9023325

ABSTRACT

These studies examined the signal transduction mechanisms by which prostaglandin (PG) E2 production can occur in human amnionic WISH cells in response to the stimuli okadaic acid, interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, phorbol-12-myristate-13-acetate (PMA) or combinations of PMA with IL-1beta or TNF-alpha. We also investigated whether WISH cells are capable of producing TNF-alpha or IL-1beta in response to stimulation, because these cytokines can be produced in an autocrine fashion to perpetuate an inflammatory response. Our data indicate that the magnitude of PGE2 production induced by a given stimulus correlated temporally with the level of PGH synthase-2 (PGHS-2) protein. PMA or IL-1beta induced PGE2 production 2 to 4 hr after treatment, whereas the combination of these agents produced the most rapid induction 2 hr after treatment. Only okadaic acid induced the production of both PGE2 and TNF-alpha, after a lag of 12 to 18 hr. PGE2 production by all stimuli was inhibited by dexamethasone, the IL-1 receptor antagonist (IL-1ra), the specific PGHS-2 inhibitor NS-398 and the protein kinase inhibitor staurosporin. In contrast, TNF-alpha production in response to okadaic acid was inhibited by the TNF-converting enzyme inhibitor GI 129471 and staurosporin but was unaffected by either IL-1ra, dexamethasone or NS-398. We conclude that WISH cells are capable of producing bioactive proinflammatory mediators such as TNF-alpha and PGE2 through separable intracellular signal transduction mechanisms. The ability of IL-1ra to reduce PGE2 production caused by all stimuli used suggests an autocrine role for IL-1 in PGHS-2 induction in these cells.


Subject(s)
Interleukin-1/pharmacology , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Amnion , Base Sequence , Blotting, Northern , Cell Line , Cyclooxygenase 2 , DNA Primers , Dinoprostone/analysis , Enzyme Induction , Female , Humans , Inflammation , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Kinetics , Membrane Proteins , Models, Biological , Molecular Sequence Data , Okadaic Acid/pharmacology , Polymerase Chain Reaction , Pregnancy , RNA, Messenger , Recombinant Proteins/pharmacology , Sialoglycoproteins/pharmacology , Tetradecanoylphorbol Acetate
12.
J Pharmacol Exp Ther ; 279(2): 633-44, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8930166

ABSTRACT

The purpose of this investigation was to pharmacologically probe the signaling pathways thought to be involved in protein kinase C (PKC)-stimulated superoxide anion (O2-) generation in all-trans retinoic acid-treated human promyelocytic HL-60 cell line (HL-60), targeting PKC, mitogen-activated protein kinase (MAPK), MAPK kinase (MEK), protein serine-threonine phosphatase(s) (PSP), protein tyrosine kinase(s) (PTK) and phosphatase(s) (PTP), secretory phospholipase A2, cyclooxygenase (CO) and 5-lipoxygenase with selected inhibitors. The following agents inhibited phorbol 12-myristate 13-acetate-stimulated O2- generation significantly in the all-trans retinoic acid-treated HL-60 cells (expressed as percentage of control, P < .05): 1) PKC inhibitors: staurosporine (100 nM, 3 +/- 1%); Ro 31-8220 (1 microM, 3 +/- 2%); sphingosine (100 microM, 15 +/- 7%); 2) PSP 1 and 2a inhibitors, okadaic acid (10 microM, 35 +/- 1%); calyculin A (10 microM, 73 +/- 1%); 3) MAPK inhibitor: SB-203580 (100 microM, 62 +/- 1%); 4) PTP inhibitors: phenylarsine oxide (1 microM, 12 +/- 9%); diamide (1 mM, 21 +/- 11%); and 5) secretory phospholipase A2 inhibitors: manoalide (1 microM, 24 +/- 10%); scalaradial (1 microM, 11 +/- 4%). Exogenously added arachidonic acid-stimulated O2- generation in a time- and dose-dependent manner. The following inhibitors enhanced or did not significantly affect phorbol 12-myristate 13-acetate-stimulated O2- generation (expressed as percentage of control): 1) PTK inhibitors: genistein (100 microM, 69 +/- 12%); CGP 53716 (100 microM, 67 +/- 10%); herbimycin A (10 microM, 67.4 +/- 1%); 2) PSP 2b inhibitors: cyclosporin A (30 microM, 71 +/- 5%); FK506 (30 microM, 88 +/- 7%); 3) CO inhibitor: indomethacin (100 microM, 111 +/- 12%); 4) 5-lipoxygenase inhibitor: WY 50,295 (100 microM, 140 +/- 23%); 5) MEK inhibitor: PD98059 (100 microM, 94 +/- 6.7%); and 6) the PTP inhibitor: orthovanadate (100 microM, 131 +/- 25%). Our pharmacological study suggests that, in neutrophil-like HL-60 cells, the signaling pathways leading to PMA-stimulated O2- generation appear to involve PKC, MAPK, phospholipase A2, arachidonic acid, PSP 1 and 2a and PTP. Furthermore, PTK, MEK, CO, 5-lipoxygenase and PSP 2b do not appear to participate in the modulation of PKC-stimulated O2- generation.


Subject(s)
Enzyme Inhibitors/pharmacology , HL-60 Cells/metabolism , Neutrophils/metabolism , Protein Kinase C/physiology , Superoxides/metabolism , Arachidonic Acid/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Humans , Mitogen-Activated Protein Kinase Kinases , Phospholipases A/physiology , Phospholipases A2 , Protein Kinases/physiology , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology
14.
Cell Signal ; 7(8): 827-35, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8593251

ABSTRACT

The regulation of phosphodiesterase-4 (PDE4) by various phospholipids was explored using PDE4s partially purified from U937 cells. Preincubation (5 min, 4 degrees C) of the large molecular weight PDE4 denoted "Peak 2 PDE4" with mixed phosphatidic acids (PAs) produced a 2-fold increase in its Vmax without changing its Km (approximately 2 microM) for cyclic AMP. This "activation" was not limited to PAs with specific fatty acid substituents: Synthetic PAs containing saturated and/or unsaturated fatty acids 16-20 carbons long produced similar effects. Lysophosphatidic acids (LPAs) and phosphatidylserines (PSs) also induced PDE4 activation, whereas phosphatidylcholines (PCs), phosphatidylethanolamines (PEs) and diacylglycerol did not. Antibodies to a peptide region near the PDE4 catalytic site specifically inhibited PA-induced activation. The data demonstrate that anionic phospholipids can act as non-essential activators of a leukocyte PDE4, and suggest biochemical crosstalk between phospholipid-dependent and cyclic AMP-dependent signalling pathways in human leukocytes.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Cyclic AMP/metabolism , Monocytes/drug effects , Phospholipids/pharmacology , Phosphoric Diester Hydrolases/metabolism , Amino Acid Sequence , Animals , Antibody Formation , Cyclic GMP/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Activation/drug effects , Fatty Acids/analysis , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Lysophospholipids/pharmacology , Molecular Sequence Data , Monocytes/enzymology , Neoplasm Proteins/metabolism , Peptide Fragments/immunology , Phosphatidic Acids/pharmacology , Phosphatidylcholines/pharmacology , Phospholipids/chemistry , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/immunology , Pyrrolidinones/pharmacology , Rabbits , Rolipram , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects
15.
Biochem Pharmacol ; 50(7): 913-22, 1995 Sep 28.
Article in English | MEDLINE | ID: mdl-7575673

ABSTRACT

The marine natural products manoalide and scalaradial are potent anti-inflammatory agents that inactivate the enzyme phospholipase A2 (PLA2) in vitro. To study the mechanism of inhibition of prostaglandin E2 (PGE2) production in human monocytes by manoalide and scalaradial, lipopolysaccharide (LPS)-induced prostaglandin biosynthesis and induction of prostaglandin H synthase (PGHS) were evaluated. LPS (10 ng/mL) and interleukin-1 beta (IL-1 beta, 50-1000 ng/mL) but not tumor necrosis factor alpha (TNF alpha, 300 ng/mL) induced the expression of the PGHS-2 isoform as determined by immunoblot analysis with a specific polyclonal antibody for PGHS-2. Manoalide and scalaradial (1-10 microM) inhibited LPS-induced endogeneous PGE2 production, reduced the LPS-induced PGHS activity, and reduced the expression of PGHS-2. Indomethacin [a PGHS inhibitor (0.01 to 0.1 microM)], zileuton [a 5-lipoxygenase inhibitor (3-10 microM)], and WEB-2806 [a platelet-activating factor (PAF) antagonist (30 microM)] did not affect the LPS-induced expression of PGHS-2 in human monocytes. These results suggest that modulation of lipid mediator production by manoalide or scalaradial may not be involved in the observed effects on the expression of PGHS-2. Manoalide and scalaradial also inhibited the release of IL-1 beta and TNF alpha from LPS-stimulated monocytes. Expression of PGHS-2 induced by either LPS or IL-1 beta was blocked by the IL-1 receptor antagonist (IL-1ra, 2 micrograms/mL) but not by rolipram, a phosphodiesterase IV inhibitor that inhibits TNF alpha but not IL-1 beta release. Similar to LPS, IL-1 beta-induced PGHS-2 expression was apparently not regulated by lipid mediators such as prostaglandins, leukotrienes or PAF as determined with specific inhibitors and antagonists. Scalaradial and to some extent manoalide were capable of blocking the IL-1 beta-induced expression of PHGS-2. These results indicate that IL-1 beta is the predominant cytokine responsible for the induction of PGHS-2 in the human monocyte. Furthermore, marine natural products such as scalaradial have novel effects on the IL-1 beta-mediated induction of PGHS-2 in human monocytes, which appears to be independent of effects on lipid mediator production.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Homosteroids/pharmacology , Monocytes/drug effects , Monocytes/enzymology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Terpenes/pharmacology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Eicosanoids/biosynthesis , Enzyme Induction/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-1/biosynthesis , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Sesterterpenes , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Pharmacol Exp Ther ; 274(1): 427-36, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7616427

ABSTRACT

Ninety minutes after i.v. injection of Escherichia coli lipopolysaccharide (LPS) (1 mg/kg) into rats, phorbol 12-myristate 13-acetate (PMA)-stimulated superoxide anion (O2-) secretion was enhanced in suspensions of in vivo LPS-treated alveolar macrophages (AM phi) when compared with saline (SAL)-treated AM phi. The purpose of this investigation was to dissect the in vitro mechanism of PMA-stimulated O2- generation in both LPS and SAL-treated rat AM phi, with a panel of inhibitors of protein kinase C (PKC), protein serine-threonine phosphatase(s) (PSP), protein tyrosine kinase(s) (PTK) and phosphatase(s) (PTP), phospholipase A2 (PLA2), cyclooxygenase (CO) and 5-lipoxygenase (5-LO). The following agents blocked PMA-stimulated O2- generation in both LPS- and SAL-treated AM phi (expressed as percentage of control): 1) PKC inhibitors: staurosporine: 100 nM, 7.0% (LPS) and 5.6% (SAL); sphingosine: 10 microM, 21% (LPS) and 10.5% (SAL); 2) PTK inhibitor: genistein: 100 microM, 44% (LPS) and 31% (SAL); 3) PTP inhibitors: phenylarsine oxide, 10 microM, 12.1% (LPS) and 18% (SAL); diamide, 1000 microM, 10.1% (LPS) and 10.5% (SAL); and 4) PLA2 inhibitors: manoalide: 1 microM, 29.3% (LPS) and 5.2% (SAL); scalaradial: 1 microM, 7.7% (LPS) and 7.1% (SAL); and WAY 125,984: 10 microM, 17.1% (LPS) and 14.5% (SAL). In addition, it was observed that exogenously added arachidonic acid (AA)-stimulated O2- generation in a time- and dose-dependent manner in both LPS and SAL-treated AM phi. The following inhibitors enhanced or did not affect PMA-stimulated O2- generation in LPS- and SAL-treated AM phi (expressed as percentage of of control): 1) PSP inhibitors: okadaic acid: 0.5 microM, 117% (LPS) and 153% (SAL); calyculin A: 1 microM, 112% (LPS) and 101% (SAL); 2) CO and 5-LO inhibitors: indomethacin: 10 microM, 107% (LPS) and 90% (SAL); WY 50, 295: 1 microM, 99% (LPS) and 103% (SAL); and 3) the PTP inhibitor orthovanadate upon prolonged preincubation. In both in vivo LPS- or SAL-primed AM phi, PMA-stimulated O2- generation appears to be modulated by PKC, PLA2, AA, PTK, PTP and PSP. No modulatory role was evident for either CO or 5-LO metabolites. These findings might bear on the design of therapeutic approaches for the modulation of O2- release by AM phi in the early stages of sepsis and adult respiratory distress syndrome.


Subject(s)
Arachidonic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Superoxides/metabolism , Animals , Macrophages, Alveolar/metabolism , Male , Phospholipases A/antagonists & inhibitors , Phospholipases A/physiology , Phospholipases A2 , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/physiology , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/physiology , Rats , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate/pharmacology
17.
Adv Pharmacol ; 32: 31-66, 1995.
Article in English | MEDLINE | ID: mdl-7748798

ABSTRACT

The area of PLA2 research has grown immensely over the past 20 years. There is a better understanding of the kinetics, or factors that affect the kinetics, of the different forms of PLA2. New forms of PLA2 are being discovered, such as the cPLA2, which fit the role of an intracellularly regulated enzyme. Multiple forms of PLA2 tend to complicate the elucidation of the cellular mechanisms that regulate AA release and the subsequent eicosanoid production. Because of the factors that affect PLA2 kinetics and the unknown nature of the PLA2 that regulates AA release (there may be more than one), it has been difficult to design or isolate specific inhibitors. This review discussed selected classes of inhibitors because these have generated the most intense research in the field. There is a multitude of structurally diverse compounds reported in the literature that have been reported to be inhibitors of PLA2 in vitro and some have been reported to have anti-inflammatory activity (Wilkerson, 1990; Connolly and Robinson, 1993a). It is clear from a brief survey of the literature that the bulk of PLA2 inhibitors have topical anti-inflammatory activity. This may be due to the nature of these inhibitors: because they are hydrophobic they may be more readily absorbed in the skin whereas when given orally they may not be absorbed. To data, manoalide has been clinically evaluated in man and a new Bristol-Myers Squibb retenoid derivative may enter clinical trials for psoriasis (BMS-181162 (XVI)); however, there are no PLA2 inhibitors on the market or significantly advanced in clinical development (Table III). This indicates the lack of understanding of this enzyme for the development of relevant inhibitors, which is related to the lack of understanding of the relevant PLA2 that regulates AA release and eicosanoid biosynthesis. The concept of regulation of eicosanoid biosynthesis by PLA2 inhibition and decreased AA availability still remains a viable therapeutic approach for the treatment of inflammatory diseases. The proof of this concept has not been obtained because of the complex nature of PLA2 and the multiple forms of PLA2 in the cell. Clinical results with cyclooxygenase inhibitors and recent clinical results with inhibitor of 5-lipoxygenase demonstrate that if inhibition of PLA2 results in reduction in both lipid mediators, a good anti-inflammatory compound should result. The added advantage of PLA2 inhibitors would be the reduction of PAF levels; however, the clinical results with potent and specific PAF antagonists has been less encouraging about the potential benefits of reduction in PAF levels.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Enzyme Inhibitors/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Animals , Annexins/pharmacology , DNA, Antisense , Drug Design , Humans , Models, Biological , Phospholipases A2 , Phospholipids/pharmacology
19.
Skin Pharmacol ; 8(6): 300-8, 1995.
Article in English | MEDLINE | ID: mdl-8688196

ABSTRACT

Several novel inhibitors of human synovial fluid phospholipase A2 (HSF-PLA2) were evaluated in cellular models of inflammatory mediator release (murine macrophage and human neutrophil) and topical in vivo inflammatory skin models in mice to ascertain the scope of effects which might be observed for PLA2 inhibitors. Potent inhibition of HSF-PLA2 in vitro can be observed with compounds such as scalaradial and ellagic acid, which both have IC50 values of 0.02 microM (using autoclaved [3H]-arachidonic-acid (AA)-labelled Escherichia coli membranes as substrate). Luffariellolide, a manoalide analog, and aristolochic acid are less potent (IC50 = 5 and 46 microM, respectively) in this assay. An interesting observation is that ellagic acid in cellular assays does not inhibit macrophage eicosanoid production and only 30% inhibition of PAF biosynthesis can be obtained at 50 microM in the human neutrophil. Possibly due to its irreversible mechanism of action, scalaradial retained its potent activity in both the macrophage (IC50 for PGE2 production = 0.05 microM) and neutrophil assays (IC50 for PAF biosynthesis = 1 microM). Aristolochic acid is active in these cellular assays (macrophage IC50 = 2.5 microM and neutrophil IC50 = 100 microM), but is consistently less active than either scalaradial or luffariellolide. The relative potencies of these compounds were determined in several murine in vivo inflammatory models such as oxazolone contact hypersensitivity, AA-induced ear edema and phorbol ester (PMA)-induced ear edema. In the mouse model of oxazolone contact hypersensitivity, these PLA2 inhibitors have little effect (< or = 30% inhibition at 400 micrograms/ear) with scalaradial and luffariellolide being less effective than either aristolochic or ellagic acid. PMA-induced ear edema was effectively inhibited by scalaradial, luffariellolide and aristolochic acid (ED50 = 70, 50 and 50 micrograms/ear, respectively) whereas ellagic acid was less effective (ED50 = 230 micrograms/ear). In AA-induced ear edema, these PLA2 inhibitors had minimal effects, as would be expected for compounds which inhibit PLA2. These results, especially those of ellagic acid, suggest that caution should be taken in the extrapolation of potency against a purified human extracellular type PLA2 to the scope of activities these compounds might have in the cellular and in vivo models. The consistency of scalaradial and luffariellolide may be inherent to their irreversible mechanism of action, which is a factor to be accounted for in the extrapolation of enzyme data to cellular and in vivo models.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Phospholipases A/antagonists & inhibitors , Animals , Eicosanoids/metabolism , Ellagic Acid/pharmacology , Homosteroids/pharmacology , Humans , Inflammation/prevention & control , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Phospholipases A2 , Platelet Activating Factor/biosynthesis , Sesterterpenes , Skin , Synovial Fluid/enzymology , Terpenes/pharmacology
20.
Trends Pharmacol Sci ; 14(3): 92-8, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8488570

ABSTRACT

The phospholipase A2 enzymes are important components of the cellular machinery that responds to inflammatory stimuli and maintains cell homeostasis by membrane remodelling. Their role as the rate-limiting step in the production of pro-inflammatory lipid mediators makes these enzymes an important therapeutic target for the treatment of inflammatory disorders. Keith Glaser and colleagues explain how the two major groups of phospholipase A2, the secretory and cytosolic forms, are very different both structurally and enzymatically. Understanding the relative contributions of these different forms of phospholipase A2 to physiological and pathological conditions requires greater insight into their cellular regulation and the development of selective inhibitors.


Subject(s)
Phospholipases A/physiology , Animals , GTP-Binding Proteins/physiology , Humans , Models, Molecular , Phospholipases A/antagonists & inhibitors , Phospholipases A/metabolism , Phospholipases A2 , Stereoisomerism , Synovial Fluid/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...