Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 8(5): e1002691, 2012.
Article in English | MEDLINE | ID: mdl-22589723

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis infection, is a major cause of morbidity and mortality in the world today. M. tuberculosis hijacks the phagosome-lysosome trafficking pathway to escape clearance from infected macrophages. There is increasing evidence that manipulation of autophagy, a regulated catabolic trafficking pathway, can enhance killing of M. tuberculosis. Therefore, pharmacological agents that induce autophagy could be important in combating tuberculosis. We report that the antiprotozoal drug nitazoxanide and its active metabolite tizoxanide strongly stimulate autophagy and inhibit signaling by mTORC1, a major negative regulator of autophagy. Analysis of 16 nitazoxanide analogues reveals similar strict structural requirements for activity in autophagosome induction, EGFP-LC3 processing and mTORC1 inhibition. Nitazoxanide can inhibit M. tuberculosis proliferation in vitro. Here we show that it inhibits M. tuberculosis proliferation more potently in infected human THP-1 cells and peripheral monocytes. We identify the human quinone oxidoreductase NQO1 as a nitazoxanide target and propose, based on experiments with cells expressing NQO1 or not, that NQO1 inhibition is partly responsible for mTORC1 inhibition and enhanced autophagy. The dual action of nitazoxanide on both the bacterium and the host cell response to infection may lead to improved tuberculosis treatment.


Subject(s)
Autophagy/drug effects , Macrophages/microbiology , Mycobacterium tuberculosis/growth & development , Proteins/metabolism , Thiazoles/pharmacology , Antiparasitic Agents/pharmacology , Cell Line , HEK293 Cells , Humans , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1 , Monocytes/microbiology , Multiprotein Complexes , Mycobacterium tuberculosis/drug effects , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Nitro Compounds , Phagosomes/metabolism , TOR Serine-Threonine Kinases , Tuberculosis/drug therapy , Tuberculosis/prevention & control
2.
J Biol Chem ; 287(21): 17530-17545, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22474287

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) signaling is frequently dysregulated in cancer. Inhibition of mTORC1 is thus regarded as a promising strategy in the treatment of tumors with elevated mTORC1 activity. We have recently identified niclosamide (a Food and Drug Administration-approved antihelminthic drug) as an inhibitor of mTORC1 signaling. In the present study, we explored possible mechanisms by which niclosamide may inhibit mTORC1 signaling. We tested whether niclosamide interferes with signaling cascades upstream of mTORC1, the catalytic activity of mTOR, or mTORC1 assembly. We found that niclosamide does not impair PI3K/Akt signaling, nor does it inhibit mTORC1 kinase activity. We also found that niclosamide does not interfere with mTORC1 assembly. Previous studies in helminths suggest that niclosamide disrupts pH homeostasis of the parasite. This prompted us to investigate whether niclosamide affects the pH balance of cancer cells. Experiments in both breast cancer cells and cell-free systems demonstrated that niclosamide possesses protonophoric activity in cells and in vitro. In cells, niclosamide dissipated protons (down their concentration gradient) from lysosomes to the cytosol, effectively lowering cytoplasmic pH. Notably, analysis of five niclosamide analogs revealed that the structural features of niclosamide required for protonophoric activity are also essential for mTORC1 inhibition. Furthermore, lowering cytoplasmic pH by means other than niclosamide treatment (e.g. incubation with propionic acid or bicarbonate withdrawal) recapitulated the inhibitory effects of niclosamide on mTORC1 signaling, lending support to a possible role for cytoplasmic pH in the control of mTORC1. Our data illustrate a potential mechanism for chemical inhibition of mTORC1 signaling involving modulation of cytoplasmic pH.


Subject(s)
Antinematodal Agents/pharmacology , Niclosamide/pharmacology , Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Hydrogen-Ion Concentration , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases
3.
J Med Chem ; 53(21): 7843-51, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20945907

ABSTRACT

Desbromoceratamine A (3) exhibits significantly less potent activity than the natural product ceratamine A (1) in a cell-based assay for antimitotic activity. Synthesis of the ceratamine A analogue 4 has shown that replacing the bromine atoms in the natural product with methyl groups generates an analogue that is more active than natural ceratamine A (1). Further enhancement of the antimitotic activity of the ceratamine pharmacophore has been achieved in the synthetic analogue 33, which has both bromine atoms replaced with methyl groups and an additional methyl substituent on the amino nitrogen at C-2. An efficient synthetic route has been developed to 33 that should enable the first in vivo evaluation of the new ceratamine microtubule-stabilizing pharmacophore and has provided several additional analogues for structure-activity relationship evaluation.


Subject(s)
Alkaloids/pharmacology , Azepines/pharmacology , Imidazoles/pharmacology , Tubulin Modulators/pharmacology , Alkaloids/chemistry , Azepines/chemical synthesis , Azepines/chemistry , Biological Products/chemistry , Biological Products/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Mitosis/drug effects , Stereoisomerism , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
4.
Exp Hematol ; 37(11): 1274-83, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19703514

ABSTRACT

OBJECTIVE: Multiple myeloma (MM) is a B-lymphocyte neoplasia that is presently incurable because the tumor cells become resistant to currently available drugs. The growth and survival signals resulting from interactions between the malignant clones and the bone marrow microenvironment are mediated chiefly through the phosphoinositide 3'-kinase/Akt kinase signaling pathway. Thus agents that can abrogate this pathway have great potential as targeted therapies. A novel approach in this regard is through activation of the Src homology 2-containing inositol 5'-phosphatase (SHIP), using the small molecule agonist, AQX-MN100. MATERIALS AND METHODS: The SHIP agonist AQX-MN100 was tested in vitro for its ability to inhibit DNA synthesis, induce apoptosis in MM cell lines, as well as inhibit phosphorylation of the kinases in the phosphoinositide 3'-kinase/Akt kinase cascade. The ability of AQX-MN100 to enhance the cytotoxicity of the current MM therapeutic drugs dexamethasone and bortezomib was also examined. RESULTS: We demonstrate herein that activation of SHIP using AQX-MN100 is sufficient to prevent growth and induce cytotoxicity of MM cell lines, while having no significant effects on nonhematopoietic cells lacking SHIP. AQX-MN100 also augments the effects of the established agents dexamethasone and bortezomib. CONCLUSION: These results provide the basis for the further study of small molecule SHIP activators to improve MM patient outcomes.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Phosphoric Monoester Hydrolases/drug effects , Polycyclic Compounds/pharmacology , Animals , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , DNA Replication/drug effects , Dexamethasone/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Activation/drug effects , Humans , Inositol Phosphates/physiology , Inositol Polyphosphate 5-Phosphatases , Mice , Neoplasm Proteins/agonists , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazines/pharmacology , Signal Transduction/drug effects
5.
J Org Chem ; 74(3): 995-1006, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19128042

ABSTRACT

Two synthetic approaches to the microtubule-stabilizing ceratamine alkaloids are described. The first approach involved attempts to graft an aminoimidazole moiety onto an azepine ring to form partially hydrogenated versions of the unprecedented aromatic imidazo[4,5-d]azepine core of the ceratamines. This route ultimately failed because it was not possible to aromatize the partially hydrogenated ceratamine intermediates. A second approach started with tribromoimidazole that was sequentially metalated and functionalized to efficiently generate a key imidazole intermediate containing vinyl bromide and amide functionalities. An intramolecular Buchwald vinyl amidation reaction converted this key intermediate into a bicyclic imidazo[4,5-d]azepine that was at the same oxidation state as the aromatic core of the ceratamines. The 2-amino functionality present on the imidazole ring of the ceratamines was installed using a Buchwald/Hartwig amination reaction on a 2-chloroimidazole precursor. Deprotection and aromatization resulted in the first synthesis of desbromoceratamine A (55) and desmethyldesbromoceratamine A (60). An unanticipated addition of atmospheric oxygen was encountered during deprotection of the imidazole ring in the last step of the synthesis leading to C-11 oxygenated ceratamine analogues as byproducts. Evaluation of the synthetic ceratamines in a TG3 cell-based assay for mitotic arrest revealed that the C-14 and C-16 bromine substituents in ceratamine A (1) play a major role in the antimitotic potency of the natural product. The synthetic route to ceratamine analogues has provided sufficient quantities of desbromoceratamine A (55) for testing in mouse models of cancer.


Subject(s)
Alkaloids/chemical synthesis , Azepines/chemical synthesis , Imidazoles/chemical synthesis , Animals , Bromobenzenes/chemical synthesis , Porifera/chemistry
6.
Org Lett ; 10(6): 1051-4, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18278926

ABSTRACT

Antimitotic analogs of the microtubule stabilizing sponge alkaloid ceratamine A (1) have been synthesized starting from tribromoimidazole. A key step in the synthesis is the formation of the azepine ring via an intramolecular Buchwald coupling between a vinyl bromide and a N-methyl amide. This represents the first synthesis of a fully unsaturated imidazo[4,5,d]azepine. NMR data obtained for the synthetic ceratamine analogs has provided support for the structure assigned to the natural product.


Subject(s)
Azepines/chemical synthesis , Imidazoles/chemical synthesis , Microtubules/chemistry , Mitosis/drug effects , Animals , Azepines/chemistry , Azepines/pharmacology , Imidazoles/chemistry , Imidazoles/pharmacology , Magnetic Resonance Spectroscopy , Molecular Structure , Porifera
7.
Blood ; 110(6): 1942-9, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17502453

ABSTRACT

Because phosphoinositide 3-kinase (PI3K) plays a central role in cellular activation, proliferation, and survival, pharmacologic inhibitors targeting components of the PI3K pathway are actively being developed as therapeutics for the treatment of inflammatory disorders and cancer. These targeted drugs inhibit the activity of either PI3K itself or downstream protein kinases. However, a previously unexplored, alternate strategy is to activate the negative regulatory phosphatases in this pathway. The SH2-containing inositol-5'-phosphatase SHIP1 is a normal physiologic counter-regulator of PI3K in immune/hematopoietic cells that hydrolyzes the PI3K product phosphatidylinositiol-3,4,5-trisphosphate (PIP(3)). We now describe the identification and characterization of potent and specific small-molecule activators of SHIP1. These compounds represent the first small-molecule activators of a phosphatase, and are able to activate recombinant SHIP1 enzyme in vitro and stimulate SHIP1 activity in intact macrophage and mast cells. Mechanism of activation studies with these compounds suggest that they bind a previously undescribed, allosteric activation domain within SHIP1. Furthermore, in vivo administration of these compounds was protective in mouse models of endotoxemia and acute cutaneous anaphylaxis, suggesting that SHIP1 agonists could be used therapeutically to inhibit the PI3K pathway.


Subject(s)
Anaphylaxis/drug therapy , Endotoxemia/drug therapy , Enzyme Inhibitors/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Polycyclic Compounds/pharmacology , Sesquiterpenes/pharmacology , Allosteric Regulation , Anaphylaxis/metabolism , Anaphylaxis/pathology , Animals , Calcium/metabolism , Cells, Cultured , Endotoxemia/metabolism , Endotoxemia/pathology , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic , Humans , Immunoprecipitation , Inositol Polyphosphate 5-Phosphatases , Kidney/cytology , Kidney/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation/drug effects , Polycyclic Compounds/chemistry , Porifera/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sesquiterpenes/chemistry , Signal Transduction/drug effects , Skin Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...