Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
J Enzyme Inhib Med Chem ; 37(1): 652-665, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35109719

ABSTRACT

The screened compound DYT-1 from our in-house library was taken as a lead (inhibiting tubulin polymerisation: IC50=25.6 µM, anti-angiogenesis in Zebrafish: IC50=38.4 µM, anti-proliferation against K562 and Jurkat: IC50=6.2 and 7.9 µM, respectively). Further investigation of medicinal chemistry conditions yielded compound 29e (inhibiting tubulin polymerisation: IC50=4.8 µM and anti-angiogenesis in Zebrafish: IC50=3.6 µM) based on tubulin and zebrafish assays, which displayed noteworthily nanomolar potency against a variety of leukaemia cell lines (IC50= 0.09-1.22 µM), especially K562 cells where apoptosis was induced. Molecular docking, molecular dynamics (MD) simulation, radioligand binding assay and cellular microtubule networks disruption results showed that 29e stably binds to the tubulin colchicine site. 29e significantly inhibited HUVEC tube formation, migration and invasion in vitro. Anti-angiogenesis in vivo was confirmed by zebrafish xenograft. 29e also prominently blocked K562 cell proliferation and metastasis in blood vessels and surrounding tissues of the zebrafish xenograft model. Together with promising physicochemical property and metabolic stability, 29e could be considered an effective anti-angiogenesis and -leukaemia drug candidate that binds to the tubulin colchicine site.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Indoles/pharmacology , Neovascularization, Pathologic/drug therapy , Tubulin Modulators/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Zebrafish
2.
J Enzyme Inhib Med Chem ; 37(1): 339-354, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34979843

ABSTRACT

α-Fluorinated chalcones were prepared and evaluated for their cell growth inhibitory properties against six human cancer cell lines. The most potent chalcone 4c demonstrated excellent selective toxicity against cancer cells versus normal human cells, with IC50 values at nanomolar concentration ranges against 5 cancer cell lines. A further study revealed that 4c could bind to the colchicine site of tubulin, disrupt the cell microtubule networks, and effectively inhibit tubulin polymerisation. Cellular-based mechanism studies elucidated that 4c arrested MGC-803 cell cycle at G2/M phase. In addition, 4c dose-dependently caused Caspase-induced apoptosis of MGC-803 cells through mitochondrial dysfunction. Notably, compound 4c was found to inhibit the HUVECs tube formation, migration, and invasion in vitro. Furthermore, our data suggested that treatment with 4c significantly reduced MGC-803 cells metastasis and proliferation in vitro. Overall, this work showed that chalcone hybrid 4c is a potent inhibitor of tubulin assembly with prominent anti-angiogenesis and anti-cancer properties.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Colchicine/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Tubulin Modulators/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemical synthesis , Chalcones/chemistry , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Halogenation , Humans , Molecular Structure , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Polymerization/drug effects , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
3.
Future Med Chem ; 13(19): 1623-1638, 2021 10.
Article in English | MEDLINE | ID: mdl-34505541

ABSTRACT

Background: Although some benzimidazole-based anthelmintic drugs are found to possess anticancer activity, their modes of binding interactions have not been reported. Methodology: In this study, we aimed to investigate the binding interactions and electronic configurations of nine benzimidazole-based anthelmintics against one of the well-known cancer targets (tubulin protein). Results: Binding affinities of docked benzimidazole drugs into colchicine-binding site were calculated where flubendazole > oxfendazole > nocodazole > mebendazole. Flubendazole was found to bind more efficiently with tubulin protein than other drugs. Quantum mechanics studies revealed that the electron density of HOMO of flubendazole and mebendazole together with their molecular electrostatic potential map are closely similar to that of nocodazole. Conclusion: Our study has ramifications for considering repurposing of flubendazole as a promising anticancer candidate.


Subject(s)
Anthelmintics/pharmacology , Benzimidazoles/pharmacology , Colchicine/antagonists & inhibitors , Density Functional Theory , Anthelmintics/chemistry , Benzimidazoles/chemistry , Binding Sites/drug effects , Colchicine/metabolism , Drug Repositioning , Molecular Docking Simulation , Molecular Structure
4.
J Enzyme Inhib Med Chem ; 36(1): 2025-2044, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34514909

ABSTRACT

Searching for improved indolesulfonamides with higher polarities, 45 new analogues with modifications on the sulfonamide nitrogen, the methoxyaniline, and/or the indole 3-position were synthesised. They show submicromolar to nanomolar antiproliferative IC50 values against four human tumour cell lines and they are not P-glycoprotein substrates as their potencies against HeLa cells did not improve upon cotreatment with multidrug resistance (MDR) inhibitors. The compounds inhibit tubulin polymerisation in vitro and in cells, thus causing a mitotic arrest followed by apoptosis as shown by cell cycle distribution studies. Molecular modelling studies indicate binding at the colchicine site. Methylated sulfonamides were more potent than those with large and polar substitutions. Amide, formyl, or nitrile groups at the indole 3-position provided drug-like properties for reduced toxicity, with Polar Surface Areas (PSA) above a desirable 75 Å2. Nitriles 15 and 16 are potent polar analogues and represent an interesting class of new antimitotics.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Sulfonamides/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Colchicine/chemistry , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Models, Molecular , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Tubulin/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tumor Cells, Cultured
5.
Bioorg Chem ; 112: 104965, 2021 07.
Article in English | MEDLINE | ID: mdl-34020238

ABSTRACT

Thirty-five new colchicine binding site inhibitors have been designed and synthesized based on the 1,2,4-triazin-3(2H)-one nucleus. Such molecules were synthesized through a cascade reaction between readily accessible α-amino ketones and phenyl carbazate as a masked N-isocyanate precursor. The synthesized derivatives are cisoid restricted combretastatin A4 analogues containing 1,2,4-triazin-3(2H)-one in place of the olefinic bond, and they have the same essential pharmacophoric features of colchicine binding site inhibitors. The synthesized compounds were evaluated in vitro for their antiproliferative activities against a panel of three human cancer cell lines (MCF-7, HepG-2, and HCT-116), using colchicine as a positive control. Among them, two compounds 5i and 6i demonstrated a significant antiproliferative effect against all cell lines with IC50 ranging from 8.2 - 18.2 µM. Further investigation was carried out for the most active cytotoxic agents as tubulin polymerization inhibitors. Compounds 5i and 6i effectively inhibited microtubule assembly with IC50 values ranging from 3.9 to 7.8 µM. Tubulin polymerization assay results were found to be comparable with the cytotoxicity results. The cell cycle analysis revealed significant G2/M cell cycle arrest of the analogue 5i in HepG-2 cells. The most active compounds 4i, 4j, 5 g, 5i and 6i did not induce significant cell death in normal human lung cells Wl-38, suggesting their selectivity against cancer cells. Also, These compounds upregulated the level of active caspase-3 and boosted the levels of the pro-apoptotic protein Bax by five to seven folds in comparison to the control. Moreover, apoptosis analyses were conducted for compound 5i to evaluate its apoptotic potential. Finally, in silico studies were conducted to reveal the probable interaction with the colchicine binding site. ADME prediction study of the designed compounds showed that they are not only with promising tubulin polymerization inhibitory activity but also with favorable pharmacokinetic and drug-likeness properties.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Drug Design , Triazines/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Triazines/chemical synthesis , Triazines/chemistry , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
6.
Eur J Med Chem ; 214: 113229, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33550186

ABSTRACT

Three different series of cis-restricted analogues of combretastatin A-4 (CA-4), corresponding to thirty-nine molecules that contained a pyrrole nucleus interposed between the two aryl rings, were prepared by a palladium-mediated coupling approach and evaluated for their antiproliferative activity against six human cancer cell lines. In the two series of 1,2-diaryl pyrrole derivatives, results suggested that the presence of the 3',4',5'-trimethoxyphenyl moiety at the N-1 position of the pyrrole ring was more favorable for antiproliferative activity. In the series of 3,4-diarylpyrrole analogues, three compounds (11i-k) exhibited maximal antiproliferative activity, showing excellent antiproliferative activity against the CA-4 resistant HT-29 cells. Inhibition of tubulin polymerization of selected 1,2 pyrrole derivatives (9a, 9c, 9o and 10a) was similar to that observed with CA-4, while the isomeric 3,4-pyrrole analogues 11i-k were generally from 1.5- to 2-fold more active than CA-4. Compounds 11j and 11k were the only compounds that showed activity as inhibitors of colchicine binding comparable to that CA-4. Compound 11j had biological properties consistent with its intracellular target being tubulin. This compound was able to block the cell cycle in metaphase and to induce significant apoptosis at a concentration of 25 nM, following the mitochondrial pathway, with low toxicity for normal cells. More importantly, compound 11j exerted activity in vivo superior to that of CA-4P, being able to significantly reduce tumor growth in a syngeneic murine tumor model even at the lower dose tested (5.0 mg/kg).


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Drug Discovery , Pyrroles/pharmacology , Tubulin Modulators/pharmacology , Antimitotic Agents/chemical synthesis , Antimitotic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Polymerization/drug effects , Pyrroles/chemical synthesis , Pyrroles/chemistry , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
7.
Curr Top Med Chem ; 20(17): 1505-1508, 2020.
Article in English | MEDLINE | ID: mdl-32543362

ABSTRACT

Butterfly-shaped structure, as a novel scaffold with an attractive and certain shape, has been widely used in new drug discovery. Tubulin, composing of α- and ß-tubulin heterodimers, plays a key role in mitosis and cell division which are regarded as an excellent target for cancer therapy. Currently, a series of butterfly shape diaryl heterocyclic compounds have been reported with strong potential against the tubulin-colchicine binding site. It is with one ring buried in the ß subunit, another ring interacts with the α subunit and the main body is located in the flat pocket. Here, we firstly introduce the concept of butterfly structure for the tubulin inhibitors, focusing on the latest advancements in a variety of molecules bearing butterfly structure, and then highlight the challenges and future direction of butterfly structure- based tubulin-colchicine binding site inhibitors.


Subject(s)
Colchicine/antagonists & inhibitors , Heterocyclic Compounds/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Binding Sites/drug effects , Colchicine/chemistry , Colchicine/metabolism , Heterocyclic Compounds/chemistry , Humans , Molecular Structure , Tubulin/chemistry , Tubulin Modulators/chemistry
8.
Bioorg Chem ; 99: 103831, 2020 06.
Article in English | MEDLINE | ID: mdl-32388203

ABSTRACT

New series of 2-amino-1,4,5,6,7,8-hexahydroquinoline-3-carbonitriles 3a,b and 2-amino-5,6,7,8-tetrahydronaphthalene-1,3-dicarbonitriles 4a-h were synthesized and evaluated for their antitumor activity. In vitro antitumor screening of the new members against HepG2, HCT-116 and MCF-7 cancer cells showed that the tetrahydronaphthalene-1,3-dicarbonitrile 4c has the highest potency against the three tested cancer cells (IC50 = 6.02, 8.45 and 6.28 µM, respectively). In addition, 4c displayed low cytotoxicity against WI38 and WISH normal cells (IC50 = 51.78 and 42.36 µM, respectively), and it might be utilized as a potent and selective antitumor agent. Compound 4c was further studied for its effect on tubulin polymerization, different phases of cell cycle, apoptosis and caspases 3/9 levels. Results revealed that analog 4c has good tubulin polymerization inhibitory activity (IC50 = 3.64 µM). Additionally, it induced significant accumulation of the tested cancer cells in G2/M phase, and induced cell death primarily via apoptosis. Besides, it showed evident increase in caspase-3 level in HepG2 and HCT-116 cells, and caspase-9 level in MCF-7 cells. Further, docking studies proved the exact fit of 4c into the colchicine binding site of tubulin.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Hydroquinones/pharmacology , Naphthyridines/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydroquinones/chemical synthesis , Hydroquinones/chemistry , Molecular Docking Simulation , Molecular Structure , Naphthyridines/chemical synthesis , Naphthyridines/chemistry , Polymerization/drug effects , Structure-Activity Relationship
9.
Eur J Med Chem ; 196: 112328, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32320841

ABSTRACT

Small molecules targeting the colchicine site of tubulin represent an attractive cancer treatment strategy. In this study, a total of 468 models derived from 1076 diverse inhibitors binding to the tubulin colchicine site were constructed based on fingerprints using three machine learning approaches: 1) naive Bayesian (NB); 2) single tree (ST); and 3) random forest (RF). The overall predictive accuracy of the best models exceeded 85.12% for both the training and test sets. We designed an integrated virtual screening (VS) strategy for identifying new tubulin inhibitors by combining established models, molecular docking, and similarity-based analog searching. Through two rounds of VS, compound 23g was identified as a novel potent anticancer agent exhibiting activity against MDA-MB-231, HeLa, A549, HepG2, CNE2, and HCT116 tumor cell lines with IC50 values of 5.45, 8.61, 7.47, 2.29, 2.91, and 4.10 µM, respectively. Compared with taxol, colchicine, and adriamycin, 23g also displayed potent cytotoxicity against the drug-resistant tumor cell lines, HepG2/ADR, A549/CDDP, and A549/TAX cells, with IC50 values of 4.12, 6.58, and 6.38 µM, respectively. Further mechanistic studies revealed that 23g inhibited microtubule polymerization by binding to the colchicine site of tubulin, arrested the cell cycle at the G2/M phase, induced cell apoptosis, and exhibited potent in vitro anti-metastasis activity. Finally, molecular docking, molecular dynamics, and free energy analyses were employed to explore the detailed binding interaction between 23g and tubulin. Collectively, these findings indicated that 23g should be further investigated as a potential novel potent antitumor agent targeting tubulin.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Drug Discovery , Small Molecule Libraries/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Colchicine/metabolism , Drug Screening Assays, Antitumor , Humans , Ligands , Molecular Dynamics Simulation , Molecular Structure , Polymerization/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship
10.
J Enzyme Inhib Med Chem ; 35(1): 139-144, 2020 Dec.
Article in English | MEDLINE | ID: mdl-31724435

ABSTRACT

A series of naphthalene-chalcone derivatives (3a-3t) were prepared and evaluated as tubulin polymerisation inhibitor for the treatment of breast cancer. All compounds were evaluated for their antiproliferative activity against MCF-7 cell line. The most of compounds displayed potent antiproliferative activity. Among them, compound 3a displayed the most potent antiproliferative activity with an IC50 value of 1.42 ± 0.15 µM, as compared to cisplatin (IC50 = 15.24 ± 1.27 µM). Additionally, the promising compound 3a demonstrated relatively lower cytotoxicity on normal cell line (HEK293) compared to tumour cell line. Furthermore, compound 3a was found to induce significant cell cycle arrest at the G2/M phase and cell apoptosis. Compound 3a displayed potent tubulin polymerisation inhibitory activity with an IC50 value of 8.4 µM, which was slightly more active than the reference compound colchicine (IC50 = 10.6 µM). Molecular docking analysis suggested that 3a interact and bind at the colchicine binding site of the tubulin.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chalcones/pharmacology , Naphthalenes/pharmacology , Tubulin Modulators/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Binding Sites/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chalcones/chemistry , Colchicine/antagonists & inhibitors , Colchicine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure , Naphthalenes/chemistry , Structure-Activity Relationship , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
11.
Bioorg Med Chem Lett ; 29(18): 2632-2634, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31362922

ABSTRACT

The colchicine site inhibitors (CSIs) displayed both antimitotic and vascular disrupting activities, therefore are promising potential antitumor agents. In this study, a series 1-phenyl-4,5-dihydro-2H-benzo[e]indazoles were found as new CSIs of which the bioactive configuration was locked. Among them, compounds C1 and C2 displayed the best activity, with tubulin polymerization IC50 of 3.4 and 1.5 µM, and growth IC50 of low nanomolar concentrations against human colon cancer cell lines. In addition, compound C1 showed excellent broad-spectrum antitumor activity in the NCI-60 Human Tumor Cell Lines Screen, encouraging further study of this antitumor compound.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship
12.
Eur J Med Chem ; 146: 185-193, 2018 Feb 25.
Article in English | MEDLINE | ID: mdl-29407949

ABSTRACT

A series of novel 4,6-diphenyl-2-(1H-pyrrol-1-yl)nicotinonitrile analogues of crolibulin and combretastatin A-4 (CA-4) were discovered using a 2-(1H-pyrrol-1-yl)pyridine ring as link-bridge to retain the cis-orientations of A-ring and B-ring. All the target compounds were synthesized and evaluated for their antiproliferative activity against five human cancer cell lines. Compounds 6a-d exhibited superior potency, with IC50 values at nanomolar levels. In particular, compound 6a exhibited antitumor activity similar to or higher than crolibulin and CA-4. Moreover, the inhibition of microtubule assembly by compound 6a was comparable to that by CA-4. A molecular modeling study of compound 6a was performed to elucidate its binding mode at the colchicine binding site in the tubulin dimer, which also provided a basis for further structure-guided design of novel colchicine binding site inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Niacin/pharmacology , Nitriles/pharmacology , Stilbenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzopyrans/chemistry , Binding Sites/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Microtubules/drug effects , Models, Molecular , Molecular Structure , Niacin/analogs & derivatives , Niacin/chemical synthesis , Niacin/chemistry , Nitriles/chemical synthesis , Nitriles/chemistry , Stilbenes/chemistry , Structure-Activity Relationship
13.
Tumour Biol ; 37(8): 10653-64, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26867767

ABSTRACT

Colchicine is a well-known and potent microtubule targeting agent, but the therapeutic value of colchicine against cancer is limited by its toxicity against normal cells. But, there is no report of its cytotoxic potential against lung cancer cell, at clinically permissible or lower concentrations, minimally toxic to non-cancerous cells. Hence, in the present study, we investigated the possible mechanism by which the efficacy of colchicine against lung cancer cells at less toxic dose could be enhanced. Colchicine at clinically admissible concentration of 2.5 nM had no cytotoxic effect and caused no G2/M arrest in A549 cells. However, at this concentration, colchicine strongly hindered the reformation of cold depolymerised interphase and spindle microtubule. Colchicine induced senescence and reactive oxygen species mediated autophagy in A549 cells at this concentration. Autophagy inhibitor 3-methyladenine (3-MA) sensitised the cytotoxicity of colchicine in A549 cells by switching senescence to apoptotic death, and this combination had reduced cytotoxicity to normal lung fibroblast cells (WI38). Together, these findings indicated the possible use of colchicine at clinically relevant dose along with autophagy inhibitor in cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Cellular Senescence/drug effects , Colchicine/pharmacology , Lung Neoplasms/pathology , A549 Cells , Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Colchicine/administration & dosage , Colchicine/antagonists & inhibitors , Dose-Response Relationship, Drug , Fibroblasts , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubules/drug effects , Reactive Oxygen Species/metabolism , Spindle Apparatus/drug effects , Tubulin Modulators/administration & dosage , Tubulin Modulators/antagonists & inhibitors , Tubulin Modulators/pharmacology
14.
Eur J Med Chem ; 77: 306-14, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24657567

ABSTRACT

2-Hydroxy-3,4,5-trimethoxybenzophenones (8-16) manifest pseudo-ring formation involving intramolecular hydrogen bonding of the 2-OH and the carbonyl group. Among the synthetic products described in this report, (3-hydroxy-4-methoxyphenyl)(2-hydroxy-3,4,5-trimethoxyphenyl)-methanone (14) and (3-amino-4-methoxyphenyl)(2-hydroxy-3,4,5-trimethoxy-phenyl)methanone (16) exhibit significant antiproliferative activity against KB cells with IC50 values of 11.1 and 11.3 nM, respectively. These two compounds also displayed tubulin affinity comparable to that of combretastatin A-4. In studies with human umbilical vein endothelial cells, compounds 14 and 16 revealed concentration-dependent vascular-disrupting properties. The results support the rationale of the pseudo-ring concept and suggest further investigation of A-ring modification in these benzophenones.


Subject(s)
Antimitotic Agents/pharmacology , Benzophenones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Antimitotic Agents/chemical synthesis , Antimitotic Agents/chemistry , Benzophenones/chemical synthesis , Benzophenones/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/antagonists & inhibitors , Dose-Response Relationship, Drug , HT29 Cells , Humans , KB Cells , Molecular Structure , Polymerization/drug effects , Structure-Activity Relationship , Tubulin/metabolism
15.
Int J Neurosci ; 122(6): 324-32, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22248034

ABSTRACT

Although nicotine is known to protect against ß-amyloid (Aß)-induced neurotoxicity, the effect of nicotine on colchicine-induced neurotoxicity remains unknown. Colchicine is a microtubule-interfering agent and is able to induce neural apoptosis. Here we investigated whether nicotine exhibits similar neuroprotective effects and the mechanism against colchicine-induced neurotoxicity of the primarily cultured cortical neurons. In this study, we investigated the effect of nicotine on the protection of neurons against colchicine damage and evaluated the associated intracellular signaling pathways. Nicotine-induced protection was blocked by an α7 nicotinic acetylcholine receptors (nAChRs) antagonist and a phosphatidylinositol 3-kinase (PI3K) inhibitor. These results suggest that the neuroprotective effects of nicotine are mediated by the α7 nAChRs and PI3K-Akt signaling pathway. In addition, we reveal that blockade of p38 and JNK (c-Jun N-terminal kinase) signaling increased Akt signaling, thus enhancing the survival of cell treatment with colchicine. On the other hand, inhibition of constitutively active Akt enhanced p38 or JNK signaling phosphorylation. These data suggested that crosstalk between PI3K Akt and p38 or JNK signaling pathways contributed to nicotine against colchicine-induced cytotoxicity.


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/drug effects , Colchicine/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Nicotine/pharmacology , Phosphatidylinositol 3-Kinase/physiology , Proto-Oncogene Proteins c-akt/drug effects , Animals , Animals, Newborn , Apoptosis/physiology , Bungarotoxins/pharmacology , Cell Survival/drug effects , Colchicine/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neurons/drug effects , Neuroprotective Agents/antagonists & inhibitors , Nicotine/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/physiology , Signal Transduction/drug effects , alpha7 Nicotinic Acetylcholine Receptor , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Chin J Integr Med ; 17(2): 126-33, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21390579

ABSTRACT

OBJECTIVE: To study the effect of anticolchicine cytotoxicity of Dan Gua-Fang, a Chinesea Chinese), a Chinese herbal compound prescription on endothelial cells of vein (ECV304) cultivated in mediums of different glucose concentrations as well as the proliferation of those cells in the same conditions, in order to reveal the value of Dan Gua-Fang in preventing and treating endothelial damage caused by hyperglycemia in diabetes mellitus. METHODS: The research was designed as three stages. The growing state and morphological changes were observed when ECV304 were cultivated in the culture mediums, which have different glucose concentrations with or without Dan Gua-Fang and at the same time with or without colchicine. RESULTS: (1) Dan Gua-Fang at all concentrations reduced the floating cell population of ECV304 cultivated in hyperglycemia mediums. (2) Dan Gua-Fang at all concentrations and hyperglycemia both had a function of promoting "pseudopod-like" structure formation in cultivated ECV304, but the function was not superimposed in mediums containing both hyperglycemia and Dan Gua-Fang. (3) Colchicine reduced and even vanished the "pseudopod-like" structure of the endotheliocyte apparently cultivated in mediums of hyperglycemia or with Dan Gua-Fang. The "pseudopod-like" structure of the endotheliocyte emerged quickly in Dan Gua-Fang groups after colchicine was removed, but it was not the case in hyperglycemia only without Dan Gua-Fang groups. (4) Dan Gua-Fang reduced the mortality of cells cultivated in mediums containing colchicine. The cell revived to its normal state fast after colchicine was removed. CONCLUSION: Dan Gua-Fang has the functions of promoting the formation of cytoskeleton and fighting against colchicine cytotoxicity.


Subject(s)
Colchicine/adverse effects , Colchicine/antagonists & inhibitors , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/drug effects , Cell Culture Techniques , Cell Line , Cell Shape/drug effects , Culture Media/adverse effects , Culture Media/pharmacology , Cytoprotection/drug effects , Cytotoxins/adverse effects , Cytotoxins/antagonists & inhibitors , Drug Antagonism , Drug Combinations , Drug Evaluation, Preclinical , Drug Synergism , Drugs, Chinese Herbal/adverse effects , Endothelial Cells/physiology , Glucose/pharmacology , Humans , Umbilical Veins/cytology , Umbilical Veins/drug effects , Up-Regulation
17.
Neurochem Int ; 58(1): 85-91, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21073911

ABSTRACT

There are few articles about the cytotoxicity evoked by secalonic acid A (SAA) in some tumor cells. It has not yet been reported whether SAA has any action on neurons of the central nervous system. The aim of this study was to investigate the protective effect of SAA against apoptosis of rat cortical neurons induced by colchicine. The protective action of SAA on the cortical neurons treated with colchicine at 1 µM was examined by Hoechst 33258, LDH release and flow cytometry methods. The results from the above tests indicated that SAA at 3 and 10 µM significantly prevented colchicine-induced apoptosis of the cortical neurons. Further studies from Western blot and confocal microscopy experiments showed that the activation of JNK, p38 MAPKs and caspase-3 during neuron apoptosis triggered by 1 µM colchicine could be obviously suppressed by SAA; on the other hand, an increase in the intracellular free Ca(2+) by 1 µM colchicine in the cortical neuron was blocked evidently by SAA. The above results suggested that SAA could antagonize the cytotoxicity of colchicine in the rat cortical neurons, which may be through inhibition of phosphorylation of JNK and p38 MAPKs, calcium influx, and the activation of caspase-3.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Cell Survival/drug effects , Colchicine/antagonists & inhibitors , Colchicine/toxicity , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mycotoxins/pharmacology , Xanthones/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Blotting, Western , Caspase Inhibitors , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Flow Cytometry , Indicators and Reagents , Microscopy, Confocal , Neurons/drug effects , Paecilomyces/chemistry , Rats , Rats, Sprague-Dawley
18.
J Enzyme Inhib Med Chem ; 24(6): 1237-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19912057

ABSTRACT

The crucial role of the microtubule in cell division has identified tubulin as a target for the development of therapeutics for cancer; in particular, tubulin is a target for antineoplastic agents that act by interfering with the dynamic stability of microtubules. A molecular modeling study was carried out to accurately represent the complex structure and the binding mode of a new class of stilbene-based tubulin inhibitors that bind at the alphabeta-tubulin colchicine site. Computational docking along with HINT (Hydropathic INTeractions) score analysis fitted these inhibitors into the colchicine site and revealed detailed structure-activity information useful for inhibitor design. Quantitative analysis of the results was in good agreement with the in vitro antiproliferative activity of these derivatives (ranging from 3 nM to 100 muM) such that calculated and measured free energies of binding correlate with an r(2) of 0.89 (standard error +/- 0.85 kcal mol(-1)). This correlation suggests that the activity of unknown compounds may be predicted.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/metabolism , Leukemia/drug therapy , Microtubules/drug effects , Stilbenes/chemistry , Stilbenes/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemistry , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/antagonists & inhibitors , Computer Simulation , Drug Evaluation, Preclinical , Humans , Leukemia/metabolism , Leukemia/pathology , Microtubules/metabolism , Structure-Activity Relationship , Thermodynamics , Tubulin Modulators/chemistry
19.
Pharmacol Biochem Behav ; 92(1): 25-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18992766

ABSTRACT

Cognitive impairment and weak intellectual capacity is a gradually progressive neurodegenerative problem. Growing evidences indicate that oxidants and antioxidant defenses interact in a vicious cycle, which plays a critical role in the pathogenesis of cognitive dysfunction. The present study was carried out to elucidate the neuroprotective effect of carvedilol against the colchicine-induced cognitive impairment and oxidative damage in rats. Colchicine (15 microg/5 microl), a microtubule disrupting agent when administered intracerebroventricularly in rats resulted in poor memory retention in both Morris water maze, elevated plus maze task paradigms and caused marked oxidative stress as indicated by significant increase in malondialdehyde, nitrite levels, depletion of SOD, catalase, glutathione-S-transferase activity and reduced glutathione levels. It also caused a significant decrease in the acetylcholinesterase activity. Chronic administration of carvedilol (2.5 and 5.0 mg/kg; p.o.) for a period of 25 days, starting 4 days prior to colchicine administration resulted in an improvement in memory retention, attenuation of oxidative damage and restoration of acetylcholinesterase activity. Present study demonstrates a neuroprotective effect of carvedilol against colchicine-induced cognitive impairment and associated oxidative damage.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Antioxidants/pharmacology , Carbazoles/pharmacology , Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Colchicine/antagonists & inhibitors , Colchicine/toxicity , Neuroprotective Agents , Oxidative Stress/physiology , Propanolamines/pharmacology , Acetylcholinesterase/metabolism , Animals , Behavior, Animal/drug effects , Carvedilol , Catalase/metabolism , Cognition Disorders/psychology , Glutathione/metabolism , Glutathione Transferase/metabolism , Injections, Intraventricular , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Nitrites/metabolism , Rats , Rats, Wistar , Space Perception/drug effects , Superoxide Dismutase/metabolism
20.
Brain Res ; 1117(1): 80-91, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-17010319

ABSTRACT

In this work, we have immunohistochemically analyzed the effects of single injections of apotransferrin (aTf) on the expression of myelin (myelin basic proteins [MBPs]) and axonal (protein gene product 9.5 [PGP 9.5] and beta(III)-tubulin [beta(III)-tub]) proteins in colchicine-injected and crushed sciatic nerves of adult rats. A protein redistribution was seen in the distal stump of injured nerves, with the appearance of MBP- and PGP 9.5-immunoreactive (IR) clusters which occurred earlier in crushed nerves (3 days post-injury [PI]) as compared to colchicine-injected nerves (7 days PI). beta(III)-tub-IR clusters appeared at 1 day PI preceding the PGP 9.5- and MBP-IR clusters in colchicine-injected nerves. With image analysis, the peak of clustering formation was found at 14 days PI for MBP and at 3 days PI for beta(III)-tub in colchicine-injected nerves. At 28 days of survival, the protein distribution patterns were almost normal. The intraneural application of aTf, at different concentrations (0.0005 mg/ml, 0.005 mg/ml, 0.05 mg/ml, 0.5 mg/ml), prevented nerve degeneration produced by colchicine, with the appearance of only a small number of MBP- and beta(III)-tub-IR clusters. However, aTf was not able to prevent clustering formation when the nerve was crushed, a kind of injury that also involves necrosis and blood flow alterations. The results suggest that aTf could prevent the colchicine effects by stabilizing the cytoskeleton proteins of the nerve fibers, avoiding the disruption of the axonal transport and thus the myelin degeneration. Transferrin is proposed as a complementary therapeutic avenue for treatment of cytotoxic nerve injuries.


Subject(s)
Apoproteins/pharmacology , Axons/drug effects , Nerve Tissue Proteins/drug effects , Sciatic Neuropathy/drug therapy , Transferrin/pharmacology , Wallerian Degeneration/drug therapy , Wallerian Degeneration/prevention & control , Animals , Axonal Transport/drug effects , Axonal Transport/physiology , Axons/metabolism , Colchicine/antagonists & inhibitors , Colchicine/toxicity , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Immunohistochemistry , Microtubules/drug effects , Microtubules/metabolism , Microtubules/pathology , Myelin Basic Protein/drug effects , Myelin Basic Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Rats , Rats, Wistar , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Treatment Outcome , Tubulin/drug effects , Tubulin/metabolism , Ubiquitin Thiolesterase/drug effects , Ubiquitin Thiolesterase/metabolism , Wallerian Degeneration/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...