Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38700005

RESUMO

BACKGROUND: The only clinically approved drug that reduces doxorubicin cardiotoxicity is dexrazoxane, but its application is limited due to the risk of secondary malignancies. So, exploring alternative effective molecules to attenuate its cardiotoxicity is crucial. Colchicine is a safe and well-tolerated drug that helps reduce the production of reactive oxygen species. High doses of colchicine have been reported to block the fusion of autophagosomes and lysosomes in cancer cells. However, the impact of colchicine on the autophagy activity within cardiomyocytes remains inadequately elucidated. Recent studies have highlighted the beneficial effects of colchicine on patients with pericarditis, postprocedural atrial fibrillation, and coronary artery disease. It remains ambiguous how colchicine regulates autophagic flux in doxorubicin-induced heart failure. METHODS AND RESULTS: Doxorubicin was administered to establish models of heart failure both in vivo and in vitro. Prior studies have reported that doxorubicin impeded the breakdown of autophagic vacuoles, resulting in damaged mitochondria and the accumulation of reactive oxygen species. Following the administration of a low dose of colchicine (0.1 mg/kg, daily), significant improvements were observed in heart function (left ventricular ejection fraction: doxorubicin group versus treatment group=43.75%±3.614% versus 57.07%±2.968%, P=0.0373). In terms of mechanism, a low dose of colchicine facilitated the degradation of autolysosomes, thereby mitigating doxorubicin-induced cardiotoxicity. CONCLUSIONS: Our research has shown that a low dose of colchicine is pivotal in restoring the autophagy activity, thereby attenuating the cardiotoxicity induced by doxorubicin. Consequently, colchicine emerges as a promising therapeutic candidate to improve doxorubicin cardiotoxicity.


Assuntos
Autofagia , Cardiotoxicidade , Colchicina , Doxorrubicina , Lisossomos , Miócitos Cardíacos , Colchicina/toxicidade , Colchicina/farmacologia , Doxorrubicina/toxicidade , Cardiotoxicidade/prevenção & controle , Autofagia/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Modelos Animais de Doenças , Masculino , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Antibióticos Antineoplásicos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Função Ventricular Esquerda/efeitos dos fármacos
2.
J Microsc ; 291(3): 229-236, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37358710

RESUMO

Many diseases are related to changes in the biomechanical properties of cells; their study can provide a theoretical basis for drug screening and can explain the internal working of living cells. In this study, the biomechanical properties of nephrocytes (VERO cells), hepatocytes (HL-7702 cells), and hepatoma cells (SMCC-7721 cells) in culture were detected by atomic force microscopy (AFM) to analyse the side effects of colchicine at different concentrations (0.1 µg/mL (A) and 0.2 µg/mL (B)) at the nanoscale for 2, 4 and 6 h. Compared with the corresponding control cells, the damage to the treated cells increased in a dose-dependent manner. Among normal cells, the injury of nephrocytes (VERO cells) was markedly worse than that of hepatocytes (HL-7702 cells) in both colchicine solutions A and B. Based on the analyses of biomechanical properties, the colchicine solution reduced the rate of division and inhibited metastasis of SMCC-7721 cells. By comparing these two concentrations, we found that the anticancer effect of colchicine solution A was greater than that of solution B. Studying the mechanical properties of biological cells can help understand the mechanism of drug action at the molecular level and provide a theoretical basis for preventing the emergence and diagnosis of diseases at the nanoscale.


Assuntos
Colchicina , Hepatócitos , Animais , Chlorocebus aethiops , Colchicina/toxicidade , Fenômenos Biomecânicos , Células Vero , Microscopia de Força Atômica
3.
ACS Chem Neurosci ; 14(1): 19-34, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36541944

RESUMO

Indibulin, a microtubule-depolymerizing agent, produces minimal neurotoxicity in animals. It is also less cytotoxic toward differentiated neuronal cells than undifferentiated cells. We found that the levels of ß-III tubulin, acetylated tubulin, and polyglutamylated tubulin were significantly increased in differentiated neuroblastoma cells (SH-SY5Y). Since neuronal cells express ß-tubulin isotypes differently from other cell types, we explored the binding of indibulin to different ß-tubulin isotypes. Our molecular docking analysis suggested that indibulin binds to ß-III tubulin with lower affinity than to other ß-tubulin isotypes. We therefore studied the implications of different ß-tubulin isotypes on the cytotoxic effects of indibulin, colchicine, and vinblastine in differentiated SH-SY5Y cells. Upon depletion of ß-III tubulin in the differentiated cells, the toxicity of indibulin and colchicine significantly increased, while sensitivity to vinblastine was unaffected. Using biochemical, bioinformatics, and fluorescence spectroscopic techniques, we have identified the binding site of indibulin on tubulin, which had not previously been established. Indibulin inhibited the binding of colchicine and C12 (a colchicine-site binder) to tubulin and also increased the dissociation constant of the interaction between tubulin and colchicine. Indibulin did not inhibit the binding of vinblastine or taxol to tubulin. Interestingly, indibulin antagonized colchicine treatment but synergized with vinblastine treatment in a combination study performed in MDA-MB-231 cells. The results indicate that indibulin is a colchicine-site binder and that the efficacy of colchicine-site binders is affected by the ß-III tubulin levels in the cells.


Assuntos
Antineoplásicos , Neuroblastoma , Animais , Humanos , Tubulina (Proteína)/metabolismo , Vimblastina/toxicidade , Simulação de Acoplamento Molecular , Antineoplásicos/farmacologia , Colchicina/toxicidade , Colchicina/química , Sítios de Ligação , Moduladores de Tubulina/farmacologia
4.
Fundam Clin Pharmacol ; 37(2): 275-286, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36203311

RESUMO

Alzheimer's disease (AD) is a complex neurodegenerative disease. There is epidemiological evidence that heart failure (HF) patients are at higher risk of developing AD, and the impact of sacubitril/valsartan, the first angiotensin receptor-neprilysin inhibitor (ARNI) approved for HF, on cognitive functions is still controversial. To investigate the effect of sacubitril/valsartan on cognitive functions in colchicine-induced AD rat model. Forty adult male Wistar rats were equally allocated into four groups (each of 10 rats): Group I: normal control, Group II: intracerebroventricular injection of colchicine (15 µg/5 µl/bilaterally), Group III: colchicine (15 µg/5 µl/bilaterally, icv) + oral sacubitril/valsartan (100 mg/kg/day) for 25 days, and Group IV: colchicine (15 µg/5 µl/bilaterally, icv) + oral valsartan (50 mg/kg/day) for 25 days. Behavioral assessment was done using Morris water maze and passive avoidance tasks. Biochemically, ß-amyloid (1-40 and 1-42) peptides, oxidative stress (malondialdehyde and superoxide dismutase) and inflammatory (tumor necrosis factor-alpha) parameters were measured in hippocampus and prefrontal cortex. Sacubitril/valsartan exaggerated colchicine-induced cognitive impairment in both Morris water maze and passive avoidance tasks and was associated with significant increase in ß-amyloid accumulation, oxidative stress, and inflammation versus valsartan. Sacubitril/valsartan caused deleterious effect on cognitive impairment and biochemical alterations in colchicine-induced AD rat model. Hence, special caution should be taken following long-term intake of ARNI on cognitive functions.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Insuficiência Cardíaca , Doenças Neurodegenerativas , Masculino , Ratos , Animais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Tetrazóis/farmacologia , Colchicina/toxicidade , Antagonistas de Receptores de Angiotensina , Ratos Wistar , Valsartana/farmacologia , Compostos de Bifenilo , Combinação de Medicamentos , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/tratamento farmacológico , Peptídeos beta-Amiloides
5.
Chem Biol Interact ; 368: 110193, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36179773

RESUMO

Colchicine (COL) has been used to treat gout for over a millennium, but its medicinal use has been controversial due to its potent toxicity in the gastrointestinal tract. Nausea, vomiting, and diarrhea are the most prominent external manifestations of COL gastrointestinal toxicity, but the cause of these adverse events remains obscure. In this study, the mice were exposed to COL (2.5 mg/kg b.w./day) for one week to study the mechanism of COL-induced diarrhea from the perspective of intestinal metabolism. The results showed that COL exposure disturbed intestinal metabolic homeostasis, resulting in a significant accumulation of 116 metabolites and, conversely, significant depletion of 64 metabolites, with the number of differential metabolites being one-eighth of the total metabolites (180/1445). Also, it was found that cAMP, Adenosine 5'-monophosphate, GDP, Inositol, and Cortisol are core metabolites that play crucial roles in COL-induced metabolic disorders. These metabolites could be used as biomarkers to differentiate control and COL-treated groups, implying that these metabolites may be closely related to COL-induced diarrhea. Furthermore, changes in the metabolic pathways (Purine metabolism, biosynthesis and metabolism of aromatic amino acids, and Bile secretion) involved in these five core metabolites increased the toxic load in the gut, which was the culprit leading to intestinal metabolic disorders. In addition, the abnormal bile secretion caused by COL exposure may play an important role in COL-induced diarrhea. In conclusion, our study opens new avenues for understanding the mechanisms of COL-induced gastrointestinal adverse reactions and broadens the scientific horizon on the interactions between COL and host metabolism.


Assuntos
Colchicina , Metaboloma , Camundongos , Animais , Colchicina/toxicidade , Colchicina/análise , Fezes/química , Diarreia/induzido quimicamente , Homeostase , Metabolômica
6.
Sci Rep ; 11(1): 9034, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33907227

RESUMO

10-Alkylthiocolchicines have been obtained and characterized by spectroscopic methods and their biological activities as: cytotoxic, anti-inflammatory and analgesic activities have been tested. Cytotoxic activity against SKOV-3 ovarian cell line for 10-alkylthiocolchicine analogues was reported and tested compounds showed to be more active than commonly used doxorubicin. Some of tested C-10 alkylthiolated colchicines have been found to exhibit cytotoxicity at levels comparable to that of the natural product-colchicine. 10-Methylthiocolchicine has IC50 = 8 nM and 10-ethylthiocolchicine has IC50 = 47 nM in comparison to colchicine IC50 = 37 nM. Moreover for 10-alkylthioderivatives apoptosis test, cyclin B1 and cell cycle tests were performed. 10-n-Butylthiocolchicine was tested for anti-inflammatory and analgesic activities it showed to produce analgesic rather than anti-inflammatory effect.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Colchicina/farmacologia , Analgésicos/química , Animais , Anti-Inflamatórios não Esteroides/química , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral , Colchicina/análogos & derivados , Colchicina/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Masculino , Ratos , Ratos Wistar , Compostos de Enxofre/farmacologia
7.
Eur J Med Chem ; 207: 112724, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32827941

RESUMO

Two series of heterocyclic colchicinoids bearing ß-methylenedihydrofuran or 2H-pyran-2-one fragments were synthesized by the intramolecular Heck reaction. Methylenedihydrofuran compounds 9a and 9h were found to be the most cytotoxic among currently known colchicinoids, exhibiting outstanding antiproliferative activity on tumor cell lines in picomolar (0.01-2.1 nM) range of concentrations. Compound 9a potently and substoichiometrically inhibits microtubule formation in vitro, being an order of magnitude more active in this assay than colchicine. Derivatives 9a and 9h revealed relatively low acute toxicity in mice (LD50 ≥ 10 mg/kg i.v.). The X-Ray structure of colchicinoid 9a bound to tubulin confirmed interaction of this compound with the colchicine binding site of tubulin.


Assuntos
Antimitóticos/química , Antimitóticos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Colchicina/análogos & derivados , Colchicina/farmacologia , Animais , Antimitóticos/toxicidade , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colchicina/toxicidade , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Furanos/química , Furanos/farmacologia , Furanos/toxicidade , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/toxicidade
8.
J Biol Chem ; 294(48): 18099-18108, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31628188

RESUMO

KXO1 (tirbanibulin or KX2-391) is as a non-ATP-competitive inhibitor of SRC proto-oncogene nonreceptor tyrosine kinase (SRC) and is being clinically investigated for the management of various cancers and actinic keratosis. Recently, KXO1 has also been shown to strongly inhibit tubulin. Interestingly, unlike conventional tubulin-targeting drugs, KXO1 has exhibited low toxicity in preclinical and clinical studies, but the reason for this remains elusive, as are the KXO1-binding site and other details of the interaction of KXO1 with tubulin. Here, cell-based experiments revealed that KXO1 induces tubulin depolymerization and G2/M phase cell cycle arrest at low nanomolar concentrations, similar to colchicine, used as a positive control. Results from biochemical experiments, including an N,N-ethylenebis(iodoacetamide) competition assay, disclosed that KXO1 binds to the colchicine-binding site on ß-tubulin, further confirmed by the crystal structure of the tubulin-KXO1 complex at 2.5-Å resolution. A high-quality electron density map of the crystallographic data enabled us to unambiguously determine the position and orientation of KXO1 in the colchicine-binding site, revealing the detailed interactions between KXO1 and tubulin. We also found that KXO1 binds reversibly to purified tubulin, induces a totally reversible cellular effect (G2/M cell cycle arrest), and possesses no cellular toxicity 5 days after drug washout, explaining KXO1's low toxicity. In summary, we show that KXO1 binds to the colchicine-binding site of tubulin and resolved the crystal structure of the tubulin-KXO1 complex. Importantly, KXO1's reversible binding to tubulin explains its clinically low toxicity, an insight that could guide further clinical applications of KXO1.


Assuntos
Antineoplásicos/química , Colchicina/química , Proteínas de Neoplasias/química , Tubulina (Proteína)/química , Antineoplásicos/farmacologia , Sítios de Ligação , Divisão Celular/efeitos dos fármacos , Colchicina/toxicidade , Cristalografia por Raios X , Fase G2/efeitos dos fármacos , Células HeLa , Humanos , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Proto-Oncogene Mas , Tubulina (Proteína)/metabolismo
9.
Artigo em Inglês | MEDLINE | ID: mdl-31561895

RESUMO

In accordance with the 3 Rs to reduce in vivo testing, more advanced in vitro models, moving from 2D monolayer to 3D cultures, should be developed for prediction of human toxicity of industrial chemicals and environmental pollutants. In this study we compared cytotoxic and genotoxic responses induced by chemicals in 2D and 3D spheroidal cultures of the human liver cancer cell line HepG2. HepG2 spheroids were prepared by hanging drop technology. Both 3D spheroids and 2D monolayer cultures were exposed to different chemicals (colchicine, chlorpromazine hydrochloride or methyl methanesulfonate) for geno- and cytotoxicity studies. Cytotoxicity was investigated by alamarBlue assay, flow cytometry and confocal imaging. DNA damage was investigated by the comet assay with and without Fpg enzyme for detection of DNA strand breaks and oxidized or alkylated base lesions. The results from the cyto- and genotoxicity tests showed differences in sensitivity comparing the 2D and 3D HepG2 models. This study shows that human 3D spheroidal hepatocellular cultures can be successfully applied for genotoxicity testing by the comet assay and represent a promising advanced in vitro model for toxicity testing.


Assuntos
Ensaio Cometa/métodos , Dano ao DNA , Células Hep G2/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Clorpromazina/toxicidade , Colchicina/toxicidade , Ensaio Cometa/normas , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/genética , Guanina/análogos & derivados , Guanina/análise , Humanos , Peróxido de Hidrogênio/toxicidade , Ensaio de Proficiência Laboratorial , Metanossulfonato de Metila/toxicidade , Sensibilidade e Especificidade
10.
J Biochem Mol Toxicol ; 33(9): e22366, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31332882

RESUMO

Colchicine (COL) is an alkaloid existing in plants of Liliaceous colchicum. It has widely been used in the treatments of many diseases, such as gout, Familial Mediterranean Fever, and tumor. However, the adverse effects of COL are an obstacle to its safe use. The present studies explored the role of metabolic demethylation in the development of COL-induced hepatotoxicity. We found that inhibition of CYP3A increased the susceptibility of mice to COL hepatotoxicity, and induction of CYP3A decreased the susceptibility of animals to the hepatotoxicity. The toxicokinetic study demonstrated that pretreatment with ketoconazole caused elevated area under the concentration-time curve of COL. Three demethylation metabolites of COL were found to be less hepatotoxic than the parent compound. It appears that the formation of electrophilic demethylation metabolites was not involved in the development of COL-induced liver injury.


Assuntos
Colchicina/farmacocinética , Colchicina/toxicidade , Fígado/efeitos dos fármacos , Animais , Citocromo P-450 CYP3A/metabolismo , Cetoconazol/administração & dosagem , Fígado/metabolismo , Masculino , Metilação , Camundongos
11.
Int J Mol Sci ; 18(10)2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-29048371

RESUMO

Our previous study demonstrated that colchicine-induced dentate granule cell death is caused by blocking axonal flow and the accumulation of intracellular zinc. Zinc is concentrated in the synaptic vesicles via zinc transporter 3 (ZnT3), which facilitates zinc transport from the cytosol into the synaptic vesicles. The aim of the present study was to identify the role of ZnT3 gene deletion on colchicine-induced dentate granule cell death. The present study used young (3-5 months) mice of the wild-type (WT) or the ZnT3-/- genotype. Colchicine (10 µg/kg) was injected into the hippocampus, and then brain sections were evaluated 12 or 24 h later. Cell death was evaluated by Fluoro-Jade B; oxidative stress was analyzed by 4-hydroxy-2-nonenal; and dendritic damage was detected by microtubule-associated protein 2. Zinc accumulation was detected by N-(6-methoxy-8-quinolyl)-para-toluenesulfonamide (TSQ) staining. Here, we found that ZnT3-/- reduced the number of degenerating cells after colchicine injection. The ZnT3-/--mediated inhibition of cell death was accompanied by suppression of oxidative injury, dendritic damage and zinc accumulation. In addition, ZnT3-/- mice showed more glutathione content than WT mice and inhibited neuronal glutathione depletion by colchicine. These findings suggest that increased neuronal glutathione by ZnT3 gene deletion prevents colchicine-induced dentate granule cell death.


Assuntos
Proteínas de Transporte/genética , Giro Denteado/metabolismo , Deleção de Genes , Proteínas de Membrana/genética , Neurônios/metabolismo , Animais , Transporte Axonal , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions , Morte Celular , Colchicina/toxicidade , Giro Denteado/citologia , Giro Denteado/efeitos dos fármacos , Glutationa/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Zinco/metabolismo
12.
Basic Clin Pharmacol Toxicol ; 120(5): 505-508, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27862994

RESUMO

The traditional gout medication colchicine has been reported to effectively prevent atrial fibrillation recurrence after atrial fibrillation ablation or cardiac surgery in a few clinical trials. Severe adverse events have not yet been reported. The aim of the present study was to assess possible direct electrophysiological effects in an experimental whole-heart model. Ten rabbit hearts were isolated and Langendorff-perfused. Thereafter, colchicine was administered in two concentrations (1 and 3 µM). Eight endo- and epicardial monophasic action potentials and a 12-lead ECG showed a stable QT interval and action potential duration during colchicine infusion. Furthermore, there was no significant increase in dispersion of repolarization. However, colchicine induced a dose-dependent significant decrease of effective refractory period (ERP; 1 µM: -19 ms, 3 µM: -22 ms; p < 0.05). In the present study, acute infusion of colchicine in isolated rabbit hearts resulted in a reduction of ERP in the presence of a stable myocardial repolarization. This led to a significantly elevated inducibility of ventricular fibrillation. In 4 of 10 hearts, incessant ventricular fibrillation occurred. These results suggest a pro-arrhythmic or toxic effect of colchicine and underline that further clinical studies on potential adverse effects should be conducted before the drug can be recommended for routine use after atrial fibrillation ablation.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Colchicina/toxicidade , Supressores da Gota/toxicidade , Fibrilação Ventricular/induzido quimicamente , Animais , Fibrilação Atrial/prevenção & controle , Colchicina/administração & dosagem , Colchicina/farmacologia , Relação Dose-Resposta a Droga , Eletrocardiografia , Técnicas Eletrofisiológicas Cardíacas , Supressores da Gota/administração & dosagem , Supressores da Gota/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Coelhos
13.
Anticancer Res ; 36(11): 5859-5866, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27793909

RESUMO

BACKGROUND/AIM: Colchicine (COL) is a well-known and potent microtubule targeting anticancer agent. The purpose of our study was to identify conditions that increase sensitization of COL-resistant cancer cells that overexpress P-glycoprotein (P-gp). MATERIALS AND METHODS: The anti-malarial drugs chloroquine (CHL), mefloquine (MEF) and primaquine (PRI) have been shown to increase sensitization in drug-resistant KBV20C cells via P-gp inhibition. Therefore, we tested whether co-treatment of COL with PRI, CHL or MEF increases sensitivity in COL-resistant KBV20C cells over that of cells treated with COL alone and whether these effects are attributable to P-gp activity. RESULTS: Interestingly, we found that both CHL and PRI, but not MEF, reduced cytotoxicity in KBV20C cells receiving high concentrations of COL, suggesting that the effects of CHL and PRI have specific mechanisms among the anti-malarial drugs. The effects of CHL and PRI were specific to COL-resistant cells, since we did not detect a reduction in cytotoxicity in drug-sensitive parent KB cells. These data suggest that CHL and PRI inhibit the signaling pathways of COL-treated-resistant cells without P-gp inhibition. Furthermore, we studied the molecular mechanisms underlying the effects of COL-CHL co-treatment in KBV20C cells. FACS analysis, annexin V staining and western blot analysis revealed that G2 arrest and apoptosis were lower in cells co-treated with COL and CHL than in cells treated with COL alone. We also found that pH2AX, pHistone H3 and pRb expression was highly reduced in COL-CHL co-treated cells but not in COL-VIB co-treated cells. In addition, expression of the p21 protein, which correlates with drug-resistant phenotypes, increased in cells receiving COL-CHL co-treatment over that of COL-treated cells. CONCLUSION: These results suggest that reduced G2 arrest and apoptosis resulting from COL-CHL co-treatment was attributable to DNA damage and reduced cell cycle progression. These findings provide important information regarding the prevention of COL toxicity in COL-resistant cells and indicate that CHL, PRI and MEF may contribute to sensitization in COL-resistant cells.


Assuntos
Antimaláricos/farmacologia , Colchicina/toxicidade , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Histonas/metabolismo , Humanos
14.
Talanta ; 161: 804-811, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27769486

RESUMO

With the development of large-scale biologic databases, precision medicine is becoming a frontier in biomedical research. As a main focus of precision medicine study, cancer has been widely accepted as a disease born out of inherited genetic variations or accumulating genomic damage. At the single-cell level, microfluidics or lab-on-a-chip technology for cancer study is an emerging tool for improving risk assessment, diagnostic categories and therapeutic strategies. This work presents a multi-layer microchip for single-cell gene expression profiling. Treated by three drug reagents (i.e. methyl methanesulfonate, docetaxel and colchicine) with varied concentrations and time lengths, individual human breast cancer cells (MCF-7) are then lysed on-chip, and the released mRNA templates are captured and reversely transcribed into cDNA on microbead surface. Three genes (GAPDH, CDKN1A, AURKA) are amplified and quantified simultaneously through triplex real-time polymerase chain reactions (qPCR). Readout per run is set to be eighteen, and can be further improved following same approach. The microchip is able to integrate all steps of single-cell gene expression profiling, and provide precision study of drug induced genotoxicity with reduced reagents consumption per reaction and instrumental cost.


Assuntos
Aurora Quinase A/genética , Colchicina/toxicidade , Inibidor de Quinase Dependente de Ciclina p21/genética , Gliceraldeído-3-Fosfato Desidrogenases/genética , Dispositivos Lab-On-A-Chip , Metanossulfonato de Metila/toxicidade , Taxoides/toxicidade , Colchicina/farmacologia , Docetaxel , Perfilação da Expressão Gênica , Humanos , Células MCF-7 , Metanossulfonato de Metila/farmacologia , Microfluídica , Testes de Mutagenicidade , Reação em Cadeia da Polimerase em Tempo Real , Taxoides/farmacologia
15.
Chem Res Toxicol ; 29(3): 342-51, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26881866

RESUMO

Human toxicity screening is an important stage in the development of safe drug candidates. Hepatotoxicity is one of the major reasons for the withdrawal of drugs from the market because the liver is the major organ involved in drug metabolism, and it can generate toxic metabolites. There is a need to screen molecules for drug-induced hepatotoxicity in humans at an earlier stage. Transcriptomics is a technique widely used to screen molecules for toxicity and to unravel toxicity mechanisms. To date, the majority of such studies were performed using animals or animal cells, with concomitant difficulty in interpretation due to species differences, or in human hepatoma cell lines or cultured hepatocytes, suffering from the lack of physiological expression of enzymes and transporters and lack of nonparenchymal cells. The aim of this study was to classify known hepatotoxicants on their phenotype of toxicity in humans using gene expression profiles ex vivo in human precision-cut liver slices (PCLS). Hepatotoxicants known to induce either necrosis (n = 5) or cholestasis (n = 5) were used at concentrations inducing low (<30%) and medium (30-50%) cytotoxicity, based on ATP content. Random forest and support vector machine algorithms were used to classify hepatotoxicants using a leave-one-compound-out cross-validation method. Optimized biomarker sets were compared to derive a consensus list of markers. Classification correctly predicted the toxicity phenotype with an accuracy of 70-80%. The classification is slightly better for the low than for the medium cytotoxicity. The consensus list of markers includes endoplasmic reticulum stress genes, such as C2ORF30, DNAJB9, DNAJC12, SRP72, TMED7, and UBA5, and a sodium/bile acid cotransporter (SLC10A7). This study shows that human PCLS are a useful model to predict the phenotype of drug-induced hepatotoxicity. Additional compounds should be included to confirm the consensus list of markers, which could then be used to develop a biomarker PCR-array for hepatotoxicity screening.


Assuntos
Colestase/induzido quimicamente , Perfilação da Expressão Gênica , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Necrose/induzido quimicamente , Toxicogenética , Acetaminofen/toxicidade , Idoso , Benzofuranos/toxicidade , Ácidos e Sais Biliares/toxicidade , Cloranfenicol/toxicidade , Clorpromazina/toxicidade , Colestase/genética , Colchicina/toxicidade , Ciclosporina/toxicidade , Dietilnitrosamina/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Etinilestradiol/toxicidade , Feminino , Hepatócitos/metabolismo , Humanos , Masculino , Metiltestosterona/toxicidade , Pessoa de Meia-Idade , Necrose/genética , Fenótipo , Adulto Jovem
16.
J Immunotoxicol ; 13(4): 474-89, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26788903

RESUMO

The neurodegeneration in intracerebroventricular (icv) colchicine injected (ICIR) rats is linked with neuroinflammation. Glutamate excitotoxicity through NMDA receptors is involved with the neuroinflammation in some animal models of Alzheimer Disease (AD), but it has not been explored in ICIR rats. The aim of this study was to investigate the role of NMDA receptors (by blocking it's activity with memantine) in colchicine-induced neuroinflammation and neurodegeneration and impacts on peripheral immune parameters in ICIR rats. Levels of inflammatory markers (IL-1ß, TNFα, ROS, nitrite) in the hippocampus and serum, histopathology of the hippocampus and select peripheral immune parameters were measured 14 and 21-days after icv colchicine injection in rats. These parameters were also measured in rats that received daily per os administration of memantine (20 mg/kg) in both study durations. Neuroinflammation in the hippocampus of ICIR rats was associated with neurodegeneration (chromatolysis, plaque formation), along with changes in inflammatory markers in the serum and alterations in peripheral immune parameters (phagocytic activity of WBC and splenic PMN, cytotoxic activity/leukocyte adhesion inhibition by splenic MNC). Administration of memantine to ICIR rats resulted in mitigation of colchicine-induced inflammation in the hippocampus, inflammatory markers in the serum and neurodegeneration and also led to recovery of the measured immune endpoints; most of these effects were greater with the longer duration of study. Phagocytic activity of WBC and splenic PMN cells appeared to correlate with levels of the measured central inflammatory markers. It appears from the results that neuroinflammation might be linked with the NMDA receptor activity in ICIR rats and that this receptor is involved in the process of progressive neuroinflammation and neurodegeneration in the hippocampus of ICIR and potentially in immunomodulation in these same hosts.


Assuntos
Doença de Alzheimer/imunologia , Memantina/administração & dosagem , Inflamação Neurogênica/imunologia , Neurotoxinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Adesão Celular , Colchicina/toxicidade , Citotoxicidade Imunológica , Modelos Animais de Doenças , Humanos , Imunomodulação , Mediadores da Inflamação/metabolismo , Infusões Intraventriculares , Masculino , Memantina/farmacologia , Camundongos , Neurotoxinas/antagonistas & inibidores , Fagocitose , Ratos , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
17.
Invest New Drugs ; 34(1): 129-37, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26686345

RESUMO

The subunit protein of microtubules is tubulin, which has been the target for some of the most successful and widely used anti-tumor drugs. Most of the drugs that target tubulin bind to the ß subunit. There are many isotypes of ß-tubulin and their distributions differ among different tissues. The ßIII isotype is over-expressed in many tumors, particularly those that are aggressive, metastatic, and drug resistant. We have previously reported the design and synthesis of a series of compounds to fit the colchicine site on ßIII but not on the other isotypes. In the current study, we tested the toxicity and the anti-tumor activity of one of these compounds, CH-35, on the human breast tumor MDA-MB-231 over-expressing ßIII in a xenogeneic mouse model. We found that CH-35 was as toxic as Taxol® in vivo. Although the ßIII-over-expressing cells developed into very fast-growing tumors, CH-35 was more effective against this tumor than was Taxol. Our results suggest that CH-35 is a promising candidate for future drug development.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Colchicina/análogos & derivados , Tubulina (Proteína)/genética , Animais , Antineoplásicos/toxicidade , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Colchicina/química , Colchicina/farmacologia , Colchicina/toxicidade , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/farmacologia , Paclitaxel/toxicidade , Testes de Toxicidade
18.
Clin Toxicol (Phila) ; 53(5): 427-32, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25858137

RESUMO

CONTEXT: Colchicine is an anti-inflammatory alkaloid used for the treatment of acute gout, but has a narrow therapeutic index. Colchicine overdoses are relatively rare, but have high mortality requiring rapid treatment. OBJECTIVE: To evaluate the ability of a newly available ovine fragment antigen-binding (Fab) antibody to colchicine (ColchiFab(™)) to protect rats against renal and other injury 24 h after colchicine ingestion. MATERIALS AND METHODS: Rats were gavaged with colchicine (5 mg/kg), then 2 h later injected intraperitoneally with 5 ml of sterile saline, or Fab anti-colchicine, a newly available ovine antibody to colchicine. Samples of blood were taken at 1, 2, 5 and 24 h after gavage, and urine was collected from 5 to 24 h after gavage. Concentrations of colchicine in tissue, blood and urine were measured by liquid chromatography/mass spectrometry, concentrations of Fab anti-colchicine, urinary neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 or KIM-1 by enzyme-linked immunosorbent assay or ELISA, while concentrations of creatine kinase and creatinine (Cr) were measured enzymatically. RESULTS: Colchicine equilibrated rapidly throughout the body and increased serum creatine kinase. Fab anti-colchicine also rapidly redistributed to the blood and remained at high concentrations over 24 h. Fab anti-colchicine caused a rapid 7.1-fold increase in serum colchicine level, followed by excretion of both colchicine and Fab anti-colchicine through the urine. This was associated with the accumulation of colchicine in the kidney, a reversal of colchicine-induced diarrhoea, and increasing urinary NGAL level; from 168 ± 48 to 477 ± 255 ng/mmol Cr [mean ± standard deviation or SD]. DISCUSSION: Fab anti-colchicine greatly increased the clearance of colchicine, although increasing NGAL level suggested the presence of mild kidney damage. CONCLUSION: These data suggest clinical utility for Fab anti-colchicine in the treatment of colchicine overdose.


Assuntos
Anti-Inflamatórios/toxicidade , Antídotos/farmacologia , Colchicina/toxicidade , Fragmentos Fab das Imunoglobulinas/farmacologia , Intoxicação/tratamento farmacológico , Proteínas de Fase Aguda/urina , Animais , Anti-Inflamatórios/sangue , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/urina , Antídotos/administração & dosagem , Antídotos/farmacocinética , Biomarcadores/sangue , Biomarcadores/urina , Moléculas de Adesão Celular/urina , Colchicina/sangue , Colchicina/imunologia , Colchicina/farmacocinética , Colchicina/urina , Creatina Quinase/sangue , Diarreia/induzido quimicamente , Diarreia/diagnóstico , Diarreia/tratamento farmacológico , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Fragmentos Fab das Imunoglobulinas/sangue , Fragmentos Fab das Imunoglobulinas/urina , Injeções Intraperitoneais , Rim/efeitos dos fármacos , Rim/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/diagnóstico , Nefropatias/tratamento farmacológico , Lipocalina-2 , Lipocalinas/urina , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Taxa de Depuração Metabólica , Intoxicação/sangue , Intoxicação/urina , Proteínas Proto-Oncogênicas/urina , Ratos , Ratos Sprague-Dawley , Eliminação Renal/efeitos dos fármacos , Ovinos , Distribuição Tecidual
19.
Artigo em Inglês | MEDLINE | ID: mdl-25344161

RESUMO

Following the initial observation that methyl methanesulphonate induced binucleated cells in the AHH-1 line and a significant number of them contained micronuclei, human lymphoblastoid TK6 and mouse lymphoma L5178Y cells were treated with methyl methanesulphonate, methylnitrosourea, mitomycin C, cytosine arabinoside, colchicine and triton X. All except triton X induced binucleated cells in both lines but an increased micronucleus incidence in them was seen only in TK6. The two lines also differed in the numbers of binucleates in the control cultures with 2.0% and 0.5% in TK6 and L5178Y, respectively, and a much higher proportion of those in TK6 contained micronuclei. The differences in behaviour between the two cell lines could not clearly be ascribed to their P53 status. Colchicine induced binucleates in both cell lines but they did not contain increased numbers of micronuclei. The effect on binucleate incidence was not a non-specific cytotoxic response because no increase was seen with triton X even at highly cytotoxic concentrations. The initial concern that not scoring micronuclei in binucleated cells might lead to erroneous results in in vitro micronucleus tests not using a cytokinesis block, was not proven because all the genotoxins tested here induced significant increases in micronucleus frequency in mononuclear cells. When testing less potently active agents in in vitro micronucleus tests not employing a cytokinesis block, care should be taken to understand better this phenomenon and not to include these damaged cells until we do.


Assuntos
Testes para Micronúcleos/métodos , Mutagênicos/toxicidade , Animais , Linhagem Celular , Colchicina/toxicidade , Citarabina/toxicidade , Citocinese/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Humanos , Metanossulfonato de Metila/toxicidade , Metilnitrosoureia/toxicidade , Camundongos , Mitomicina/toxicidade
20.
Eur J Pharmacol ; 721(1-3): 373-81, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24075937

RESUMO

Oxidative-nitrosative stress and mitochondrial dysfunction plays an important role in the onset of various neurodegenerative diseases. Lycopene, a carotenoid antioxidant, has received considerable scientific interest in recent years. Present study was designed to evaluate the possible nitric oxide mechanism in protective effects of lycopene against the colchicine induced cognitive impairment and mito-oxidative damage in rats. Wistar rats were received i.c.v. colchicine (15 µg/5 µl). Lycopene (2.5 and 5mg/kg), NO modulators e.g. l-Arginine (50mg/kg) l-NAME (5mg/kg) administered for 21 days. Behavioural alterations were assessed in between study period. Animals were killed immediately following the last behavioral session, and mitochondrial enzymes, oxidative parameters, inflammatory mediators (TNF-α, IL-6) and caspase-3 activity were measured. I.C.V. administration of colchicine impaired memory performance in Morris water maze, oxidative defense and mitochondrial complex enzymes activities as compared to sham group. A significant increase of TNF-α, IL-6 and caspase-3 activity in hippocampus and cortex was also noted. Chronic treatment lycopene significantly improved memory retention and attenuated mito-oxidative damage parameters, inflammatory markers and apoptosis in colchicine treated rats. Further, l-arginine pretreatment with sub effective dose of lycopene significantly reversed the protective effect of lycopene. However, l-NAME pretreatment with sub effective dose of lycopene significantly potentiated the protective effect of lycopene which was significant as compared to their effect per se. These results suggest that lycopene exhibit a neuroprotective effect by accelerating brain anti-oxidant defense mechanisms and down regulating nitric oxide pathways. Thus, lycopene may be used as therapeutic agent in preventing complications in memory dysfunction.


Assuntos
Carotenoides/farmacologia , Colchicina/toxicidade , Memória/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Acetilcolinesterase/metabolismo , Animais , Arginina/farmacologia , Caspase 3/metabolismo , Cognição/efeitos dos fármacos , Interleucina-6/metabolismo , Licopeno , Masculino , Mitocôndrias/efeitos dos fármacos , Atividade Motora/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA