Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 413
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Appl Toxicol ; 44(6): 863-873, 2024 06.
Article in English | MEDLINE | ID: mdl-38311468

ABSTRACT

Although the medicinal properties of colchicine (COL) have been widely known for centuries, its toxicity has been the subject of controversy. The narrow therapeutic window causes COL to induce gastrointestinal adverse effects even when taken at recommended doses, mainly manifested as nausea, vomiting, and diarrhea. However, the mechanism of COL-induced gastrointestinal toxic reactions remains obscure. In the present study, the mice were dosed with COL (2.5 mg/kg b.w./day) for a week to explore the effect of COL on bile acid metabolism and the mechanism of COL-induced diarrhea. The results showed that COL treatment affected liver biochemistry in mice, resulting in a significant down-regulation of the mRNA expression levels of bile acid biosynthesis regulators Cyp7a1, Cyp8b1, Cyp7b1, and Cyp27a1 in liver tissues. The mRNA expression levels of bile acid transporters Ntcp, Oatp1, Mrp2, Ibabp, Mrp3, Osta, and Ostb in liver and ileum tissues were also significantly down-regulated. In addition, COL treatment significantly inhibited the mRNA expression levels of Fxr and its downstream target genes Shp, Lrh1, and Fgf15 in liver and ileum tissues, affecting the feedback regulation of bile acid biosynthesis. More importantly, the inhibition of COL on bile acid transporters in ileal and hepatic tissues affected bile acid recycling in the ileum as well as their reuptake in the liver, leading to a significantly increased accumulation of bile acids in the colon, which may be an important cause of diarrhea. In conclusion, our study revealed that COL treatment affected bile acid biosynthesis and enterohepatic circulation, thereby disrupting bile acid metabolic homeostasis in mice.


Subject(s)
Bile Acids and Salts , Colchicine , Enterohepatic Circulation , Homeostasis , Liver , Animals , Bile Acids and Salts/metabolism , Enterohepatic Circulation/drug effects , Colchicine/toxicity , Homeostasis/drug effects , Liver/drug effects , Liver/metabolism , Mice , Male , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Ileum/drug effects , Ileum/metabolism , Diarrhea/chemically induced
2.
J Microsc ; 291(3): 229-236, 2023 09.
Article in English | MEDLINE | ID: mdl-37358710

ABSTRACT

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.


Subject(s)
Colchicine , Hepatocytes , Animals , Chlorocebus aethiops , Colchicine/toxicity , Biomechanical Phenomena , Vero Cells , Microscopy, Atomic Force
3.
Mutagenesis ; 37(3-4): 213-225, 2022 10 26.
Article in English | MEDLINE | ID: mdl-35869703

ABSTRACT

Two prototypical genotoxicants, benzo[a]pyrene (B[a]P) and colchicine (COL), were selected as model compounds to deduce their quantitative genotoxic dose-response relationship at low doses in a multi-endpoint genotoxicity assessment platform. Male Sprague-Dawley rats were treated with B[a]P (2.5-80 mg/kg bw/day) and COL (0.125-2 mg/kg bw/day) daily for 28 days. The parameters included were as follows: comet assay in the peripheral blood and liver, Pig-a gene mutation assay in the peripheral blood, and micronucleus test in the peripheral blood and bone marrow. A significant increase was observed in Pig-a mutant frequency in peripheral blood for B[a]P (started at 40 mg/kg bw/day on Day 14, started at 20 mg/kg bw/day on Day 28), whereas no statistical difference for COL was observed. Micronucleus frequency in reticulocytes of the peripheral blood and bone marrow increased significantly for B[a]P (80 mg/kg bw/day on Day 4, started at 20 mg/kg bw/day on Days 14 and 28 in the blood; started at 20 mg/kg bw/day on Day 28 in the bone marrow) and COL (started at 2 mg/kg bw/day on Day 14, 1 mg/kg bw/day on Day 28 in the blood; started at 1 mg/kg bw/day on Day 28 in the bone marrow). No statistical variation was found in indexes of comet assay at all time points for B[a]P and COL in the peripheral blood and liver. The dose-response relationships of Pig-a and micronucleus test data were analyzed for possible point of departures using three quantitative approaches, i.e., the benchmark dose, breakpoint dose, and no observed genotoxic effect level. The practical thresholds of the genotoxicity of B[a]P and COL estimated in this study were 0.122 and 0.0431 mg/kg bw/day, respectively, and our results also provided distinct genotoxic mode of action of the two chemicals.


Subject(s)
Benzo(a)pyrene , Colchicine , Rats , Animals , Male , Benzo(a)pyrene/toxicity , Colchicine/toxicity , Rats, Sprague-Dawley , Erythrocytes , Micronucleus Tests/methods , Comet Assay/methods , Reticulocytes , DNA Damage , Dose-Response Relationship, Drug , Mutagenicity Tests/methods
4.
Arch Toxicol ; 96(11): 3067-3076, 2022 11.
Article in English | MEDLINE | ID: mdl-36102954

ABSTRACT

Colchicine is an anti-inflammatory drug with a narrow therapeutic index. Its binding to tubulin prevents microtubule polymerization; however, little is known about how depolymerization of microtubules interferes with the phagocytosis function of Kupffer cells (KC). Here, we applied functional intravital imaging techniques to investigate the influence of microtubule disruption by colchicine on KC morphology, as well as its capacity to clear foreign particles and bacterial lipopolysaccharide (LPS) in anesthetized mice. Intravital imaging of KC in healthy mice showed the typical elongated morphology, localization at the luminal side of the sinusoidal endothelial cells, and moving cell protrusions. In contrast, at colchicine doses of 1 mg/kg and higher (intraperitoneal), KC appeared roundish with strongly reduced protrusions and motility. To study the functional consequences of these alterations, we analyzed the capacity of KC to phagocytose fluorescent nanospheres (100 nm-size) and LPS. After tail vein injection, the nanospheres formed aggregates of up to ~ 5 µm moving along the sinusoidal bloodstream. In controls, the nanosphere aggregates were rapidly captured by the Kupffer cell protrusions, followed by an internalization process that lasted up to 10 min. Similar capture events and internalization processes were observed after the administration of fluorescently labeled LPS. In contrast, capture and internalization of both nanospheres and LPS by KC were strongly reduced in colchicine-treated mice. Reduced phagocytosis of LPS was accompanied by aggravated production of inflammatory cytokines. Since 0.4 mg/kg colchicine in mice has been reported to be bio-equivalent to human therapeutic doses, the here-observed adverse effects on KC occurred at doses only slightly above those used clinically, and may be critical for patients with endotoxemia due to a leaky gut-blood barrier.


Subject(s)
Kupffer Cells , Lipopolysaccharides , Animals , Anti-Inflammatory Agents/pharmacology , Colchicine/metabolism , Colchicine/toxicity , Cytokines/metabolism , Endothelial Cells/metabolism , Endotoxins , Humans , Lipopolysaccharides/toxicity , Mice , Tubulin/metabolism
5.
J Appl Toxicol ; 41(4): 595-606, 2021 04.
Article in English | MEDLINE | ID: mdl-33067908

ABSTRACT

The micronucleus test (MNT) is the most widely applied short-term assay to detect clastogens or spindle disruptors. The use of flow cytometry (FCM) has been reported for micronucleated erythrocytes scoring in peripheral blood. The aim of this study was to develop a novel and practical protocol for MNT in rat peripheral blood by FCM, with the method validation. CD71-fluorescein isothiocyanate and DRAQ5 were adopted for the fluorescent staining of proteins and DNA, respectively, to detect micronuclei. To validate the method, groups of male Sprague-Dawley rats (five per group) received two oral gavage doses at 0 and 24 h of six chemicals (four positive mutagens: ethyl methanesulphonate [EMS], cyclophosphamide [CP], colchicine [COL], and ethyl nitrosourea [ENU]; two nongenotoxic chemicals: sodium saccharin and eugenol). Blood samples were collected from the tail vein before and on the five continuous days after treatments; all of which were analyzed for micronuclei presence by both the manual (Giemsa staining) and FCM methods. The FCM-based method consistently demonstrated highly sensitive responses for micronucleus detection at all concentrations and all time points for EMS, CP, COL, and ENU. Sodium saccharin and eugenol could be identified as negative in this protocol. Results obtained with the FCM-based method correlated well with the micronucleus frequencies (r = 0.659-0.952), and the proportion of immature erythrocytes (r = 0.915-0.981) tested by Giemsa staining. The method reported here, with easy operation, low background, and requirement for a regular FCM, could be an efficient system for micronucleus scoring.


Subject(s)
Flow Cytometry/methods , Leukocytes, Mononuclear/chemistry , Micronucleus Tests/methods , Mutagens/toxicity , Nitrosourea Compounds/toxicity , Reticulocytes , Animals , Colchicine/toxicity , Cyclophosphamide/toxicity , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/toxicity , Eugenol/toxicity , Male , Rats , Rats, Sprague-Dawley , Saccharin/toxicity
6.
J Biol Chem ; 294(48): 18099-18108, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31628188

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/chemistry , Colchicine/chemistry , Neoplasm Proteins/chemistry , Tubulin/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Division/drug effects , Colchicine/toxicity , Crystallography, X-Ray , G2 Phase/drug effects , HeLa Cells , Humans , Neoplasm Proteins/metabolism , Protein Binding , Proto-Oncogene Mas , Tubulin/metabolism
7.
J Biochem Mol Toxicol ; 33(9): e22366, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31332882

ABSTRACT

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.


Subject(s)
Colchicine/pharmacokinetics , Colchicine/toxicity , Liver/drug effects , Animals , Cytochrome P-450 CYP3A/metabolism , Ketoconazole/administration & dosage , Liver/metabolism , Male , Methylation , Mice
8.
Biol Pharm Bull ; 41(7): 1001-1005, 2018.
Article in English | MEDLINE | ID: mdl-29962396

ABSTRACT

Colchicine or vincristine depolymerize microtubules, an action which blocks neuron axonal transport. Thus, these chemicals showed selective neurotoxicity in hippocampal neurons. However, the mechanism of neurotoxicity by these antimicrotubule agents has remained unclear. Our previous studies have suggested that colchicine-induced hippocampal neuron death is caused by incremental increases in intraneuronal free zinc. We have demonstrated that zinc transporter 3 gene deletion (ZnT3-/-) reduces dentate granule cell death after colchicine injection. This ZnT3-/--mediated reduction of dentate granule cell death was accompanied by a decrease in the incidence of oxidative injury. Unexpectedly, we found that ZnT3-/- mice contain a higher glutathione (GSH) level in the hippocampal neurons than wild type mice. Thus, ZnT3-/- mice showed less neuronal GSH depletion by colchicine injection, and thus less neuronal death. These results suggest that the higher levels of neuronal GSH in ZnT3-/- mice result in less dentate granule cell death after colchicine injection. In addition to colchicine, our lab also demonstrated that a chemotherapeutic agent, pacritaxel (Taxol), which is a microtubule stabilizing agent, depleted vesicular zinc in the presynaptic terminals and induced a reduction of neurogenesis. Therefore, in the present review, we discussed how antimicrotubule agent-induced neurotoxicity and cognitive impairment is associated with zinc dyshomeostasis in the brain.


Subject(s)
Cell Death/drug effects , Hippocampus/drug effects , Microtubules/drug effects , Neurotoxicity Syndromes/pathology , Tubulin Modulators/toxicity , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Colchicine/toxicity , Disease Models, Animal , Hippocampus/cytology , Hippocampus/pathology , Humans , Mice , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Paclitaxel/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/pathology , Vincristine/toxicity
9.
Int J Mol Sci ; 18(10)2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29048371

ABSTRACT

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.


Subject(s)
Carrier Proteins/genetics , Dentate Gyrus/metabolism , Gene Deletion , Membrane Proteins/genetics , Neurons/metabolism , Animals , Axonal Transport , Carrier Proteins/metabolism , Cation Transport Proteins , Cell Death , Colchicine/toxicity , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Glutathione/metabolism , Male , Membrane Proteins/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Oxidative Stress , Zinc/metabolism
10.
Invest New Drugs ; 34(1): 129-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26686345

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Colchicine/analogs & derivatives , Tubulin/genetics , Animals , Antineoplastic Agents/toxicity , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Colchicine/chemistry , Colchicine/pharmacology , Colchicine/toxicity , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/pharmacology , Paclitaxel/toxicity , Toxicity Tests
11.
Chem Res Toxicol ; 29(3): 342-51, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26881866

ABSTRACT

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.


Subject(s)
Cholestasis/chemically induced , Gene Expression Profiling , Hepatocytes/drug effects , Liver/drug effects , Liver/metabolism , Necrosis/chemically induced , Toxicogenetics , Acetaminophen/toxicity , Aged , Benzofurans/toxicity , Bile Acids and Salts/toxicity , Chloramphenicol/toxicity , Chlorpromazine/toxicity , Cholestasis/genetics , Colchicine/toxicity , Cyclosporine/toxicity , Diethylnitrosamine/toxicity , Drug-Related Side Effects and Adverse Reactions , Ethinyl Estradiol/toxicity , Female , Hepatocytes/metabolism , Humans , Male , Methyltestosterone/toxicity , Middle Aged , Necrosis/genetics , Phenotype , Young Adult
12.
Genet Mol Res ; 15(2)2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27173261

ABSTRACT

Six different colchicine concentrations: 0, 400, 800, 1200, 1600, and 2000 ppm, in combination with four soaking time treatments (1, 2, 3, and 4 h), were selected to assess the effects on germination, vegetative growth, and flower yield components in calendula plants. The molecular diversity among the treatments was assessed using ten SRAP marker combinations. Seed soaking in colchicine significantly enhanced both the fresh and the dry shoot and root masses, flowering date, number of flowers per plant, and flower diameter. At 1200-ppm colchicine combined with a 4-h soaking time, a superior effect on seed germination was observed, whereas 800 ppm for 4 h produced the highest number of flowers and the largest flower diameter. The earliest flowering time was found at 800 ppm combined with a short soaking time (1 h), while the 4-h soaking time with 800 ppm, is recommended for growing calendula outdoors, since it enhances flower development. At the molecular level, 752 fragments were successfully amplified using the SRAP primers, with 280 genetic loci found throughout the calendula genome. The polymorphism percentage ranged from 79 to 100% and the polymorphic information content (PIC) values ranged between 0.85 and 0.97. The high number of detected loci and PIC values suggests a great power of SRAP markers in detecting mutant molecular diversity. Our results clearly show the existence of genetic variation among colchicine treated calendula plants and the clustering of the studied mutants was concordant with the colchicine concentration used.


Subject(s)
Calendula/drug effects , Colchicine/toxicity , Mutation , Polymorphism, Genetic , Calendula/genetics , Calendula/growth & development , Flowers/drug effects , Phenotype , Seeds/drug effects
13.
Neuroimmunomodulation ; 21(6): 304-21, 2014.
Article in English | MEDLINE | ID: mdl-24662962

ABSTRACT

BACKGROUND: The components of the immune system have been indicated to be linked with the neurotoxicity in Alzheimer's disease (AD). The participation of the immune system in the neurodegeneration in a rat model of colchicine-induced AD has not been explored. METHODS: In the present study, hippocampal neurodegeneration along with reactive oxygen species (ROS), nitrite and TNF-α in the hippocampus and some systemic immune responses were measured after 15 and 21 days of intracerebroventricular colchicine injection in rats and again after oral administration of different doses of the anti-inflammatory drug naproxen in AD rats. RESULTS: Chromatolysis and amyloid plaques were found along with higher ROS, nitrite and TNF-α levels in the hippocampus of colchicine-induced AD rats, and these changes were prevented by naproxen in a dose-dependent manner. Alterations in immunological parameters [increased phagocytic activity of white blood cells and splenic polymorphonuclear cells (PMN), increased cytotoxicity and decreased leucocyte adhesive inhibition index (LAI) of splenic mononuclear cells (MNC)] were also observed in colchicine-injected rats, which showed a dose-dependent recovery after oral administration of naproxen in AD rats. The number of plaques, chromatolysis of Nissl granules, TNF-α, nitrite and ROS levels in the hippocampus, phagocytic activity of splenic PMN and LAI of splenic MNC in AD rats showed greater changes in the 21- than in the 15-day study, and the recovery of these parameters after administration of naproxen differed between the two study durations. CONCLUSION: The present study shows that colchicine-induced neurodegeneration is time dependent and mediated by cyclooxygenase-induced neuroinflammation, which is reflected in the systemic immunological responses.


Subject(s)
Alzheimer Disease , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colchicine/toxicity , Cytokines/blood , Naproxen/therapeutic use , Tubulin Modulators/toxicity , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Animals , Body Weight/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Hydrocortisone/blood , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Nitrites/metabolism , Phagocytes/drug effects , Plaque, Amyloid/pathology , Rats , Reactive Oxygen Species/metabolism , Spleen/cytology , Time Factors
14.
Angew Chem Int Ed Engl ; 53(41): 10945-8, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25154925

ABSTRACT

Light-activatable drugs offer the promise of controlled release with exquisite temporal and spatial resolution. However, light-sensitive prodrugs are typically converted to their active forms using short-wavelength irradiation, which displays poor tissue penetrance. We report herein erythrocyte-mediated assembly of long-wavelength-sensitive phototherapeutics. The activating wavelength of the constructs is readily preassigned by using fluorophores with the desired excitation wavelength λ(ex). Drug release from the erythrocyte carrier was confirmed by standard analytical tools and by the expected biological consequences of the liberated drugs in cell culture: methotrexate, binding to intracellular dihydrofolate reductase; colchicine, inhibition of microtubule polymerization; dexamethasone, induced nuclear migration of the glucocorticoid receptor.


Subject(s)
Drug Carriers/chemistry , Erythrocytes/chemistry , Prodrugs/chemistry , Cell Survival/drug effects , Colchicine/chemistry , Colchicine/toxicity , Dexamethasone/chemistry , Dexamethasone/toxicity , Fluorescein/chemistry , HeLa Cells , Humans , Light , Methotrexate/chemistry , Methotrexate/toxicity , Prodrugs/toxicity , Receptors, Glucocorticoid/metabolism , Rhodamines/chemistry , Tetrahydrofolate Dehydrogenase/metabolism , Vitamin B 12/chemistry
15.
Acute Med ; 13(4): 171-3, 2014.
Article in English | MEDLINE | ID: mdl-25521087

ABSTRACT

A 19 year old male presented with a deliberate overdose of colchicine (50mg). He had no other significant medical history. 36 hours following admission he developed widespread surgical emphysema. An urgent CT scan of his chest and abdomen demonstrated mediastinal gas of lung origin. He also developed bone marrow suppression and disseminated intravascular coagulopathy. He was treated supportively with intravenous fluids, high flow oxygen and intravenous antibiotics and made a full recovery. Colchicine toxicity is a rare, but important presentation with high levels of morbidity and mortality. Pneumomediastinum is a potentially important complication. It may be appropriate to monitor patients in the later stages of the condition through an ambulatory setting.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Colchicine/toxicity , Drug Overdose , Fluid Therapy/methods , Mediastinal Emphysema , Oxygen Inhalation Therapy/methods , Drug Overdose/complications , Drug Overdose/diagnosis , Drug Overdose/physiopathology , Drug Overdose/therapy , Humans , Male , Mediastinal Emphysema/diagnostic imaging , Mediastinal Emphysema/etiology , Mediastinal Emphysema/physiopathology , Mediastinal Emphysema/therapy , Subcutaneous Emphysema/etiology , Subcutaneous Emphysema/physiopathology , Subcutaneous Emphysema/therapy , Tomography, X-Ray Computed/methods , Treatment Outcome , Young Adult
16.
Toxins (Basel) ; 16(1)2024 01 09.
Article in English | MEDLINE | ID: mdl-38251251

ABSTRACT

Colchicine, a natural compound extracted from Colchicum autumnale, is a phytotoxin, but interestingly, it also has multiple pharmacological activities. Clinically, colchicine is widely used for the treatment of gouty arthritis, familial Mediterranean fever, cardiovascular dysfunction and new coronary pneumonia. However, overdose intake of colchicine could cause lethal liver damage, which is a limitation of its application. Therefore, exploring the potential mechanism of colchicine-induced hepatotoxicity is meaningful. Interestingly, it was found that CYP1A1 played an important role in the hepatotoxicity of colchicine, while it might also participate in its metabolism. Inhibition of CYP1A1 could alleviate oxidative stress and pyroptosis in the liver upon colchicine treatment. By regulating CYP1A1 through the CASPASE-1-GSDMD pathway, colchicine-induced liver injury was effectively relieved in a mouse model. In summary, we concluded that CYP1A1 may be a potential target, and the inhibition of CYP1A1 alleviates colchicine-induced liver injury through pyroptosis regulated by the CASPASE-1-GSDMD pathway.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Colchicine , Animals , Mice , Colchicine/toxicity , Cytochrome P-450 CYP1A1/genetics , Oxidative Stress , Caspase 1
17.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700005

ABSTRACT

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.


Subject(s)
Autophagy , Cardiotoxicity , Colchicine , Doxorubicin , Lysosomes , Myocytes, Cardiac , Colchicine/toxicity , Colchicine/pharmacology , Doxorubicin/toxicity , Cardiotoxicity/prevention & control , Autophagy/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Male , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/metabolism , Antibiotics, Antineoplastic/toxicity , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Ventricular Function, Left/drug effects
18.
Neurobiol Learn Mem ; 106: 112-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23880567

ABSTRACT

The aim of this study was to determine the role of the dorsal dentate gyrus (dDG) in object recognition memory using a black box and object-context recognition memory using a clear box with available cues that define a spatial context. Based on a 10 min retention interval between the study phase and the test phase, the results indicated that dDG lesioned rats are impaired when compared to controls in the object-context recognition test in the clear box. However, there were no reliable differences between the dDG lesioned rats and the control group for the object recognition test in the black box. Even though the dDG lesioned rats were more active in object exploration, the habituation gradients did not differ. These results suggest that the dentate gyrus lesioned rats are clearly impaired when there is an important contribution of context. Furthermore, based on a 24 h retention interval in the black box the dDG lesioned rats were impaired compared to controls.


Subject(s)
Dentate Gyrus/physiology , Recognition, Psychology/physiology , Animals , Colchicine/toxicity , Cues , Dentate Gyrus/drug effects , Rats , Rats, Long-Evans , Recognition, Psychology/drug effects , Space Perception/drug effects , Space Perception/physiology , Spatial Behavior/drug effects , Spatial Behavior/physiology , Time Factors
19.
Virol J ; 10: 121, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23597412

ABSTRACT

BACKGROUND: It is generally thought that viruses require the cytoskeleton during their replication cycle. However, recent experiments in our laboratory with rubella virus, a member of the family Togaviridae (genus rubivirus), revealed that replication proceeded in the presence of drugs that inhibit microtubules. This study was done to expand on this observation. FINDINGS: The replication of three diverse viruses, Sindbis virus (SINV; family Togaviridae family), vesicular stomatitis virus (VSV; family Rhabdoviridae), and Herpes simplex virus (family Herpesviridae), was quantified by the titer (plaque forming units/ml; pfu/ml) produced in cells treated with one of three anti-microtubule drugs (colchicine, noscapine, or paclitaxel) or the anti-actin filament drug, cytochalasin D. None of these drugs affected the replication these viruses. Specific steps in the SINV infection cycle were examined during drug treatment to determine if alterations in specific steps in the virus replication cycle in the absence of a functional cytoskeletal system could be detected, i.e. redistribution of viral proteins and replication complexes or increases/decreases in their abundance. These investigations revealed that the observable impacts were a colchicine-mediated fragmentation of the Golgi apparatus and concomitant intracellular redistribution of the virion structural proteins, along with a reduction in viral genome and sub-genome RNA levels, but not double-stranded RNA or protein levels. CONCLUSIONS: The failure of poisons affecting the cytoskeleton to inhibit the replication of a diverse set of viruses strongly suggests that viruses do not require a functional cytoskeletal system for replication, either because they do not utilize it or are able to utilize alternate pathways when it is not available.


Subject(s)
Cytoskeleton/metabolism , Herpesvirus 1, Human/physiology , Sindbis Virus/physiology , Vesiculovirus/physiology , Virus Replication , Animals , Cell Line , Colchicine/toxicity , Cytoskeleton/drug effects , Humans , Noscapine/toxicity , Paclitaxel/toxicity , Viral Load , Viral Plaque Assay
20.
Mutat Res ; 755(1): 73-80, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23726961

ABSTRACT

When characterizing the genotoxicity of chemicals that induce micronuclei, it is practical to be able to classify the chemicals as aneugens or clastogens. This classification gives information on the mechanistic properties of chemicals and is indispensable for setting the threshold safety margins for genotoxicity in pharmaceutical development. A widely used method for detecting aneugens is fluorescence in situ hybridization (FISH) but, even though the rat is an experimental animal generally used in preclinical studies in drug development, DNA probes that hybridize to all the centromeres of rat chromosomes have not yet been established. In the present study, in addition to the previously known satellite I sequence, we identified two novel satellite sequences, satellite II and satellite III, from the rat genome database. DNA probes with a mixture of these satellite DNA sequences were used to establish a FISH method for pan-centromeric staining of rat chromosomes. To confirm the feasibility of the method, vinblastine (VBS) and mitomycin C (MMC) were administered to rats as a typical aneugen and clastogen, respectively. Micronucleated polychromatic erythrocytes (MNPCE) from bone marrow were enriched by sorting in flow cytometry and subjected to the FISH method. As a result, the ratio of centromere-positive MNPCE increased in VBS-treated rats but not in MMC-treated ones. Since the FISH method using the novel DNA probes clearly discriminates the aneugens from the clastogens, we suggest this method as a useful tool for providing mechanistic information for micronucleus induction in vivo.


Subject(s)
Aneugens/toxicity , Bone Marrow/drug effects , Centromere/drug effects , DNA Probes , In Situ Hybridization, Fluorescence , Micronuclei, Chromosome-Defective/drug effects , Mutagens/toxicity , Animals , Antibiotics, Antineoplastic/toxicity , Base Sequence , Centromere/genetics , Chromosomes/genetics , Colchicine/toxicity , DNA, Satellite/analysis , DNA, Satellite/genetics , Erythrocytes/drug effects , Flow Cytometry , Male , Micronucleus Tests , Mitomycin/toxicity , Molecular Sequence Data , Rats , Sequence Homology, Nucleic Acid , Tubulin Modulators/toxicity , Vinblastine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL