Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
1.
Environ Sci Pollut Res Int ; 29(42): 64225-64231, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35896873

RESUMO

With their wide application in chemical industry, human health and environmental toxic effects of hydrazines arise extensive concerns. Although hydrazine exposure is known to lead to inhibition of seed germination and seedling growth in plants, there are few reports about the mechanism of oxidation or transformation ways of hydrazines in plant tissue. In this research, garden cress (Lepidium sativum L.) and zucchini (Cucurbita pepo L.) were used as model plant objects to study 1,1-dimethylhydrazine exposure in vitro using the GC-MS system. The seed germination and plant growth experiments were carried out in Petri dishes using an aqueous media. Among the detected 1,1-dimethylhydrazine transformation products in plant tissues, 1-methyl-1H-1,2,4-triazole had the highest intensity and reproducibility. In our research, 1-methyl-1H-1,2,4-triazole formed at all contaminant concentrations in an extremely short period. This preliminary study determined 1,1-dimethylhydrazine environment contamination by detecting 1-methyl-1H-1,2,4-triazole in combination with other transformation products.


Assuntos
Dimetilidrazinas , Lepidium sativum , Dimetilidrazinas/farmacologia , Humanos , Reprodutibilidade dos Testes , Triazóis
2.
J Toxicol Environ Health A ; 84(14): 582-592, 2021 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-33825664

RESUMO

Styrax camporum Pohl, a typical species from the Brazilian cerrado, commonly known as "benjoeiro", is used to treat gastroduodenal diseases. In previous studies carried out by our research group, hydroalcoholic extract of S. camporum stems (SCHE) exhibited antigenotoxic and antiproliferative effects. For a comparative analysis of the chemopreventive effect of SCHE, the aim of this study was to investigate the influence of SCHE against carcinogen 1,2-dimethylhydrazine (DMH)-induced DNA damage and pre-neoplastic lesions in Wistar rat colon. Animals were treated orally with SCHE at 250, 500 or 1000 mg/kg body weight in conjunction with a subcutaneous injection of DMH. DNA damage was assessed using the comet assay while tpre-neoplastic lesions by aberrant crypt foci (ACF) assay. The following hepatic oxidative stress markers were determined including activities of catalase (CAT) and glutathione S-transferase (GST) as well as levels of reduced glutathione (GSH) and malondialdehyde (MDA). Treatment with SCHE was not genotoxic or carcinogenic at the highest dose tested (1000 mg/kg b.w.). The extract effectively inhibited DNA damage and pre-neoplastic lesions induced by DMH administration at all concentrations tested. Measurement of CAT, and GST activities and levels of GSH showed that SCHE did not reduce oxidative processes. In contrast, treatment with SCHE (1000 mg/kg b.w.) decreased liver MDA levels. Taken together, these findings suggested the chemopreventive effect attributed to SCHE in colon carcinogenesis, may be related to its capacity to inhibit DNA damage as well as an antioxidant action associated with its chemical constituents egonol and homoegonol.


Assuntos
Anticarcinógenos/farmacologia , Dano ao DNA/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Styrax/química , Animais , Carcinógenos/farmacologia , Carcinógenos/toxicidade , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Ensaio Cometa , Dimetilidrazinas/farmacologia , Dimetilidrazinas/toxicidade , Masculino , Extratos Vegetais/química , Caules de Planta/química , Substâncias Protetoras/química , Ratos , Ratos Wistar
3.
FEBS Open Bio ; 9(12): 2117-2125, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31665829

RESUMO

Colorectal cancer (CRC) is the third most common tumor worldwide, and recent epidemiological studies have indicated that obesity contributes to the morbidity and mortality of CRC. Furthermore, obesity-related adipokines have been shown to be closely related to the incidence of CRC, but the underlying mechanisms are unclear. Here, we investigated the effects of high-fat diet-induced adipokines and cytokines on the development of CRC in vitro and in vivo. For the in vivo assays, we divided 2-week-old C57BL/6J-ApcMin/J male mice into three groups: normal-fat diet (ND), high-fat and high-sugar feed (HFHS), and high-fat and low-sugar feed (HFLS). After 1 week, all mice were injected with 20 mg·kg-1 1,2-dimethylhydrazine once weekly for 10 consecutive weeks. Body weight, liver weight, epididymal fat weight and blood glucose levels were greatly increased in HFHS and HFLS groups compared with the ND group, and the expression levels of some adipokines and cytokines were obviously higher in HFHS or HFLS mice compared with ND mice. For the in vitro assays, HCT116 CRC cells were treated with sera of ND, HFHS or HFLS groups, or serum-free media as a negative control. We observed that sera derived from HFHS or HFLS mice that contain excess adipokines and cytokines promoted the proliferation, migration and invasion of HCT116 cells compared with the ND sera-conditioned medium or serum-free medium group. Therefore, high-fat diet-induced adipokines and cytokines may promote the progression of CRC in vivo and in vitro.


Assuntos
Neoplasias Colorretais/etiologia , Neoplasias Colorretais/metabolismo , Dieta Hiperlipídica/efeitos adversos , Adipocinas/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Dimetilidrazinas/farmacologia , Progressão da Doença , Masculino , Camundongos , Invasividade Neoplásica , Obesidade/metabolismo , beta-Glucanas
4.
J Environ Pathol Toxicol Oncol ; 37(2): 93-101, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30055545

RESUMO

The combination approach is now a well-established treatment for cancer. The present study evaluated the potential of curcumin and resveratrol on p53 post-translational modifications during gastric cancer. We segregated rats into five groups that included normal controls, dimethylhydrazine (DMH) treated, DMH + curcumin treated, DMH + resveratrol treated, and DMH + curcumin + resveratrol treated. Morphological analyses of tumor nodules confirmed carcinogenesis in rats after 25 weeks of DMH administration. The DMH treatment significantly induced carcinogenesis, as evidenced by high tumor burden in DMH-treated rats compared with controls. Moreover, DMH treatment caused a significant increase in the protein expressions of p53 as well as p53 phosphorylation in the DMH-treated rats. In addition, a significant rise was observed in 14C glucose uptake and 3H-thymidin uptakes in DMH-treated rats. Furthermore, enzyme activities of lactate dehydrogenase and alkaline phosphatase also showed a significant rise. On the contrary, significant decline was noticed in the p53 acetylation at residue 382 of DMH-treated rats. Conversely, combined treatment with curcumin and resveratrol to DMH-treated rats resulted in significant moderation in the tumor burden. In addition, a significant rise in p53 acetylation was at residue 382 of DMH-treated rats after treatment with phytochemicals. Supplementation with phytochemicals significantly modulated other biophysical and biochemical indices to near normal levels. Therefore, we conclude that curcumin and resveratrol significantly modulated p53 post-translational modifications during gastric cancer.


Assuntos
Antineoplásicos/farmacologia , Curcumina/farmacologia , Processamento de Proteína Pós-Traducional , Estilbenos/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Proteína Supressora de Tumor p53/genética , Acetilação , Animais , Antineoplásicos Fitogênicos/farmacologia , Carcinogênese , Dimetilidrazinas/farmacologia , Combinação de Medicamentos , Masculino , Fosforilação , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Resveratrol , Neoplasias Gástricas/etiologia , Proteína Supressora de Tumor p53/metabolismo
5.
Biomed Pharmacother ; 100: 282-295, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29448205

RESUMO

In this study, we investigated the in vivo antiproliferative activity of 6,7-dimethoxy-1,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid (M1) in dimethylhydrazine (DMH) induced colorectal carcinoma (CRC) using albino Wistar rats. M1 was administered to DMH induced CRC rats at 10 and 25 mg/kg doses for 15 days. Various physiological, oxidative parameters, histopathology, ELISA, gene and protein expression studies were conducted to evaluate the anti-CRC potential of M1. The histopathology and biochemical tests indicated the protective action of M1 in DMH-induced colon cancer. ELISA confirms that M1 reduced the increased concentration of IL-6 more prominently than those of IL-2 and COX-2. Gene expression analysis revealed that M1 attenuated the increased mRNA over-expression of IL-6, JAK2 and STAT3. The result obtained from quantitative western blot analysis demonstrated that the CRC condition was produced by the IL-6 induced activation/phosphorylation of JAK2 and STAT3 and further down-regulated with M1 treatment. This evidence was supported well with the application of data-based mathematical modeling. Applying the fitted model, we predicted the quantitative behavior of STAT3 populations not accessible to experimental measurement. Later, 1H NMR based serum metabolic profiling was carried out using rat sera to investigate the impact of M1 on CRC-induced metabolic alterations. M1 showed its ability to restore the perturbed metabolites in CRC condition. Altogether, our study provided the first time evidence that M1 exhibits anti-CRC potential through the blockade of IL-6/JAK2/STAT3 oncogenic signaling.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Interleucina-6/antagonistas & inibidores , Tetra-Hidroisoquinolinas/uso terapêutico , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/isolamento & purificação , Proliferação de Células/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Dimetilidrazinas/farmacologia , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Interleucina-6/genética , Masculino , Modelos Teóricos , Mucuna/química , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar , Transdução de Sinais , Tetra-Hidroisoquinolinas/administração & dosagem , Tetra-Hidroisoquinolinas/isolamento & purificação
6.
Eur J Pharmacol ; 818: 174-183, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29074413

RESUMO

The present study reveals the effect of galantamine (GAL) against 1, 2-dimethylhydrazine (DMH) induced colon cancer. Wistar albino rats were arbitrarily divided into four groups (n = 8). Group 1 served as normal control (normal saline, 3ml/kg/day, p.o.); group 2, 3 and 4 received DMH (20mg/kg/week, s.c.), for 6 weeks; groups 3 and 4 also received GAL (2 and 4mg/kg/day, p.o) for 6 weeks. DMH treated rats showed decreased heart rate variability (HRV) factors, increased incidence of aberrant crypt foci (ACF), increased thiobarbituric acid reactive substances (TBARs) along with the decrease in the enzymatic activity of superoxide dismutase (SOD) and catalase. Increased levels of inflammatory marker cyclooxygenase (COX) and lipoxygenase (LOX) was also evident in DMH treated animals. The colonic surface architecture was studied using scanning electron microscopy revealed aberrant crypts(X500) and neoplastic nodules (X2000). GAL treatment helped to minimize the ACF count, restored oxidative stress and inflammatory markers favorably. To further validate our results, our study was directed to define the effect of GAL on acetylcholine neurotransmission using a simple model organism, Caenorhabditis elegans (C. elegans). Increased synaptic cholinergic transmission by GAL (32µM) was evident in the worms when studied through aldicarb assay. However, GAL (32µM) treatment negatively modulated α7 nicotinic acetylcholine receptor (α7nAch receptor), when evaluated using the levamisole assay. GAL (32µM) treatment down regulated the genomic expression of ace-1, ace-2 along with unc-29, unc-38, and unc-50 (essential components of α7 nAch receptor). GAL by inhibiting AchE and regulating Alpha7nACh activity can improve cholinergic neurotransmission.


Assuntos
Acetilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Neoplasias do Colo/patologia , Dimetilidrazinas/farmacologia , Galantamina/farmacologia , Receptores Nicotínicos/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Concentração de Íons de Hidrogênio , Estresse Oxidativo/efeitos dos fármacos , Ratos Wistar
7.
Environ Mol Mutagen ; 52(4): 269-79, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20872852

RESUMO

The in vivo bone marrow (BM) micronucleus assay is one of the three tests in the standard test battery to assess the genotoxic potential of a pharmaceutical candidate. In some cases, depending on results of in vitro studies, the route of administration or the degree of systemic exposure, in vivo assessment of genotoxicity in the BM alone may not be sufficient. Based on the potential for high gut exposures to orally administered compounds with low systemic exposures as well as the potential susceptibility of rapidly dividing cells of the intestinal tissues, we have developed a modified technique for evaluating micronuclei formation in both the duodenum and colon of rats based on earlier publications. Adult male Sprague Dawley rats were treated once daily for 2 days with either vehicle control or with the test articles acetyl salicylic acid (ASA), carbendazim (CAR), cyclophosphamide (CP), dimethylhydrazine (DMH), mitomycin C (MMC) or vinblastine sulfate (VIN). The duodenum, colon, and BM were harvested, processed, and analyzed for micronucleus induction. Results from these studies demonstrated differences in the susceptibility for different test compounds in the three tissues tested. When MMC and VIN were dosed by different routes at the same dose levels both compounds produced positive results in all three tissues by intraperitoneal injection but not oral administration. These studies suggest that overall the GI micronucleus assay might be a useful tool for clastogenic and aneugenic compounds that are expected to produce high sustained concentrations in the gastrointestinal tract with little systemic exposure.


Assuntos
Colo/efeitos dos fármacos , Duodeno/efeitos dos fármacos , Testes para Micronúcleos/métodos , Animais , Aspirina/efeitos adversos , Aspirina/farmacologia , Benzimidazóis/efeitos adversos , Benzimidazóis/farmacologia , Carbamatos/efeitos adversos , Carbamatos/farmacologia , Ciclofosfamida/efeitos adversos , Ciclofosfamida/farmacologia , Dano ao DNA/efeitos dos fármacos , Dimetilidrazinas/efeitos adversos , Dimetilidrazinas/farmacologia , Masculino , Mitomicina/efeitos adversos , Mitomicina/farmacologia , Testes de Mutagenicidade/métodos , Mutagênicos/efeitos adversos , Mutagênicos/farmacologia , Ratos , Ratos Sprague-Dawley , Vimblastina/efeitos adversos , Vimblastina/farmacologia
8.
Aviakosm Ekolog Med ; 42(1): 32-5, 2008.
Artigo em Russo | MEDLINE | ID: mdl-18564567

RESUMO

Asymmetric dimethyl hydrazine (ADH), a highly toxic propellant component, is known to cause metabolic disturbances in humans and laboratory animals. Focus of this investigation was placed on crystal-forming properties of blood serum, and types of plasma protein destructions in ADH-intoxicated rats. Object of investigation was blood plasma collected from rats exposed to 100 mg/kg of per oral ADH total (5 mg/kg per a day, 5 days a wk, 4 wks.). The crystal-forming ability of plasma was assessed with the teziographic technique. Severity of proteins destruction was determined by the content of carbonyl products of oxidative protein modification in plasma. Chronic exposure to low ADH doses caused oxidative destruction of proteins in blood plasma and, consequently, significant degradation of the plasma ability to form crystals reflected in an altered zonal phase structure and phase bursting on teziograms. Symmetry gave way to pathological structures, e.g. petals and loops. Amorphous areas grew in size, bursting lines thinned out. Concretions became less numerous. These data suggest a close relationship between the impairments in physical and chemical properties of blood plasma of experimental animals by highly toxic asymmetric dimethyl hydrazine.


Assuntos
Proteínas Sanguíneas/efeitos dos fármacos , Dimetilidrazinas/farmacologia , Plasma/efeitos dos fármacos , Animais , Dimetilidrazinas/administração & dosagem , Feminino , Masculino , Modelos Animais , Ratos
9.
Mol Endocrinol ; 22(4): 868-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18096695

RESUMO

The orphan nuclear receptor pregnane X receptor (PXR) plays an important role in the detoxification of foreign and endogenous chemicals, including bile acids. PXR promotes bile acid elimination by activating bile acid-detoxifying enzymes and transporters. Certain bile acids are known to promote colonic carcinogenesis by inducing colon cancer cell apoptosis. However, whether and how PXR plays a role in colon cancer apoptosis has not been reported. In this study, we showed that activation of PXR by genetic (using a constitutively activated PXR) or pharmacological (using PXR agonist rifampicin) means protected the PXR-overexpressing colon cancer HCT116 cells from deoxycholic acid-induced apoptosis. Interestingly, activation of PXR also protected HCT116 cells from adriamycin-induced cell death, suggesting that the antiapoptotic effect of PXR was not bile acid specific. Moreover, the antiapoptotic effect of PXR in HCT116 cells appeared to be independent of xenobiotic enzyme regulation, because these cells had little basal and inducible expression of bile acid-detoxifying enzymes. Instead, SuperArray analysis showed that PXR-mediated deoxycholic acid resistance was associated with up-regulation of multiple antiapoptotic genes, including BAG3, BIRC2, and MCL-1, and down-regulation of proapoptotic genes, such as BAK1 and TP53/p53. Treatment with rifampicin in colon cancer LS180 cells, a cell line known to express endogenous PXR, also inhibited apoptosis. Activation of PXR in transgenic mice inhibited bile acid-induced colonic epithelial apoptosis and sensitized mice to dimethylhydrazine-induced colonic carcinogenesis, suggesting that the antiapoptotic effect of PXR is conserved in normal colon epithelium. In summary, our results have established the antiapoptotic role of PXR in both human colon cancer cells and normal mouse colon epithelium.


Assuntos
Apoptose/fisiologia , Receptores de Esteroides/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Western Blotting , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ácido Desoxicólico/farmacologia , Dimetilidrazinas/farmacologia , Doxorrubicina/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Citometria de Fluxo , Células HCT116 , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Camundongos Transgênicos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Receptor de Pregnano X , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rifampina/farmacologia , Ubiquitina-Proteína Ligases , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
10.
Zhongguo Zhong Yao Za Zhi ; 33(22): 2653-7, 2008 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-19216165

RESUMO

OBJECTIVE: : To profile urinary metabolite variations from 1, 2-dimethylhydrazine (DMH)-induced precancerous colon rats, Jinfu Kang treated rats and healthy controls. METHOD: We used ethyl chloroformate derivatization and gas chromatography-mass spectrometry (GC-MS) based metabonomic method to analyze rat urines. RESULT: The time-dependent variations of metabolite profile showed a progressive deviation of the metabolism in the model group from the initial pattern over time and a systemic recovery of the metabolism in the treatment group, which is consistent with the histological results. The in-depth analysis indicated that the disorder of tricarboxylic acid cycle (TCA), tryptophan metabolism, polyamine metabolism and gut flora structure were associated with DMH intervention. CONCLUSION: Metabolic study revealed that Jinfu Kang can effectively reverse metabolic departures in DMH-induced precancerous colon rat, which is consistent with pathological results.


Assuntos
Neoplasias do Colo/patologia , Pólipos do Colo/tratamento farmacológico , Pólipos do Colo/urina , Medicamentos de Ervas Chinesas/farmacologia , Animais , Neoplasias do Colo/induzido quimicamente , Pólipos do Colo/induzido quimicamente , Dimetilidrazinas/farmacologia , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Ratos , Ratos Wistar
11.
Biol Trace Elem Res ; 103(2): 133-45, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15772437

RESUMO

Cell culture studies have suggested that arsenic exposure results in decreased S-adenosylmethionine (SAM), causing DNA hypomethylation. Previously, we have shown that hepatic SAM is decreased and/or S-adenosylhomocysteine increased in arsenic-deprived rats; these rats tended to have hypomethylated DNA. To determine the effect of dietary arsenic on dimethylhydrazine (DMH)-induced aberrant crypt formation in the colon, Fisher 344 weanling male rats were fed diets containing 0, 0.5, or 50 microg As (as NaAsO2)/g. After 12 wk, dietary arsenic affected the number of aberrant crypts (p<0.02) and aberrant crypt foci (p<0.007) in the colon and the amount of global DNA methylation (p<0.04) and activity of DNA methyltransferase (DNMT) (p<0.003) in the liver. In each case, there were more aberrant crypts and aberrant crypt foci, a relative DNA hypomethylation, and increased activity of DNMT in the rats fed 50 microg As/g compared to those fed 0.5 microg As/g. The same phenomenon, an increased number of aberrant crypts and aberrant crypt foci, DNA hypomethylation, and increased DNMT tended to hold when comparing rats fed the diet containing no supplemental arsenic compared to rats fed 0.5 microg As/g. The data suggest that there is a threshold for As toxicity and that possibly too little dietary As could also be detrimental.


Assuntos
Arsênio/toxicidade , Colo/efeitos dos fármacos , Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Dieta , Dimetilidrazinas/farmacologia , Fígado/efeitos dos fármacos , Animais , Arsênio/administração & dosagem , Carcinógenos/toxicidade , Colo/anatomia & histologia , Humanos , Fígado/fisiologia , Masculino , Ratos , Ratos Endogâmicos F344
12.
Indian J Exp Biol ; 42(6): 595-600, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15260111

RESUMO

Effect of prefeeding dehydrated amaranth (A. gangeticus) leaves at 10 and 20% levels on a chemical toxicant, dimethylhydrazine (DMH)-induced free radical stress in rat liver was evaluated. DMH-induced rise in hepatic malondialdehyde (MDA), was diminished by AL. AL intake resulted in a significant increase in hepatic glutathione (GSH). The feeding of AL at 10% level increased the hepatic glucose-6-phosphate dehydrogenase (G-6-PDH) activity, while that at 20% level increased the hepatic glutathione reductase (GSSGR) as well, in addition to G-6-PDH. Amaranth leaves at 10 and 20% levels of feeding diminished the hepatic superoxide dismutase and glutathione peroxidase (GSH-Px) activities. DMH influenced adversely the hepatic antioxidant enzyme activities. Simultaneous administration of DMH and feeding of AL enhanced the DMH-induced decrease in hepatic GSH-Px. DMH enhanced formation of micronuclei was reverted significantly by AL intake. Hence, it was concluded that the consumption of AL at 20% level reduced DMH-induced impaired antioxidant status in rat liver.


Assuntos
Amaranthus/metabolismo , Antioxidantes/farmacologia , Dimetilidrazinas/farmacologia , Fígado/metabolismo , Extratos Vegetais/farmacologia , Animais , Antioxidantes/metabolismo , Peso Corporal , Medula Óssea/metabolismo , Colo/metabolismo , Radicais Livres , Glucosefosfato Desidrogenase/metabolismo , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Peroxidação de Lipídeos , Fígado/enzimologia , Masculino , Malondialdeído/farmacologia , Micronúcleos com Defeito Cromossômico/metabolismo , Tamanho do Órgão , Estresse Oxidativo , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo
14.
Biofactors ; 15(1): 11-26, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11673641

RESUMO

Low dietary copper has been shown to decrease the expression of various protein kinase C (PKC) isozymes and increase the risk of colon cancer development in experimental animals. The purpose of this study was to investigate the relationship between dietary copper and carcinogen administration on PKC isozyme accumulation and aberrant crypt foci (ACF) formation in rats fed 0.9 and 7.7 microg Cu/g diet. After 24 and 31 d on the diets, the rats were injected with either dimethylhydrazine (DMH) (25 mg/kg i.p.) or saline and killed at two time points (2 wk and 8 wk after DMH). Rats fed low dietary copper had significantly lower (p<0.0001) hematocrits, hemoglobin, ceruloplasmin activity and plasma and liver copper concentrations than rats fed adequate dietary copper. Ingestion of low dietary copper significantly (p<0.005) increased the formation of DMH-induced ACF (116.8 vs 59.6). Low dietary copper significantly (p<0.05) decreased the concentration of PKC alpha, delta, and zeta in the colon at 2 wk but not at 8 wk. Thus, changes in PKC isoform protein concentration may be related to increased susceptibility of copper-deficient animals to colon cancer.


Assuntos
Carcinógenos/farmacologia , Colo/efeitos dos fármacos , Cobre/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Animais , Colo/enzimologia , Colo/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Cobre/análise , Cobre/deficiência , Dieta , Dimetilidrazinas/farmacologia , Isoenzimas/genética , Fígado/química , Masculino , Proteína Quinase C/genética , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Oncol Rep ; 6(4): 883-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10373675

RESUMO

We studied the tissue-specific expression of the p53 gene in different parts of the intestine of mice treated with low doses of a carcinogen and exposed to different p53 antibodies. The human p53 promoter-CAT transgenic mice were immunized with different p53 antibodies (monoclonal - PAb 421 and DO1, and polyclonal - H-p53 and anti-soluble p53 IgG) and then exposed to low doses of dimethylhydrazine (DMH). Enzymatic CAT activity was determined in the ileum and colon 8 weeks later after the final injection of DMH. Expression of the p53 transgene in the normal ileum was twice as high as in the colon. Treatment with DMH significantly decreased the expression of the p53 transgene both in the ileum (from 18% to 100%) and in the colon (from 10% to 52%). Vaccination of mice protected at least in part such a decrease. The most effective results were found after exposure of mice to polyclonal H-p53 and to a lesser extent to anti-p53 IgG. No difference was found in the effects of antibodies on the small and large intestines. We concluded that polyclonal antibodies were more effective than monoclonal ones in protection against anti-p53 action of DMH. The observation of these effects may make it possible to explain the higher antitumor activity of polyclonal antibodies.


Assuntos
Carcinógenos/farmacologia , Dimetilidrazinas/farmacologia , Genes Supressores de Tumor , Intestinos/efeitos dos fármacos , Proteína Supressora de Tumor p53/biossíntese , Animais , Anticorpos/farmacologia , Genes Supressores de Tumor/efeitos dos fármacos , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia
16.
Am J Pathol ; 151(3): 843-52, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9284833

RESUMO

Crypt fission is now established as an important mechanism of intestinal growth and regeneration. It has been proposed that increased crypt size is the stimulus for crypt fission, because crypts preparing for fission are generally larger. Consequently, we investigated the effects of epidermal growth factor (EGF) and dimethylhydrazine, which are both known to stimulate crypt cell proliferation, on crypt fission in the rat intestine. We also examined whether the effects of EGF on both proliferation and crypt fission are modified by the pretreatment with dimethylhydrazine for 16 weeks, dimethylhydrazine was then discontinued for 8 weeks, followed by intravenous infusion of EGF for 1 week. There were four groups: vehicle alone, EGF alone, dimethylhydrazine alone, and dimethylhydrazine followed by EGF infusion. The rats were killed at 25 weeks and rates of intestinal crypt cell production, crypt size, and crypt fission were determined. Intravenously infused EGF significantly increased crypt cell production rate, but the magnitude of the effect decreased from the proximal to the distal colon. EGF caused an increase in crypt area, possibly reflecting an increase in crypt size. Importantly dimethylhydrazine had no significant effect on crypt cell production rate nor on crypt area in the distal colon, but it did cause an increase in crypt area in the mid-colon. The crypt fission index was significantly decreased by EGF and increased by dimethylhydrazine. There was no qualitative interaction between EGF and dimethylhydrazine. These results demonstrate the marked proliferative effect of intravenously infused EGF in the colon of orally fed rats, with significant site effects (P = 0.0007); the effect was greatest in the proximal colon and disappeared in the distal colon. The observation that EGF reduced crypt fission indicates that increased cell proliferation, per se, is not a stimulus for crypt fission. This is further supported by the observation that dimethylhydrazine increases crypt fission in crypts of normal size in the distal colon without significantly increasing cell proliferation. These results suggest that increasing crypt cellularity by proliferation is not sufficient to induce crypt fission, and factors other than increased crypt size by proliferation can control crypt fission. It is also probable that cell proliferation and crypt fission are independently regulated. Crypt fission appears to play a considerable role in the intestinal response to carcinogens.


Assuntos
Colo/patologia , Dimetilidrazinas/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Mucosa Intestinal/patologia , Animais , Divisão Celular/efeitos dos fármacos , Colo/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Wistar
17.
Biochem Biophys Res Commun ; 234(1): 44-7, 1997 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-9168957

RESUMO

Activation of 1,2-dimethylhydrazine (DMH) by prolonged auto-oxidation (24-h) induced proliferation of mouse fibroblasts at low hydrazine concentrations (0.1-1.0 mM) as determined by [3H-methyl]-thymidine uptake of confluent quiescent cells. Incubations were performed under conditions in which alkyl radicals are slowly formed by DMH auto-oxidation. The proliferative stimulus induced by DMH auto-oxidation complements that induced by insulin, PMA, and EGF. Inhibition by the iron chelators, o-phenanthroline and desferrioxamine, demonstrates that the induction of the proliferative effect is dependent on simple iron complexes. Proliferation was also inhibited by superoxide dismutase, catalase, and mannitol, implicating reactive oxygen species, although superoxide dismutase and catalase also inhibited alkyl radical formation, as determined by spin-trapping. These results suggest that cell proliferation induced by DMH auto-oxidation is mediated by reactive oxygen species, mainly the hydroxyl radical, and is dependent on simple iron complexes, possibly involving the Fenton reaction.


Assuntos
Divisão Celular/efeitos dos fármacos , Dimetilidrazinas/farmacologia , Ferro/metabolismo , 1,2-Dimetilidrazina , Animais , Carcinógenos/farmacologia , Catalase/metabolismo , Catalase/farmacologia , Linhagem Celular , Quelantes/farmacologia , DNA/biossíntese , Dimetilidrazinas/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Sequestradores de Radicais Livres/farmacologia , Radicais Livres , Insulina/farmacologia , Manitol/farmacologia , Camundongos , Mutagênicos/farmacologia , Oxirredução , Ácido Pentético/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase/farmacologia
18.
Botucatu; s.n; 1997. 100 p. ilus, tab.
Tese em Português | LILACS, SES-SP | ID: lil-226102

RESUMO

O crescimento neoplásico pode ser estimulado por um trauma cirúrgico e pelo processo reparativo subseqüente. A retirada parcial de uma neoplasia, pode favorecer a proliferaçäo de células tumorais remanescentes. Da mesma forma, a remoçäo de um tumor primário, a cirurgia simulada ou a amputaçäo de um membro sadio influenciam positivamente o crescimento de diferentes tipos de neoplasias. No presente estudo, avaliamos a influência do processo reparativo cutâneo sobre as lesöes pré-neoplásicas induzidas pela 1,2 Dimetilhidrazina (DMH) no cólon de ratos Wistar. Foram utilizados cinquenta animais, divididos em seis grupos. Quatro grupos, constituídos por dez animais cada, receberam injeçöes subcutâneas de DMH (20mg/Kg/semana), durante oito semanas. Dois grupos controles, constituídos por cinco animais cada, receberam injeçöes de soluçäo salina por igual período. Na 9ª semana dois grupos de animais, tratados com DMH e dois controles, sofreram intervençäo cirúrgica para retirada de retalho cutâneo do flanco direito, medindo 16 cm². Os animais foram submetidos à eutanásia na 12ª e 20ª semanas do experimento. O número de focos de lesöes pré-neoplásicas, assim como o número e a área das neoplasias bem ou moderadamente diferenciadas, näo foram influenciadas pelo processo reparativo cutâneo, nos momentos estudados. No entanto, o número e o tamanho dos tumores indiferenciados foi maior, na 12ª semana, no grupo submetido a retirada do retalho cutâneo. Estes resultados demonstram que o processo reparativo cutâneo influenciou positivamente o crescimento de neoplasias colônicas indiferenciadas, induzidas pela 1,2 Dimetilhidrazina. O estímulo induzido foi transitório, sendo detectado no período correspondente a fase de reparaçäo da ferida cutânea.


Assuntos
Animais , Ratos , Masculino , Dimetilidrazinas/farmacologia , Cicatrização , Neoplasias do Colo/cirurgia , Ratos Wistar , Dimetilidrazinas/administração & dosagem , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia
19.
Jpn J Cancer Res ; 87(12): 1199-203, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9045953

RESUMO

Morphological analysis of isolated colonic crypts in rats, postnatally, indicated that the crypts reproduce themselves by a fission mechanism, the division beginning at the crypt base and proceeding upwards until there are two separate crypts. Occasionally, before the separation is complete, a second fission process starts on one or both sides of a bifurcating crypt and a triple-branched or quadruple-branched crypt results. Analysis of isolated aberrant crypt foci (ACF) in rats treated with 1,2-dimethylhydrazine revealed that the development of ACF consisting of multiple crypts is also due to a fission mechanism. Initially, an indentation appears at the base of a single ACF crypt, with subsequent formation of a bifurcation and eventual crypt division.


Assuntos
Colo/patologia , 1,2-Dimetilidrazina , Animais , Colo/efeitos dos fármacos , Colo/ultraestrutura , Dimetilidrazinas/farmacologia , Microscopia Eletrônica de Varredura , Mutagênicos/farmacologia , Ratos , Ratos Endogâmicos F344
20.
Z Gastroenterol ; 34(12): 801-8, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9017896

RESUMO

Weekly administrations of the potent carcinogen 1,2-dimethylhydrazine (DMH) predominantly induce carcinoma of the colon by nearly 100% after six months' treatment in rats. Polyamines, and especially the key enzyme of polyamine de novo synthesis ornithine decarboxylase (ODC) are well-known to play an important role in cell growth and tumor carcinogenesis. Male Wistar rats were s. c.-injected with a single dose of 20 mg DMH/kg b. wt. and five to eight animals were sacrificed 4, 8, 12, 24, 72, 120, 168, and 240 hours after injection of DMH or the basic solution, respectively. Additionally, seven animals were simultaneously treated with the ODC inhibitor alpha-difluoromethylornithine (DFMO) and sacrificed seven days after a single DMH injection. A single s. c.-dosage of the colon carcinogen DMH resulted in dissimilar activation patterns of polyamine metabolism in the various organs studied: in distal and less pronounced in proximal colonic mucosa ODC and putrescine are significantly increased seven days after application of DMH and DNA polymerase after ten days; in small intestinal mucosa ODC activity is significantly elevated after seven days and especially S-adenosylmethionine decarboxylase activity is significantly and prolonged increased between twelve and 72 hours after DMH injection; while spermidine/spermine N1-acetyltransferase activity is significantly elevated in liver after 168 and 240 hours, no changes compared to controls are found in the pancreas. DFMO treatment completely prevents DMH-induced activation of polyamine de novo synthesis and DNA polymerase in colon and small intestine. These data prove completely different and -interestingly-late appearing activation patterns of DMH on intracellular polyamine metabolism in various organ systems and further elucidate the complex metabolic changes following carcinogen treatment.


Assuntos
Carcinógenos/farmacologia , Neoplasias do Colo/induzido quimicamente , Dimetilidrazinas/farmacologia , Ornitina Descarboxilase/metabolismo , Putrescina/metabolismo , Espermidina/metabolismo , Espermina/metabolismo , 1,2-Dimetilidrazina , Acetiltransferases/metabolismo , Adenosilmetionina Descarboxilase/metabolismo , Animais , Neoplasias do Colo/enzimologia , Eflornitina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Masculino , Inibidores da Ornitina Descarboxilase , Ratos , Ratos Wistar , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA