Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 11(11): e0165971, 2016.
Article in English | MEDLINE | ID: mdl-27832139

ABSTRACT

Curcumin is extensively investigated as a good chemo-preventive agent in the development of many cancers and particularly in leukemia, including treatment of chronic myelogenous leukemia and it has been proposed as an adjuvant for leukemia therapies. Human chronic myeloid leukemia cells (K562), were treated with 20 µM of curcumin, and we found that a subpopulation of these cells were arrested and accumulate in the G2/M phase of the cell cycle. Characterization of this cell subpopulation showed that the arrested cells presented nuclear morphology changes resembling those described for mitotic catastrophe. Mitotic cells displayed abnormal chromatin organization, collapse of the mitotic spindle and abnormal chromosome segregation. Then, these cells died in an apoptosis dependent manner and showed diminution in the protein levels of BCL-2 and XIAP. Moreover, our results shown that a transient activation of the nuclear factor κB (NFκB) occurred early in these cells, but decreased after 6 h of the treatment, explaining in part the diminution of the anti-apoptotic proteins. Additionally, P73 was translocated to the cell nuclei, because the expression of the C/EBPα, a cognate repressor of the P73 gene, was decreased, suggesting that apoptosis is trigger by elevation of P73 protein levels acting in concert with the diminution of the two anti-apoptotic molecules. In summary, curcumin treatment might produce a P73-dependent apoptotic cell death in chronic myelogenous leukemia cells (K562), which was triggered by mitotic catastrophe, due to sustained BAX and survivin expression and impairment of the anti-apoptotic proteins BCL-2 and XIAP.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , M Phase Cell Cycle Checkpoints/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mitosis/drug effects
2.
Parasit Vectors ; 9: 19, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26767976

ABSTRACT

BACKGROUND: In human hosts, Entamoeba histolytica cysts can develop into trophozoites, suggesting that the life cycle of this parasite are regulated by changes in gene expression. To date, some evidence has suggested that epigenetic mechanisms such as DNA methylation and histone modification are involved in the regulation of gene expression in Entamoeba. Some post-translational modifications (PTMs) at the N-terminus of E. histolytica's histones have been reported experimentally, including tri-methylation in the lysine 4 of histone H3 (H3K4me3) and dimethylation in the lysine 27 of histone H3 (H3K27me2), dimethylation of arginine 3 (H4R3me2) and the indirect acetylation of histone H4 in the N-terminal region. However, it is not known which residues of histone H4 are subject to acetylation and/or methylation or where in the nucleus these epigenetic marks are located. METHODS: Histones from trophozoites of E. histolytica were obtained and analyzed by LC-MS/MS. WB assays were performed using antibodies against epigenetic marks (acetylated lysines and methylated arginines). Immunofluorescence assays (IFA) were carried out to determine the distribution of PTMs and the localization of DNA methylation as a heterochromatin marker. Nuclear bodies such as the nucleolus were identified by using antibodies against fibrillarin and nucleolin and speckles by using anti-PRP6 antibody. RESULTS: Some new PTMs in histone H4 of E. histolytica, such as the acetylation of lysines 5, 8, 12 and 16 and the monomethylation of arginine 3, were identified by WB. IFA demonstrated that some marks are associated with transcriptional activity (such as acetylation and/or methylation) and that these marks are distributed throughout the E. histolytica nucleus. Staining with antibodies against anti-pan-acetylated lysine H4 histone and 5-methyl cytosine showed that the activation and transcriptional repression marks converge. Additionally, two nuclear bodies, the nucleolus and speckles, were identified in this parasite. CONCLUSIONS: This study provides the first evidence that the nucleus of E. histolytica is not compartmentalized and contains two nuclear bodies, the nucleolus and speckles, the latter of which was not identified previously. The challenge is now to understand how these epigenetic marks and nuclear bodies work together to regulate gene expression in E. histolytica.


Subject(s)
Entamoeba histolytica/genetics , Entamoebiasis/microbiology , Epigenesis, Genetic , Acetylation , Arginine , DNA Methylation , Histones/genetics , Humans , Lysine , Tandem Mass Spectrometry
3.
Toxicol Lett ; 241: 19-31, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26589970

ABSTRACT

Cellular senescence is characterized by irreversible cell arrest and is associated with the development of chronic diseases, including cancer. Here, we investigated the induction of cellular senescence during liver carcinogenesis. Liver cancer was induced in Fischer 344 rats with a weekly intraperitoneal injection of diethylnitrosamine (50mg/kg body weight) for 16 weeks. Double-detection of ß-galactosidase with Ki67 for cell proliferation; a-SMA and Pdgfrb for cell specificity; p53, p21, p16, and cyclin D1, CDK2, and CDK4 for senescence-associated molecular pathways and γ-glutamyltranspeptidase (GGT) for hepatocarcinogenesis was assessed to determine the association of these markers with cellular senescence. DNA damage was measured through senescence-associated heterochromatin foci (SAHF) detection. Progressive cellular senescence was observed in both fibrotic septa and hepatocytes from week 10 to 18. The maximum peak of positive senescent and fibrotic cells was observed at week 16 and decreased at week 18, but cell proliferation remained high. Whereas the increased p16 expression and SAHF were concomitant with that of ß-galactosidase, those of p53 and p21 were barely detected. Furthermore, ß-galactosidase positive myofibroblast-like cells were mainly surrounding GGT-positive tumors. Our findings showed that in hepatocarcinogenesis by diethylnitrosamine, cellular senescence is associated with p16 pathway activation and is mainly localized in myofibroblast-like cells.


Subject(s)
Biomarkers, Tumor/analysis , Carcinogenesis/drug effects , Carcinogens/toxicity , Cellular Senescence/drug effects , Diethylnitrosamine/toxicity , Liver Neoplasms/pathology , Neoplasms/enzymology , beta-Galactosidase/metabolism , Animals , Cell Proliferation/drug effects , DNA Damage , Fibrosis , Ki-67 Antigen/metabolism , Liver Neoplasms/chemically induced , Male , Rats , Rats, Inbred F344 , beta-Galactosidase/analysis , gamma-Glutamyltransferase/analysis , gamma-Glutamyltransferase/metabolism
4.
Front Immunol ; 7: 666, 2016.
Article in English | MEDLINE | ID: mdl-28111575

ABSTRACT

Pediatric oncology, notably childhood acute lymphoblastic leukemia (ALL), is currently one of the health-leading concerns worldwide and a biomedical priority. Decreasing overall leukemia mortality in children requires a comprehensive understanding of its pathobiology. It is becoming clear that malignant cell-to-niche intercommunication and microenvironmental signals that control early cell fate decisions are critical for tumor progression. We show here that the mesenchymal stromal cell component of ALL bone marrow (BM) differ from its normal counterpart in a number of functional properties and may have a key role during leukemic development. A decreased proliferation potential, contrasting with the strong ability of producing pro-inflammatory cytokines and an aberrantly loss of CXCL12 and SCF, suggest that leukemic lymphoid niches in ALL BM are unique and may exclude normal hematopoiesis. Cell competence ex vivo assays within tridimensional coculture structures indicated a growth advantage of leukemic precursor cells and their niche remodeling ability by CXCL12 reduction, resulting in leukemic cell progression at the expense of normal niche-associated lymphopoiesis.

5.
Toxicol Sci ; 104(1): 100-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18397917

ABSTRACT

Caffeic acid phenethyl ester (CAPE), a natural component of propolis, shows anticarcinogenic properties in the modified resistant hepatocyte model when administered before initiation or promotion of hepatocarcinogenesis process; however, information about the mechanism underlying this chemoprotection is limited. The aim of this work was to characterize the effect of CAPE on cytochrome P450 (CYP), which is involved in diethylnitrosamine (DEN) metabolism during the initiation stage of chemical hepatocarcinogenesis. Male Fischer-344 rats were treated as in the modified resistant hepatocyte model. Liver samples were obtained at four different times: at 12 h after pretreatment with CAPE and at 12 and 24 h and 25 days after DEN administration. Liver damage was determined by histology with hematoxylin and eosin, measurement of total CYP levels and enzyme activity, and gamma-glutamyl transpeptidase-positive (GGT+) staining of hepatocyte foci. CAPE administration prevented DEN-induced necrosis at 24 h. It also decreased O-dealkylation of 7-ethoxy-resorufin (EROD), O-dealkylation of 7-methoxyresorufin (MROD), and 7-pentoxy-resorufin activities at 12 h after its administration and EROD and MROD activities at 12 h after administration of DEN. CAPE treatment decreased GGT+ foci by 59% on day 25. Our results suggest that CAPE modifies the enzymatic activity of CYP isoforms involved in the activation of DEN, such as CYP1A1/2 and CYP2B1/2. These findings describe an alternative mechanism for understanding the ability of CAPE to protect against chemical hepatocarcinogenesis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Caffeic Acids/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Liver Neoplasms, Experimental/enzymology , Phenylethyl Alcohol/analogs & derivatives , 2-Acetylaminofluorene , Animals , Diethylnitrosamine , Hepatocytes , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/pathology , Male , Microsomes, Liver/enzymology , Phenylethyl Alcohol/pharmacology , Rats , Rats, Inbred F344 , gamma-Glutamyltransferase/metabolism
6.
Mol Carcinog ; 46(7): 524-33, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17393424

ABSTRACT

Cyclohexanol is a basic industrial chemical widely used because of its versatility as an industrial solvent. No studies have been conducted to evaluate the carcinogenic/co-carcinogenic hazards associated with cyclohexanol exposure. In male Fisher 344 rats liver preneoplastic lesions were induced by N-nitrosodiethylamine (150 mg/Kg) i.p., followed by the tumor promoter 2-acetylaminofluorene (2-AAF: 20 mg/kg) orally administered on three consecutive days before partial hepatectomy. The cyclohexanol administration in this hepatocarcinogenesis assay revealed that it has a strong tumor co-promoter potential. There is clear evidence that oxidative stress and the CYP2E1 are components of carcinogenesis. Although no changes in the lipid peroxidation levels were observed between treated and untreated animals, a significant increase in CYP2E1 expression was observed when cyclohexanol was administered 24 h after the last 2-AAF dose. On the other hand, levels of the proliferation markers PCNA and Ki-67 were not increased after treatment with cyclohexanol, but a marked downregulation of the Bax proapoptotic protein was found exclusively in mitochondrial extracts of animals treated with cyclohexanol. This study represents the first report of the ability of cyclohexanol-induced lesions, when administered simultaneously with 2-AAF, to potentiate the development of preneoplastic liver.


Subject(s)
Alkylating Agents/toxicity , Cyclohexanols/toxicity , Diethylnitrosamine/toxicity , Liver Neoplasms, Experimental/chemically induced , Liver/drug effects , Precancerous Conditions/chemically induced , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Disease Models, Animal , Lipid Peroxidation , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred F344
7.
Neoplasia ; 8(5): 373-83, 2006 May.
Article in English | MEDLINE | ID: mdl-16790086

ABSTRACT

In this study, we investigated the time course gene expression profile of preneoplastic nodules and hepatocellular carcinomas (HCC) to define the genes implicated in cancer progression in a resistant hepatocyte model. Tissues that included early nodules (1 month, ENT-1), persistent nodules (5 months, ENT-5), dissected HCC (12 months), and normal livers (NL) from adult rats were analyzed by cDNA arrays including 1185 rat genes. Differential genes were derived in each type of sample (n = 3) by statistical analysis. The relationship between samples was described in a Venn diagram for 290 genes. From these, 72 genes were shared between tissues with nodules and HCC. In addition, 35 genes with statistical significance only in HCC and with extreme ratios were identified. Differential expression of 11 genes was confirmed by comparative reverse transcription-polymerase chain reaction, whereas that of 2 genes was confirmed by immunohistochemistry. Members involved in cytochrome P450 and second-phase metabolism were downregulated, whereas genes involved in glutathione metabolism were upregulated, implicating a possible role of glutathione and oxidative regulation. We provide a gene expression profile related to the progression of nodules into HCC, which contributes to the understanding of liver cancer development and offers the prospect for chemoprevention strategies or early treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Precancerous Conditions/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , DNA, Complementary/metabolism , Disease Progression , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Models, Biological , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Rats , Rats, Inbred F344 , Time Factors
8.
World J Gastroenterol ; 12(12): 1895-904, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16609996

ABSTRACT

AIM: To determine the role of c-Jun N-terminal kinase (JNK) activity in ethanol-induced apoptosis and the modulation of this signaling cascade by S-Adenosyl-methionine (AdoMet). METHODS: Primary hepatocyte cultures were pretreated with 100 micromol/L SP600125, a selective JNK inhibitor, 1 mL/L DMSO or 4 mmol/L AdoMet and then exposed to 100 mmo/L ethanol. Hepatocyte apoptosis was determined by the TUNEL and DNA ladder assays. JNK activity and its inhibition by SP600125 and AdoMet were determined by Western blot analysis of c-jun phosphorylation and Bid fragmentation. SP600125 and AdoMet effects on the apoptotic signaling pathway were determined by Western blot analysis of cytochrome c release and pro-caspase 3 fragmentation. The AdoMet effect on glutathione levels was measured by Ellman's method and reactive oxygen species (ROS) generation by cell cytometry. RESULTS: The exposure of hepatocytes to ethanol induced JNK activation, c-jun phosphorylation, Bid fragmentation, cytochrome c release and pro-caspase 3 cleavage; these effects were diminished by SP600125, and caused a significant decrease in ethanol-induced apoptosis (P< 0.05). AdoMet exerted an antioxidant effect maintaining glutathione levels and decreasing ROS generation, without a significant effect on JNK activity, and prevented cytochrome c release and pro-caspase 3 cleavage. CONCLUSION: The JNK signaling cascade is a key component of the proapoptotic signaling pathway induced by ethanol. JNK activation may be independent from ROS generation, since AdoMet which exerted antioxidant properties did not have a significant effect on JNK activity. JNK pathway modulator agents and AdoMet may be components of promising therapies for alcoholic liver disease (ALD) treatment.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , JNK Mitogen-Activated Protein Kinases/physiology , S-Adenosylmethionine/pharmacology , Signal Transduction/drug effects , Animals , Anthracenes/pharmacology , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cytochromes c/metabolism , Ethanol/pharmacology , Glutathione/metabolism , Hepatocytes/physiology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mitochondria , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
9.
Cancer Lett ; 217(1): 25-32, 2005 Jan 10.
Article in English | MEDLINE | ID: mdl-15596293

ABSTRACT

Oxidative stress during carcinogen metabolism seems to participate in liver tumor production in the rat. N-diethylnitrosamine is an important carcinogen used in liver cancer animal models. This indirect alkylating agent produces DNA-ethyl adducts and oxidative stress. In contrast, N-ethyl-N-nitrosourea, a direct mutagen, which generates DNA-ethyl adducts, does not produce liver tumors in rat unless it is given under oxidative stress conditions such as partial hepatectomy or phenobarbital treatment. To gain insight into the relation between oxidative stress and hepatocarcinogenicity, the induction of preneoplastic liver lesions was compared among three different initiation protocols related to the initiation-promotion-resistant hepatocyte model. In addition, liver lipid peroxidation levels, determined as thiobarituric acid reactive substances were studied early during the initiation stage. Rats initiated with N-ethyl-N-nitrosourea, 25 days after treatment developed fewer and smaller gamma-glutamyl transpeptidase positive preneoplastic lesions than rats initiated with N-diethylnitrosamine. A pre-treatment with the antioxidant quercetin 1 h before N-diethylnitrosamine initiation, significantly prevented development of gamma-glutamyl transpeptidase-positive lesions. Increased lipid peroxidation levels were induced with N-diethylnitrosamine from 3 to 24 h after initiation, while N-ethyl-N-nitrosourea did not induce increments, and importantly, pre-treatment with quercetin decreased lipid peroxidation induced by N-diethylnitrosamine. These results show correlation between lipid peroxidation and hepatocarcinogenicity and support the important role of oxidative stress on liver carcinogenesis.


Subject(s)
Alkylating Agents/toxicity , Lipid Peroxidation/drug effects , Liver Neoplasms, Experimental/chemically induced , Oxidative Stress/drug effects , Precancerous Conditions/chemically induced , Animals , Antioxidants/pharmacology , Diethylnitrosamine/toxicity , Ethylnitrosourea/toxicity , Liver Neoplasms, Experimental/pathology , Precancerous Conditions/pathology , Quercetin/pharmacology , Rats , Thiobarbituric Acid Reactive Substances/metabolism , gamma-Glutamyltransferase/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...