Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 294(31): 11762-11771, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31189653

ABSTRACT

Gap junctions contain intercellular channels that enable intercellular communication of small molecules while also serving as a signaling scaffold. Connexins, the proteins that form gap junctions in vertebrates, are highly regulated and typically have short (<2 h) half-lives. Connexin43 (Cx43), the predominate connexin in the myocardium and epithelial tissues, is phosphorylated on more than a dozen serine residues and interacts with a variety of protein kinases. These interactions regulate Cx43 and gap junction formation and stability. Casein kinase 1 (CK1)-mediated phosphorylation of Cx43 promotes gap junction assembly. Using murine knock-in technology and quantitative PCR, immunoblotting, and immunoprecipitation assays, we show here that mutation of the CK1 phosphorylation sites in Cx43 reduces the levels of total Cx43 in the myocardium and increases Cx43 phosphorylation on sites phosphorylated by extracellular signal-regulated kinase (ERK). In aged myocardium, we found that, compared with WT Cx43, mutant Cx43 expression increases ERK activation, phosphorylation of Akt substrates, and protection from ischemia-induced injury. Our findings also uncovered that Cx43 interacts with the hypoxia-inducible protein N-Myc downstream-regulated gene 1 protein (NDRG1) and that Cx43 phosphorylation status controls this interaction and dramatically affects NDRG1 stability. We propose that, in addition to altering gap junction stability, Cx43 phosphorylation directly and dynamically regulates cellular signaling through ERK and Akt in response to ischemic injury. We conclude that gap junction-dependent NDRG1 regulation might explain some cellular responses to hypoxia.


Subject(s)
Cell Cycle Proteins/metabolism , Connexin 43/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Aging , Animals , Casein Kinase I/metabolism , Connexin 43/genetics , Dogs , Gap Junctions/metabolism , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Mutagenesis , Myocardium/metabolism , Phosphorylation/drug effects , Protein Stability , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Substrate Specificity
2.
Can J Physiol Pharmacol ; 91(2): 157-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23458200

ABSTRACT

Connexin43 has been recognized as forming gap junctions in Leydig cells. However, previous work has shown that mouse Leydig cells lacking this connexin do not suffer any limitation of their ability to produce testosterone when stimulated with luteinizing hormone. The objective of this study was to identify additional connexins in mouse Leydig cells that could be required for steroidogenesis. A reverse transcription - polymerase chain reaction screen involving isolated adult Leydig cells identified connexin36 and connexin45 as expressed along with connexin43. Treatment of dissociated testes with carbenoxolone, a nonspecific blocker of gap junctional coupling, significantly reduced testosterone output as did treatment with quinine, which disrupts coupling provided by connexin36 and connexin45 gap junctions but not those composed of connexin43, indicating that either or both of connexins 36 and 45 could be involved in supporting Leydig cell steroidogenesis. Immunolabeling of adult mouse testis sections confirmed the localization of connexin36 along with connexin43 in Leydig cell gap junctions but not connexin45, which is distributed throughout the cells. It was concluded that connexin36, connexin43, and connexin45 are coexpressed in Leydig cells with connexins 36 and 43 contributing to gap junctions. The role of connexin45 remains to be elucidated.


Subject(s)
Connexin 43/metabolism , Connexins/metabolism , Leydig Cells/metabolism , Testosterone/biosynthesis , Animals , Carbenoxolone/pharmacology , Cells, Cultured , Connexin 43/genetics , Connexins/genetics , Fluorescent Antibody Technique , Gap Junctions/metabolism , Leydig Cells/drug effects , Luteinizing Hormone/pharmacology , Male , Mice , Quinine/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Testis/cytology , Testis/drug effects , Testis/metabolism , Gap Junction delta-2 Protein
3.
J Cell Sci ; 125(Pt 3): 695-702, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22389404

ABSTRACT

Vertebrate gap junctions are composed of proteins from the connexin family. Co-immunoprecipitation, in vitro binding and far western experiments demonstrate that mammalian CASK (also known as LIN2) directly interacts with Cx43. Immunoprecipitation studies indicate that the CASK mainly interacts with the hypophosphorylated form of Cx43. Functional co-regulation of these proteins was found in MDCK cells migrating into a scratch wound, where expression of either protein individually inhibits migration but their coexpression abrogates this inhibitory effect. Immunofluorescence shows colocalization of Cx43 and CASK in mouse brain astrocytes and in response to wounding in human foreskin. During wounding, CASK is mobilized to the plasma membrane where it colocalizes with Cx43 and CADM1 1 hour after skin explant wounding. Together, these studies indicate that CASK interaction with Cx43 occurs relatively early in the connexin life cycle and imply a plasma membrane targeting role for the interaction that apparently affects cellular processes including cellular migration and wound healing.


Subject(s)
Connexin 43/metabolism , Guanylate Kinases/metabolism , Wound Healing/physiology , Animals , Astrocytes/metabolism , Base Sequence , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement/physiology , Connexin 43/genetics , Dogs , Gene Knockdown Techniques , Guanylate Kinases/antagonists & inhibitors , Guanylate Kinases/genetics , Humans , Immunoglobulins/metabolism , Male , Mice , Phosphorylation , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin/cytology , Skin/injuries , Skin/metabolism , Transfection
4.
Biochim Biophys Acta ; 1818(8): 1985-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21819962

ABSTRACT

Gap junctions, composed of proteins from the connexin family, allow for intercellular communication between cells in essentially all tissues. There are 21 connexin genes in the human genome and different tissues express different connexin genes. Most connexins are known to be phosphoproteins. Phosphorylation can regulate connexin assembly into gap junctions, gap junction turnover and channel gating. Given the importance of gap junctions in development, proliferation and carcinogenesis, regulation of gap junction phosphorylation in response to wounding, hypoxia and other tissue insults is proving to be critical for cellular response and return to homeostasis. Connexin43 (Cx43) is the most widely and highly expressed gap junction protein, both in cell culture models and in humans, thus more research has been done on it and more reagents to it are available. In particular, antibodies that can report Cx43 phosphorylation status have been created allowing temporal examination of specific phosphorylation events in vivo. This review is focused on the use of these antibodies in tissue in situ, predominantly looking at Cx43 phosphorylation in brain, heart, endothelium and epithelium with reference to other connexins where data is available. These data allow us to begin to correlate specific phosphorylation events with changes in cell and tissue function. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Subject(s)
Brain/metabolism , Connexin 43/physiology , Endothelium/metabolism , Epithelium/metabolism , Gap Junctions/physiology , Myocardium/metabolism , Animals , Connexin 43/metabolism , Genome, Human , Homeostasis , Humans , Models, Biological , Neoplasms/metabolism , Phosphorylation , Signal Transduction , Tissue Distribution
5.
J Cardiovasc Dis Res ; 2(4): 206-12, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22135478

ABSTRACT

BACKGROUND: GJA1 gene encodes a gap junction protein known as connexin 43 (Cx43). Cx43 is abundantly expressed in the ventricular myocardium and in cardiac neural crest cells. Cx43 is proposed to play an important role in human congenital heart disease, as GJA1 knock-out mice die neonatally from outflow tract obstruction. In addition, patients with visceroatrial heterotaxia or hypoplastic left heart syndrome were reported to have point mutations in GJA1 at residues that affect protein kinase phosphorylation and gating of the gap junction channel. However, as these clinical findings were not replicated in subsequent studies, the question remains about the contribution of GJA1 mutations in human congenital heart disease (CHD). MATERIALS AND METHODS: We analyzed the GJA1 coding sequence in 300 patients with CHD from two clinical centers, focusing on outflow tract anomalies. This included 152 with Tetralogy of Fallot from over 200 patients exhibiting outflow tract anomalies, as well as other structural heart defects including atrioventricular septal defects and other valvar anomalies. Our sequencing analysis revealed only two silent nucleotide substitutions in 8 patients. To further assess the possible role of Cx43 in CHD, we also generated two knock-in mouse models with point mutations at serine residues subject to protein kinase C or casein kinase phosphorylation, sites that are known to regulate gating and trafficking of Cx43, respectively. RESULTS: Both heterozygous and homozygous knock-in mice were long term viable and did not exhibit overt CHD. CONCLUSION: The combined clinical and knock-in mouse mutant studies indicate GJA1 mutation is not likely a major contributor to CHD, especially those involving outflow tract anomalies.

6.
Circ Res ; 104(3): 365-71, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19096029

ABSTRACT

Pressure overload is a common pathological insult to the heart and the resulting hypertrophy is an independent risk factor for sudden cardiac death. Gap junction remodeling (GJR) has been described in hypertrophied hearts; however, a detailed understanding of the remodeling process and its effects on impulse propagation is lacking. Moreover, there has been little progress developing therapeutic strategies to diminish GJR. Accordingly, transverse aortic banding (TAC) was performed in mice to determine the effects of progressive pathological hypertrophy on connexin (Cx)43 expression, posttranslational phosphorylation, gap junction assembly, and impulse propagation. Within 2 weeks after TAC, total and phospho-Cx43 abundance was reduced and incorporation of Cx43 into gap junctional plaques was markedly diminished. These molecular changes were associated with progressive slowing of impulse propagation, as determined by optical mapping with voltage-sensitive dyes. Treatment with the aldosterone receptor antagonist spironolactone, which has been shown to diminish sudden arrhythmic death in clinical trials, was examined for its effects on GJR. We found that spironolactone blunted the development of GJR and also potently reversed established GJR, both at the molecular and functional levels, without diminishing the extent of hypertrophy. These data suggest a potential mechanism for some of the salutary electrophysiological and clinical effects of mineralocorticoid antagonists in myopathic hearts.


Subject(s)
Cardiomegaly/drug therapy , Cardiomegaly/pathology , Diuretics/pharmacology , Gap Junctions/pathology , Spironolactone/pharmacology , Animals , Cardiomegaly/diagnostic imaging , Connexin 43/metabolism , Disease Models, Animal , Gap Junctions/drug effects , Gap Junctions/physiology , Heart Conduction System/physiology , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Myocardial Contraction/physiology , Phosphorylation , Ultrasonography
7.
J Cell Biol ; 179(6): 1301-9, 2007 Dec 17.
Article in English | MEDLINE | ID: mdl-18086922

ABSTRACT

Phosphorylation at unspecified sites is known to regulate the life cycle (assembly, gating, and turnover) of the gap junction protein, Cx43. In this paper, we show that Cx43 is phosphorylated on S365 in cultured cells and heart tissue. Nuclear magnetic resonance structural studies of the C-terminal region of Cx43 with an S365D mutation indicate that it forms a different stable conformation than unphosphorylated wild-type Cx43. Immunolabeling with an antibody specific for Cx43 phosphorylated at S365 shows staining on gap junction structures in heart tissue that is lost upon hypoxia when Cx43 is no longer specifically localized to the intercalated disk. Efficient phosphorylation at S368, an important Cx43 channel regulatory event that increases during ischemia or PKC activation, depends on S365 being unphosphorylated. Thus, phosphorylation at S365 can serve a "gatekeeper" function that may represent a mechanism to protect cells from ischemia and phorbol ester-induced down-regulation of channel conductance.


Subject(s)
Connexin 43/metabolism , Connexins/metabolism , Down-Regulation , Protein Kinase C/metabolism , Amino Acid Substitution , Animals , Blotting, Western , Cell Line , Connexin 43/chemistry , Connexin 43/genetics , Connexins/chemistry , Connexins/genetics , Dogs , Fluorescent Antibody Technique , Hypoxia/metabolism , Mice , Myocardium/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Serine/metabolism
8.
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
9.
World J Gastroenterol ; 12(42): 6779-85, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17106925

ABSTRACT

AIM: To study of the effect of caffeic acid phenethyl ester (CAPE) on the initiation period in a medium-term assay of hepatocarcinogenesis. METHODS: Male Wistar rats were subjected to a carcinogenic treatment (CT) and sacrificed at 25th d; altered hepatic foci (AHF) were generated efficiently. To a second group of rats a single 20 mg/kg doses of CAPE was given 12 h before initiation with CT and were sacrificed at 25th d. We evaluated the expression of preneoplastic markers as gamma-glutamyltranspeptidase (GGT) and glutathione S-transferase type pi protein (GSTp) by histochemistry, RT-PCR and Western blot analyses, respectively. We measured thiobarbituric acid reactive substances (TBARS) in homogenates of liver and used Unscheduled DNA Synthesis (UDS) assay by incorporation of [3H] thymidine (3HdT) in primary hepatocyte cultures (PHC). RESULTS: At 25th d after CT CAPE reduced the observed increase of GGT+AHF by 84% and liver expression of ggt mRNA by 100%. In case of the GSTp protein, the level was reduced by 90%. As indicative of oxidative stress generated by diethylnitrosamine (DEN) 12 h after its administration, we detected a 68% increase of TBARS. When CAPE was administered before DEN, it completely protected from liver TBARS induction. To have an indication of the sole effect of CAPE on initiation, two carcinogens were tested in a UDS assay in PHC, we used methyl-n-nitrosoguanidine as a direct carcinogen and DEN, as indirect carcinogen. In this assay, genotoxic damage caused by carcinogens was abolished at 5 microM CAPE concentration. CONCLUSION: Our results demonstrated that CAPE possesses anti-genotoxic and antineoplastic capabilities, by an anti-oxidative and free-radical scavenging mechanism.


Subject(s)
Caffeic Acids/pharmacology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/drug effects , Cytotoxins/pharmacology , Liver Neoplasms, Experimental/prevention & control , Phenylethyl Alcohol/analogs & derivatives , 2-Acetylaminofluorene , Animals , Caffeic Acids/administration & dosage , Carcinogens , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cytotoxins/administration & dosage , Diethylnitrosamine/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Lipid Peroxidation/drug effects , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , NF-kappa B/antagonists & inhibitors , Oxidative Stress/drug effects , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , gamma-Glutamyltransferase/genetics , gamma-Glutamyltransferase/metabolism
10.
Anticancer Res ; 26(2A): 1271-80, 2006.
Article in English | MEDLINE | ID: mdl-16619534

ABSTRACT

BACKGROUND: Celecoxib, a cyclooxygenase-2 (COX-2) inhibitor, may reduce the risk and mortality of certain types of human cancer. The chemopreventive effect of celecoxib on preneoplastic lesions induced by chemical hepatocarcinogenesis was investigated. MATERIALS AND METHODS: Male Sprague Dawley rats were fed a celecoxib-supplemented diet between days 18 and 26 post-initiation (1500 ppm) and sacrificed on day 26. The effects of celecoxib on proliferation, apoptosis, COX-2 activity and liver function were evaluated by immunohistochemistry, TUNEL assay, enzyme-immunoassay and spectrophotometry, respectively. RESULTS: Celecoxib decreased, in area and number, gamma-glutamyltranspeptidase and glutathione S-transferase placental-positive lesions, below levels found after 18 days, by 55.2% and 62.2%, and by 50.5% and 71.1%, respectively, (p < 0.05). Celecoxib neither induced apoptosis nor altered the levels of prostaglandin E2, bilirubin or alanine aminotransferase in the plasma; however, proliferating cell nuclear antigen and cyclin D1 decreased by 77.7% and 94.9%, respectively, (p < 0.05). CONCLUSION: Celecoxib regresses existing preneoplastic liver lesions through antiproliferative processes, without altering liver function.


Subject(s)
Liver Neoplasms, Experimental/prevention & control , Precancerous Conditions/drug therapy , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Growth Processes/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/blood , Liver/anatomy & histology , Liver/drug effects , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/enzymology , Male , Organ Size/drug effects , Precancerous Conditions/blood , Precancerous Conditions/enzymology , Rats , Rats, Sprague-Dawley
11.
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
12.
Neoplasia ; 7(6): 563-74, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16036107

ABSTRACT

In this work, we investigated the effects of Casiopeina II-gly (Cas IIgly)--a new copper compound exhibiting antineoplastic activity--on glioma C6 cells under both in vitro and in vivo conditions, as an approach to identify potential therapeutic agents against malignant glioma. The exposure of C6 cells to Cas IIgly significantly inhibited cell proliferation, increased reactive oxygen species (ROS) formation, and induced apoptosis in a dose-dependent manner. In cultured C6 cells, Cas IIgly caused mitochondrio-nuclear translocation of apoptosis induction factor (AIF) and endonuclease G at all concentrations tested; in contrast, fragmentation of nucleosomal DNA, cytochrome c release, and caspase-3 activation were observed at high concentrations. Administration of N-acetyl-L-cystein, an antioxidant, resulted in significant inhibition of AIF translocation, nucleosomal DNA fragmentation, and caspase-3 activation induced by Cas IIgly. These results suggest that caspase-dependent and caspase-independent pathways both participate in apoptotic events elicited by Cas IIgly. ROS formation induced by Cas IIgly might also be involved in the mitochondrio-nuclear translocation of AIF and apoptosis. In addition, treatment of glioma C6-positive rats with Cas IIgly reduced tumor volume and mitotic and cell proliferation indexes, and increased apoptotic index. Our findings support the use of Cas IIgly for the treatment of malignant gliomas.


Subject(s)
Caspases/metabolism , Copper/pharmacology , Glioma/drug therapy , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Acetylcysteine/pharmacology , Active Transport, Cell Nucleus , Animals , Antioxidants/pharmacology , Apoptosis , Blotting, Western , Caspase 3 , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , DNA Fragmentation , Dose-Response Relationship, Drug , In Vitro Techniques , Lipid Peroxidation , Membrane Potentials , Mitochondria/pathology , Nucleosomes/metabolism , Protein Transport , Rats , Rats, Wistar , Reactive Oxygen Species , Subcellular Fractions
13.
J Hepatol ; 43(4): 653-60, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16023763

ABSTRACT

BACKGROUND/AIMS: Several studies suggest that cyclooxygenase-2 (COX-2) inhibitors are chemopreventive agents against colon, breast and skin cancer. In this study, we evaluated the chemopreventive effect of celecoxib, a specific COX-2 inhibitor, on the development of liver preneoplastic lesions in rats. METHODS: Male Sprague-Dawley rats were fed during 5 weeks either a control or an experimental diet containing 1500 ppm celecoxib on a medium-term hepatocarcinogenesis protocol. Livers were collected and evaluated by histological and biochemical assays. RESULTS: A reduction by 80 and 90% both in the number and size of altered hepatic foci was observed in the group treated with celecoxib during hepatocarcinogenesis treatment, respectively. No evidence of apoptosis was observed in our present study, however, the expression of the proliferation markers such as PCNA and Ki-67 was drastically reduced. Interestingly, neither COX-2 expression nor prostaglandin-E2 (PGE2) production were altered by the hepatocarcinogenic treatment or celecoxib treatment. Finally, celecoxib inhibited the translocation of Rel A/p65 to the nucleus with significant effect on stability of the repressor IkappaB-alpha. CONCLUSIONS: This is the first demonstration that a specific COX-2 inhibitor, celecoxib, possesses striking chemopreventive activity, inhibiting preneoplastic lesions during hepatocarcinogenesis in vivo, suggesting that celecoxib effects are mediated by PGE2-independent mechanisms.


Subject(s)
Cyclooxygenase Inhibitors/therapeutic use , Liver Neoplasms/prevention & control , Precancerous Conditions/prevention & control , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Animals , Celecoxib , Liver Neoplasms, Experimental/prevention & control , Male , Models, Animal , Rats , Rats, Sprague-Dawley
14.
Int J Cancer ; 108(4): 488-92, 2004 Feb 10.
Article in English | MEDLINE | ID: mdl-14696111

ABSTRACT

Caffeic acid phenethyl ester (CAPE), a natural honeybee product exhibits a spectrum of biological activities including anti-microbial, anti-inflammatory, antioxidant and anti-tumoral actions. CAPE is also chemopreventive against intestinal, colon and skin cancer. Our aim was to extend the study of its chemoprotective features to the promotion of hepatocarcinogenesis. Male Wistar rats were subjected to a protocol under a modified promotion regimen of the resistant hepatocyte model. The altered hepatic foci (AHF) were quantitatively analyzed by histochemistry and image processing. When given during promotion, CAPE (20 mg/kg) decreased the expression of number and area gamma-glutamyl transpeptidase (GGT) positive AHF by 91% and 97%, respectively. When GGT expression was analyzed by RT-PCR, CAPE drastically decreased and prevented expression of almost all GGT transcripts at this stage of the carcinogenic process. Glutathione S-transferase placental form (GST-P), another protein marker for preneoplastic lesions was measured by Western blot and a decrease of 82% was observed. Additionally, we evaluated the effect of CAPE on the expression of nuclear factor NF-kappaB and found an 85% decrease in nuclear localization of the p65 subunit of NF-kappaB; however, their repressor, IkappaBalpha was not modified. Our results showed that CAPE given during promotion in hepatocarcinogenesis protects against induction of GGT-positive AHF, GST-P protein, GGT mRNA expression and translocation of p65. This phenomenon was independent of IkappaBalpha degradation.


Subject(s)
Caffeic Acids/therapeutic use , Carcinoma, Hepatocellular/prevention & control , Liver Neoplasms, Experimental/prevention & control , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/therapeutic use , Precancerous Conditions/prevention & control , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Nucleus/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , I-kappa B Proteins/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Transport , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , gamma-Glutamyltransferase/genetics , gamma-Glutamyltransferase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...