Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 110(11): 2747-55, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24809783

ABSTRACT

BACKGROUND: Elevated expression of focal adhesion kinase (FAK) occurs in numerous human cancers including colon-, cervix- and breast cancer. Although several studies have implicated FAK in mammary tumour formation induced by ectopic oncogene expression, evidence supporting a role for FAK in spontaneous mammary tumour development caused by loss of tumour suppressor genes such as p53 is lacking. Alterations in the tumour suppressor gene p53 have been implicated in over 50% of human breast cancers. Given that elevated FAK expression highly correlates with p53 mutation status in human breast cancer, we set out to investigate the importance of FAK in p53-mediated spontaneous mammary tumour development. METHODS: To directly assess the role of FAK, we generated mice with conditional inactivation of FAK and p53. We generated female p53(lox/lox)/FAK(+/+)/WapCre, p53(lox/lox)/FAK(flox/+)/WapCre and p53(lox/lox)/FAK(flox/-)/WapCre mice, and mice with WapCre-mediated conditional expression of p53(R270H), the mouse equivalent of human p53(R273H) hot spot mutation, together with conditional deletion of FAK, P53(R270H/+)/FAK(lox/+)/WapCre and p53(R270H/+)/FAK(flox/-)/WapCre mice. All mice were subjected to one pregnancy to induce WapCre-mediated deletion of p53 or expression of p53 R270H, and Fak genes flanked by two loxP sites, and subsequently followed the development of mammary tumours. RESULTS: Using this approach, we show that FAK is important for p53-induced mammary tumour development. In addition, mice with the mammary gland-specific conditional expression of p53 point mutation R270H, the mouse equivalent to human R273H, in combination with conditional deletion of Fak showed reduced incidence of p53(R270H)-induced mammary tumours. In both models these effects of FAK were related to reduced proliferation in preneoplastic lesions in the mammary gland ductal structures. CONCLUSIONS: Mammary gland-specific ablation of FAK hampers p53-regulated spontaneous mammary tumour formation. Focal adhesion kinase deletion reduced proliferative capacity of p53 null and p53(R270H) mammary epithelial cells but did not lead to increased apoptosis in vivo. Our data identify FAK as an important regulator in mammary epithelial cell proliferation in p53-mediated and p53(R270H)-induced mammary tumour development.


Subject(s)
Carcinoma/enzymology , Carcinosarcoma/enzymology , Focal Adhesion Kinase 1/genetics , Mammary Neoplasms, Experimental/enzymology , Tumor Suppressor Protein p53/metabolism , Animals , Carcinogenesis/metabolism , Carcinoma/genetics , Carcinoma/pathology , Carcinosarcoma/genetics , Carcinosarcoma/pathology , Cell Proliferation , Epithelial Cells/enzymology , Female , Focal Adhesion Kinase 1/deficiency , Humans , Incidence , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mutation, Missense , Tumor Burden , Tumor Suppressor Protein p53/genetics
2.
Biochem Pharmacol ; 62(8): 1087-97, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11597577

ABSTRACT

Caspase activation is a central event in the execution phase of apoptosis and is associated with phosphatidylserine (PS) externalization and DNA fragmentation. We investigated the role of caspase activity in anticancer drug-induced PS externalization and DNA fragmentation in MTLn3 cells. Caspase activation (DEVD-AMC cleavage) occurred in a time- and concentration-dependent manner after exposure to doxorubicin, in association with cleavage of poly(ADP) ribose polymerase and protein kinase C delta, two caspase-3 substrates. Caspase activation was closely followed by oligonucleosomal DNA fragmentation and PS externalization as determined by flow cytometric analysis. Similar observations were made for etoposide and cisplatin. Inhibition of caspases with zVAD-fmk inhibited almost completely doxorubicin-induced DNA fragmentation as well as proteolysis of protein kinase C delta. In contrast, PS externalization induced by doxorubicin was only partly affected by caspase inhibition. Flow cytometric cell sorting demonstrated that DNA fragmentation in the remaining PS positive cells after doxorubicin treatment in the presence of zVAD-fmk was fully blocked. In conclusion, these data indicate that while DNA fragmentation in anticancer drug-induced apoptosis of MTLn3 cells is fully dependent on caspase activity, PS externalization is controlled by both caspase-dependent and caspase-independent pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Caspases/physiology , DNA Fragmentation/drug effects , Phosphatidylserines/metabolism , Adenocarcinoma/pathology , Animals , Caspase 3 , Cisplatin/pharmacology , Doxorubicin/pharmacology , Etoposide/pharmacology , Exocytosis/drug effects , Mammary Neoplasms, Animal/pathology , Necrosis , Rats , Tumor Cells, Cultured
3.
J Biol Chem ; 276(39): 36183-93, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11447217

ABSTRACT

Decreased phosphorylation of focal adhesion kinase (FAK) is associated with loss of focal adhesions and actin stress fibers and precedes the onset of apoptosis in renal epithelial cells caused by nephrotoxicants (Van de Water, B., Nagelkerke, J. F., and Stevens, J. L. (1999) J. Biol. Chem. 274, 13328-13337). The role of FAK in the control of apoptosis caused by nephrotoxicants was further investigated in LLC-PK1 cells that were stably transfected with either green fluorescent protein (GFP)-FAK or dominant negative acting deletion mutants of FAK, GFP-FAT, and GFP-FRNK. GFP-FAT and GFP-FRNK delayed the formation of focal adhesions and prevented the localization of endogenous (phosphorylated) FAK at these sites. GFP-FAT and GFP-FRNK overexpression potentiated the onset of apoptosis caused by the nephrotoxicant dichlorovinyl-cysteine. This was associated with an increased activation of caspase-3. GFP-FAT also potentiated apoptosis caused by doxorubicin but not cisplatin. The potentiation of apoptosis by GFP-FAT was related to an almost complete dephosphorylation of FAK; this did not occur in cells overexpressing only GFP. This dephosphorylation was associated with a pronounced loss of focal adhesion organization in GFP-FAT cells, in association with loss of tyrosine phosphorylation of paxillin. In conclusion, the data indicate an important role of cell-matrix signaling in the control of chemically induced apoptosis; loss of FAK activity caused by toxic chemicals results in perturbations of focal adhesion organization with a subsequent inactivation of associated (signaling) molecules and loss of survival signaling.


Subject(s)
Apoptosis , Epithelial Cells/enzymology , Necrosis , Protein-Tyrosine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Caspase 3 , Caspases/metabolism , Cell Cycle , Cell Division , Cell Survival , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Focal Adhesion Protein-Tyrosine Kinases , Gene Deletion , Genes, Dominant , Green Fluorescent Proteins , LLC-PK1 Cells , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Recombinant Fusion Proteins/metabolism , Signal Transduction , Swine , Time Factors , Transfection
4.
J Biol Chem ; 275(33): 25805-13, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10823823

ABSTRACT

Decreased phosphorylation of focal adhesion kinase and paxillin is associated with loss of focal adhesions and stress fibers and precedes the onset of apoptosis (van de Water, B., Nagelkerke, J. F., and Stevens, J. L. (1999) J. Biol. Chem. 274, 13328-13337). The cortical actin cytoskeletal network is also lost during apoptosis, yet little is known about the temporal relationship between altered phosphorylation of proteins that are critical in the regulation of this network and their potential cleavage by caspases during apoptosis. Adducins are central in the cortical actin network organization. Cisplatin caused apoptosis of renal proximal tubular epithelial cells, which was associated with the cleavage of alpha-adducin into a 74-kDa fragment; this was blocked by a general caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (z-VAD-fmk). Hemagglutinin-tagged human alpha-adducin was cleaved into a similar 74-kDa fragment by caspase-3 in vitro but not by caspase-6 or -7. Asp-Arg-Val-Asp(29)-Glu, Asp-Ile-Val-Asp(208)-Arg, and Asp-Asp-Ser-Asp(633)-Ala were identified as the principal caspase-3 cleavage sites; Asp-Asp-Ser-Asp(633)-Ala was key in the formation of the 74-kDa fragment. Cisplatin also caused an increased phosphorylation of alpha-adducin and gamma-adducin in the MARCKS domain that preceded alpha-adducin cleavage and was associated with loss of adducins from adherens junctions; this was not affected by z-VAD-fmk. In conclusion, the data support a model in which increased phosphorylation of alpha-adducin due to cisplatin leads to dissociation from the cytoskeleton, a situation rendered irreversible by caspase-3-mediated cleavage of alpha-adducin at Asp-Asp-Ser-Asp(633)-Ala.


Subject(s)
Apoptosis , Calmodulin-Binding Proteins/metabolism , Caspases/metabolism , Kidney/metabolism , Actins/metabolism , Amino Acid Chloromethyl Ketones/metabolism , Animals , Antineoplastic Agents/pharmacology , COS Cells , Carrier Proteins/metabolism , Caspase 3 , Cell Death , Cisplatin/pharmacology , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Immunoblotting , L-Lactate Dehydrogenase/metabolism , Microfilament Proteins/metabolism , Phosphorylation , Plasmids/metabolism , Protein Isoforms , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Rats , Serine/metabolism , Time Factors , Transfection
5.
Biochem Pharmacol ; 44(12): 2339-45, 1992 Dec 15.
Article in English | MEDLINE | ID: mdl-1472098

ABSTRACT

Isolated rat hepatocytes were incubated with the carcinogen N-hydroxy-2-acetylaminofluorene (N-OH-AAF). Cells from fasted rats were much more susceptible to the cytotoxic effects of 1 mM N-OH-AAF than cells from fed rats: after approximately 90 min exposure the former were all dead but the latter still viable. Even after 240 min 25% of the "fed" cells were still viable. The loss of viability was preceded by a decrease in mitochondrial membrane potential (MMP) and inhibition of respiration; the mitochondrial respiration as measured in permeabilized cells appeared uncoupled. Addition of 15 mM fructose prevented cell death and the loss of MMP in cells both from fed and fasted rats to a large extent; however, uncoupling was not prevented. After incubation of hepatocytes from fasted rats with 1 mM [3H]N-OH-AAF for 120 min, 12 nmol [3H]N-OH-AAF became bound per mg cell protein. Addition of fructose decreased this to 7 nmol. In cells from fed animals 4 nmol [3H]N-OH-AAF became bound after 120 min, in this case fructose had no effect. Part of the protective effect of fructose might be explained by a decrease in intracellular ATP, which prevents the formation of reactive intermediates of N-OH-AAF resulting in a decrease of covalent binding, in addition, fructose protects via a yet to be determined mechanism.


Subject(s)
Fasting/metabolism , Hydroxyacetylaminofluorene/pharmacology , Liver/drug effects , Adenosine Triphosphate/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured/drug effects , Fructose/pharmacology , Liver/metabolism , Liver/pathology , Male , Membrane Potentials/drug effects , Mitochondria, Liver/drug effects , Oxidation-Reduction , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...