Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Oncogene ; 36(16): 2328-2336, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27819670

ABSTRACT

Heat shock protein 110 (HSP110) is induced by different stresses and, through its anti-apoptotic and chaperoning properties, helps cells survive these adverse situations. In colon cancers, HSP110 is abnormally abundant. We have recently shown that colorectal cancer patients with microsatellite instability (MSI) had an improved response to chemotherapy because they harbor an HSP110-inactivating mutation (HSP110DE9). In this work, we used patient biopsies, human colorectal cancer cells grown in vitro and in vivo (xenografts), and intestinal crypts to demonstrate that HSP110 is also involved in colon cancer growth. We showed that HSP110 induces colon cancer cell proliferation and that this effect is associated with STAT3 activation, specifically an increase in STAT3 phosphorylation, nuclear translocation and transcription factor activity. STAT3 inhibition blocks the proliferative effect of HSP110. From a molecular standpoint, we demonstrated that HSP110 directly binds to STAT3, thereby facilitating its phosphorylation by JAK2. Finally, we showed a correlation between HSP110 expression and STAT3 phosphorylation in colon cancer patient samples. Thus, the expression of HSP110 in colon cancer contributes to STAT3-dependent tumor growth and the frequent inactivating mutation of this chaperone is probably an important event underlying the improved prognosis in colon cancer displaying MSI.


Subject(s)
Colorectal Neoplasms/pathology , HSP110 Heat-Shock Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Protein Binding
3.
Cell Death Dis ; 7: e2195, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27077811

ABSTRACT

Inactivation of p53 found in more than half of human cancers is often associated with increased tumor resistance to anti-cancer therapy. We have previously shown that overexpression of the phosphatase Wip1 in p53-negative tumors sensitizes them to chemotherapeutic agents, while protecting normal tissues from the side effects of anti-cancer treatment. In this study, we decided to search for kinases that prevent Wip1-mediated sensitization of cancer cells, thereby interfering with efficacy of genotoxic anti-cancer drugs. To this end, we performed a flow cytometry-based screening in order to identify kinases that regulated the levels of γH2AX, which were used as readout. Another criterion of the screen was increased sensitivity of p53-negative tumor cells to cisplatin (CDDP) in a Wip1-dependent manner. We have found that a treatment with a low dose (75 nM) of MK-1775, a recently described specific chemical inhibitor of Wee1, decreases CDDP-induced H2AX phosphorylation in p53-negative cells and enhances the Wip1-sensitization of p53-negative tumors. We were able to reduce CDDP effective concentration by 40% with a combination of Wip1 overexpression and Wee1 kinase inhibition. We have observed that Wee1 inhibition potentiates Wip1-dependent tumor sensitization effect by reducing levels of Hipk2 kinase, a negative regulator of Wip1 pathway. In addition, during CDDP treatment, the combination of Wee1 inhibition and Wip1 overexpression has a mild but significant protective effect in normal cells and tissues. Our results indicate that inhibition of the negative regulators of Wip1 pathway, Wee1 and Hipk2, in p53-negative tumors could potentiate efficiency of chemotherapeutic agents without concomitant increase of cytotoxicity in normal tissues. The development and clinical use of Wee1 and Hipk1 kinase chemical inhibitors might be a promising strategy to improve anti-cancer therapy.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Protein Phosphatase 2C/metabolism , Protein-Tyrosine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Damage/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Histones/metabolism , Humans , Mice , Mice, Transgenic , Mitochondria/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phosphorylation/drug effects , Protein Phosphatase 2C/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RNA Interference , Survival Rate , Tumor Suppressor Protein p53/deficiency
4.
Oncogene ; 35(22): 2842-51, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26364610

ABSTRACT

Graft versus host disease (GvHD), which is the primary complication of allogeneic bone marrow transplantation, can alter the intestinal barrier targeted by activated donor T-cells. Chemical inhibition of the stress protein HSP90 was demonstrated in vitro to inhibit T-cell activation and to modulate endoplasmic reticulum (ER) stress to which intestinal cells are highly susceptible. Since the HSP90 inhibitor 17-allylamino-demethoxygeldanamycin (17AAG) is developed in clinics, we explored here its ability to control intestinal acute GvHD in vivo in two mouse GvHD models (C57BL/6BALB/c and FVB/NLgr5-eGFP), ex vivo in intestine organoids and in vitro in intestinal epithelial cultures. We show that 17AAG decreases GvHD-associated mortality without impairing graft versus leukemia effect. While 17AAG effect in T-cell activation is just moderate at the dose used in vivo, we observe a striking intestinal integrity protection. At the intestine level, the drug promotes the splicing of the transcription factor X-box binding protein 1 (XBP1), which is a key component of the ER stress. This effect is associated with a decrease in intestinal damage and an increase in Lgr5(+) stem cells, Paneth cells and defensins production. The importance of XBP1 splicing control is further confirmed in cultured cells and organoids of primary intestinal epithelium where XBP1 is either shRNA depleted or inhibited with toyocamycin. In conclusion, 17AAG has a protective effect on the epithelial intestinal barrier in mouse models of acute GvHD. This compound deserves to be tested in the therapeutic control of acute GvHD.


Subject(s)
Benzoquinones/pharmacology , Cytoprotection/drug effects , Graft vs Host Disease/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Intestines/pathology , Lactams, Macrocyclic/pharmacology , Stem Cell Niche/drug effects , Animals , Benzoquinones/therapeutic use , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestines/drug effects , Lactams, Macrocyclic/therapeutic use , Mice , Mice, Inbred C57BL , RNA Splicing/drug effects , X-Box Binding Protein 1/genetics
5.
Cell Death Dis ; 5: e1016, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24457960

ABSTRACT

The proapoptotic protein, prostate apoptosis response-4 (Par-4), acts as a tumor suppressor in prostate cancer cells. The serine/threonine kinase casein kinase 2 (CK2) has a well-reported role in prostate cancer resistance to apoptotic agents or anticancer drugs. However, the mechanistic understanding on how CK2 supports survival is far from complete. In this work, we demonstrate both in rat and humans that (i) Par-4 is a new substrate of the survival kinase CK2 and (ii) phosphorylation by CK2 impairs Par-4 proapoptotic functions. We also unravel different levels of CK2-dependent regulation of Par-4 between species. In rats, the phosphorylation by CK2 at the major site, S124, prevents caspase-mediated Par-4 cleavage (D123) and consequently impairs the proapoptotic function of Par-4. In humans, CK2 strongly impairs the apoptotic properties of Par-4, independently of the caspase-mediated cleavage of Par-4 (D131), by triggering the phosphorylation at residue S231. Furthermore, we show that human Par-4 residue S231 is highly phosphorylated in prostate cancer cells as compared with their normal counterparts. Finally, the sensitivity of prostate cancer cells to apoptosis by CK2 knockdown is significantly reversed by parallel knockdown of Par-4. Thus, Par-4 seems a critical target of CK2 that could be exploited for the development of new anticancer drugs.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Casein Kinase II/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Casein Kinase II/genetics , Cell Line, Tumor , Humans , Male , Phosphorylation , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/physiopathology , Rats
6.
Skin Pharmacol Physiol ; 23(6): 328-32, 2010.
Article in English | MEDLINE | ID: mdl-20588084

ABSTRACT

AIM: To compare the tissue tolerance and efficacy of two wound antiseptics with tissue-tolerable plasma (TTP) on enucleated contaminated eyes from slaughtered pigs in order to draw consequences for the use of TTP on wounds. METHOD: The corneas of extracted eyes were contaminated with Staphylococcus aureus or Pseudomonas aeruginosa. One and 10 min after application of 10% povidone (PVP)-iodine and 0.04% polyhexanide, respectively, the eyes were rinsed with inactivating solution. To test TTP, the plasma pen meandered over the eyes at a speed of 30 mm/s and a distance of 5 mm; the eyes were then rinsed with balanced salt solution. The reduction factor was calculated by the difference between the logarithm of colony-forming units in the rinse before and after antisepsis or TTP application. RESULTS: The efficacy of TTP (reduction factor 2.4-2.9) was significantly higher (p < 0.001) than that of PVP-iodine and polyhexanide (reduction factor 1.7-2.1). CONCLUSION: TTP is more effective than the tested wound antiseptics. The lack of histological damage to the eyes of slaughtered pigs would seem to make its use as a wound antiseptic a viable alternative. In contrast to antiseptics, it supplies additional energy in the form of heat, electric fields and radicals by TTP.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Bacteria/drug effects , Biguanides/pharmacology , Cornea/microbiology , Plasma Gases/pharmacology , Povidone-Iodine/pharmacology , Wounds and Injuries/microbiology , Animals , Anti-Infective Agents, Local/toxicity , Antisepsis , Biguanides/toxicity , Colony Count, Microbial , Povidone-Iodine/toxicity , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Swine
7.
Oncogene ; 28(37): 3332-44, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19597476

ABSTRACT

Heat shock protein 27 (HSP27) accumulates in stressed cells and helps them to survive adverse conditions. We have already shown that HSP27 has a function in the ubiquitination process that is modulated by its oligomerization/phosphorylation status. Here, we show that HSP27 is also involved in protein sumoylation, a ubiquitination-related process. HSP27 increases the number of cell proteins modified by small ubiquitin-like modifier (SUMO)-2/3 but this effect shows some selectivity as it neither affects all proteins nor concerns SUMO-1. Moreover, no such alteration in SUMO-2/3 conjugation is achievable by another HSP, such as HSP70. Heat shock factor 1 (HSF1), a transcription factor responsible for HSP expression, is one of the targets of HSP27. In stressed cells, HSP27 enters the nucleus and, in the form of large oligomers, binds to HSF1 and induces its modification by SUMO-2/3 on lysine 298. HSP27-induced HSF1 modification by SUMO-2/3 takes place downstream of the transcription factor phosphorylation on S303 and S307 and does not affect its DNA-binding ability. In contrast, this modification blocks HSF1 transactivation capacity. These data show that HSP27 exerts a feedback inhibition of HSF1 transactivation and enlighten the strictly regulated interplay between HSPs and HSF1. As we also show that HSP27 binds to the SUMO-E2-conjugating enzyme, Ubc9, our study raises the possibility that HSP27 may act as a SUMO-E3 ligase specific for SUMO-2/3.


Subject(s)
DNA-Binding Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Ubiquitins/metabolism , Animals , Cell Nucleus/metabolism , HSP27 Heat-Shock Proteins/chemistry , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Proteins , Humans , Molecular Chaperones , Protein Multimerization , Protein Structure, Quaternary , Protein Transport , Substrate Specificity , Transcriptional Activation
8.
Oncogene ; 20(2): 260-9, 2001 Jan 11.
Article in English | MEDLINE | ID: mdl-11313953

ABSTRACT

Procaspase-2 is one of the cysteine aspartate proteases involved in apoptotic cell death. Alternative splicing of CASP-2 messenger RNA generates a long isoform, procaspase-2L, whose overexpression induces cell death and a truncated isoform, procaspase-2S, whose function remains poorly defined. The present study explored the consequences of procaspase-2S overexpression in U937 human leukemic cells exposed to the topoisomerase II inhibitor etoposide as an apoptotic stimulus. Overexpression of procaspase-2S in U937 cells partially prevented nuclear changes associated with etoposide-induced cell death, as determined by Hoechst 33342 staining of nuclear chromatin and electron microscopy studies. Procaspase-2S also prevented the maturation of apoptotic bodies, delayed phosphatidylserine externalization on the plasma membrane and prevented the cleavage and activation of procaspase-2L. These effects were not observed when the cysteine 289 in the consensus QACRG motif was mutated into a serine. Wild-type procaspase-2S overexpression did not influence the cleavage of procaspase-3, procaspase-7 and poly(ADP-ribose)polymerase nor the fragmentation of nuclear DNA into nucleosome-sized fragments. Altogether, these results indicate that the short isoform of procaspase-2 negatively interferes with selective features of apoptosis, an activity that is suppressed by mutation of the cysteine 289.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Chromatin/ultrastructure , Enzyme Precursors/metabolism , Phosphatidylserines/metabolism , Amino Acid Motifs , Apoptosis/drug effects , Base Sequence , Caspase 2 , Caspases/genetics , Cysteine , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Enzyme Precursors/genetics , Etoposide/pharmacology , Humans , Isoenzymes , Leukemia , Molecular Sequence Data , Mutation , Topoisomerase II Inhibitors , Transfection , Tumor Cells, Cultured
9.
Cancer Res ; 61(4): 1645-51, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245478

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a new cytokine that was proposed to specifically induce apoptosis of cancer cells. In tumor cells that are resistant to the cytokine, subtoxic concentrations of chemotherapeutic drugs can restore the response to TRAIL. The present study further explores the mechanisms that determine tumor cell sensitivity to TRAIL by comparing four human colon carcinoma cell lines We show that colon cancer cell sensitivity to TRAIL-induced apoptosis and cytotoxicity correlates with the expression of the death receptors TRAIL-R1 and TRAIL-R2 at the cell surface, as determined by now cytometry, whereas the two decoy receptors TRAIL-R3 and TRAIL-R4 can be detected only in permeabilized cells. Clinically relevant concentrations of cisplatin and doxorubicin sensitize the most resistant colon cancer cell lines to TRAIL-induced cell death without modifying the expression nor the localization of TRAIL receptors in these cells. TRAIL induces the activation of procaspase-8 and triggers caspase-dependent apoptosis off colon cancer cells. Cytotoxic drugs lower the signaling threshold required for TRAIL-induced procaspase-8 activation. In turn, caspase-8 cleaves Bid, a BH3 domain-containing proapoptotic molecule of the Bcl-2 family and activates effector caspases. Together, these data indicate that chemotherapeutic drugs sensitize colon tumor cells to TRAIL-mediated caspase-8 activation and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Colonic Neoplasms/pathology , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caspase 3 , Caspase 8 , Caspase 9 , Cell Membrane/metabolism , Cisplatin/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Doxorubicin/pharmacology , Drug Synergism , Enzyme Activation/drug effects , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Solubility , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
10.
Blood ; 97(6): 1835-44, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238127

ABSTRACT

Engagement of the plasma membrane receptor Fas can induce apoptosis of leukemic cells. Signaling through Fas requires the formation of a death-inducing signaling complex (DISC) that involves the cytoplasmic domain of Fas, the adaptor molecule FADD/MORT-1, and procaspase-8. The present study investigated whether another caspase, known as procaspase-2L, played a role in Fas-mediated cell death. A series of human leukemic variant cells was derived by stable transfection with a CASP2L antisense construct (CASP2L/AS). Specific down-regulation of procaspase-2L decreased the sensitivity of these cells to apoptosis induced by an agonistic anti-Fas antibody (Ab, clone CH11), as determined by studying DNA fragmentation, chromatin condensation, and externalization of phosphatidylserine on the plasma membrane. In leukemic cells transfected with an empty vector, anti-Fas Ab treatment activated caspase-8, decreased the expression of the BH3 domain-only protein Bid, triggered the release of cytochrome c from the mitochondria to the cytosol, and activated caspase-3. All these events could not be observed when CASP2L/AS cells were similarly treated with anti-Fas Abs. CASP2L/AS transfection did not inhibit the formation of the DISC and no direct interaction between procaspase-2L and either Fas or FADD or procaspase-8 was identified. Down-regulation of procaspase-2L inhibited anti-Fas Ab-mediated cleavage of c-FLIP (FLICE-inhibitory protein), a protein that interferes with the formation of a functional DISC. These results suggest that the long isoform of caspase-2 plays a role in the Fas-mediated pathway to cell death by contributing to caspase-8 activation at the DISC level.


Subject(s)
Apoptosis/drug effects , Caspases/physiology , Intracellular Signaling Peptides and Proteins , Leukemia/drug therapy , fas Receptor/pharmacology , BH3 Interacting Domain Death Agonist Protein , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Caspase 2 , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/drug effects , Caspases/genetics , Caspases/metabolism , Caspases/pharmacology , Cytochrome c Group/drug effects , Cytochrome c Group/metabolism , DNA, Antisense/pharmacology , Humans , Leukemia/pathology , Protein Isoforms/genetics , Protein Isoforms/pharmacology , Protein Isoforms/physiology , Transfection , Tumor Cells, Cultured
11.
J Immunol ; 164(10): 5023-7, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799856

ABSTRACT

The counterattack hypothesis, suggesting that cancer cells express Fas ligand (FasL) and are able to kill Fas-expressing tumor-infiltrating activated T cells, was supported by reports of the killing of Jurkat cells by FasL-expressing human colon cancer cell lines. Through the use of an improved cytotoxic assay in which soluble FasL and FasL-transfected KFL9 cells were used as positive controls, we show that none of seven human colon cancer cell lines induce apoptosis of two Fas-expressing target cell lines, Jurkat and L1210-Fas cells. Moreover, in coculture experiments, cancer cell monolayers do not inhibit the growth of Fas-expressing lymphoid cells. Although FasL mRNA and protein were detected in the extracts of the colon cancer cell lines, flow cytometry and confocal microscopy failed to detect the protein on the surface of tumor cells. These results suggest that the counterattack of tumor-infiltrating T lymphocytes by cancer cells may not account for immune tolerance toward tumor cells.


Subject(s)
Apoptosis/immunology , Colonic Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , fas Receptor/physiology , Animals , Antigens, Neoplasm/analysis , Antigens, Surface/analysis , Caco-2 Cells , Cell Division/immunology , Coculture Techniques , Colonic Neoplasms/metabolism , Fas Ligand Protein , Growth Inhibitors/immunology , HT29 Cells , Humans , Jurkat Cells , Leukemia L1210 , Membrane Glycoproteins/analysis , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Signal Transduction/immunology , fas Receptor/metabolism
12.
Anticancer Res ; 20(2A): 849-52, 2000.
Article in English | MEDLINE | ID: mdl-10810365

ABSTRACT

We have previously shown that p27KiP1 plays a role in the tumor cell resistance of HT29 confluent monolayers to cytotoxic drugs in vitro. To determine whether p27KiP1 was a resistance factor to cytotoxic drugs in vivo we tested the effect of doxorubicin on p27KiP1-overexpressing HT29 tumors in nude mice. In this study we show that ectopic overexpression of p27KiP1 in HT29 human colon cancer cells decreases their tumorigenicity in vivo in nude mice. This decreased tumor growth was associated with increased p27KiP1 protein expression, studied by Western blotting in tumor extracts. Interestingly, the overexpressing-p27KiP1 tumors were significantly more resistant to intraveneous doxorubicin treatment than the control tumors. These results indicate that p27KiP1, which delays tumor growth could also increase tumor resistance to cytotoxic drugs in vivo.


Subject(s)
Cell Cycle Proteins , Cell Division/drug effects , Doxorubicin/toxicity , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Animals , Cyclin-Dependent Kinase Inhibitor p27 , Doxorubicin/therapeutic use , Enzyme Inhibitors/metabolism , HT29 Cells , Humans , Mice , Mice, Nude , Microtubule-Associated Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
13.
FASEB J ; 13(14): 2061-70, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10544189

ABSTRACT

We have previously shown that the small heat shock protein HSP27 inhibited apoptotic pathways triggered by a variety of stimuli in mammalian cells. The present study demonstrates that HSP27 overexpression decreases U937 human leukemic cell sensitivity to etoposide-induced cytotoxicity by preventing apoptosis. As observed for Bcl-2, HSP27 overexpression delays poly(ADP-ribose)polymerase cleavage and procaspase-3 activation. In contrast with Bcl-2, HSP27 overexpression does not prevent etoposide-induced cytochrome c release from the mitochondria. In a cell-free system, addition of cytochrome c and dATP to cytosolic extracts from untreated cells induces the proteolytic activation of procaspase-3 in both control and bcl-2-transfected U937 cells but fails to activate procaspase-3 in HSP27-overexpressing cells. Immunodepletion of HSP27 from cytosolic extracts increases cytochrome c/dATP-mediated activation of procaspase-3. Overexpression of HSP27 also prevents procaspase-9 activation. In the cell-free system, immunodepletion of HSP27 increases LEDH-AFC peptide cleavage activity triggered by cytochrome c/dATP treatment. We conclude that HSP27 inhibits etoposide-induced apoptosis by preventing cytochrome c and dATP-triggered activity of caspase-9, downstream of cytochrome c release.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cytochrome c Group/physiology , Enzyme Precursors/antagonists & inhibitors , Etoposide/pharmacology , Heat-Shock Proteins/physiology , Caspase 3 , Caspase 9 , Deoxyadenine Nucleotides/physiology , Drug Resistance, Neoplasm , Enzyme Activation , Humans , Proto-Oncogene Proteins c-bcl-2/physiology , U937 Cells
14.
J Biol Chem ; 274(12): 7987-92, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075697

ABSTRACT

Trimerization of the Fas receptor (CD95, APO-1), a membrane bound protein, triggers cell death by apoptosis. The main death pathway activated by Fas receptor involves the adaptor protein FADD (for Fas-associated death domain) that connects Fas receptor to the caspase cascade. Anticancer drugs have been shown to enhance both Fas receptor and Fas ligand expression on tumor cells. The contribution of Fas ligand-Fas receptor interactions to the cytotoxic activity of these drugs remains controversial. Here, we show that neither the antagonistic anti-Fas antibody ZB4 nor the Fas-IgG molecule inhibit drug-induced apoptosis in three different cell lines. The expression of Fas ligand on the plasma membrane, which is identified in untreated U937 human leukemic cells but remains undetectable in untreated HT29 and HCT116 human colon cancer cell lines, is not modified by exposure to various cytotoxic agents. These drugs induce the clustering of Fas receptor, as observed by confocal laser scanning microscopy, and its interaction with FADD, as demonstrated by co-immunoprecipitation. Overexpression of FADD by stable transfection sensitizes tumor cells to drug-induced cell death and cytotoxicity, whereas down-regulation of FADD by transient transfection of an antisense construct decreases tumor cell sensitivity to drug-induced apoptosis. These results were confirmed by transient transfection of constructs encoding either a FADD dominant negative mutant or MC159 or E8 viral proteins that inhibit the FADD/caspase-8 pathway. These results suggest that drug-induced cell death involves the Fas/FADD pathway in a Fas ligand-independent fashion.


Subject(s)
Antigens, Surface/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arabidopsis Proteins , Fatty Acid Desaturases/metabolism , Membrane Glycoproteins/metabolism , Plant Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , fas Receptor/metabolism , Cisplatin/pharmacology , Etoposide/pharmacology , Fas Ligand Protein , Fatty Acid Desaturases/biosynthesis , Fatty Acid Desaturases/genetics , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Jurkat Cells , Ligands , Oligonucleotides, Antisense/metabolism , Plant Proteins/biosynthesis , Plant Proteins/genetics , Transfection , Tumor Cells, Cultured , U937 Cells , Vinblastine/pharmacology
15.
Biochem Biophys Res Commun ; 256(3): 603-7, 1999 Mar 24.
Article in English | MEDLINE | ID: mdl-10080945

ABSTRACT

We have previously shown that treatment by anticancer drugs sensitized tumor cells to Fas (APO-1/CD95)-mediated cell death. The present study demonstrates that the cytotoxic drugs cisplatin, doxorubicin and mitomycin C induce the accumulation of the Fas receptor, the FADD adaptor molecule, the procaspases-8, -3 and -2L and the proapoptotic molecule Bax in several human colon cancer cells. This upregulation is also observed in U3A myeloblastoma cells that do not express STAT-1, a transcription factor involved in the constitutive expression of procaspases. We conclude that anticancer drugs sensitize tumor cells to Fas-mediated cell death by a STAT-1-independent upregulation of molecules involved in this apoptotic pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Caspases/metabolism , Colonic Neoplasms/metabolism , DNA-Binding Proteins/physiology , Enzyme Precursors/metabolism , Proto-Oncogene Proteins c-bcl-2 , Trans-Activators/physiology , Up-Regulation/drug effects , Apoptosis/drug effects , Carrier Proteins/genetics , Caspase 2 , Caspase 3 , Caspase 8 , Caspase 9 , Cisplatin/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Dactinomycin/pharmacology , Doxorubicin/pharmacology , Fas-Associated Death Domain Protein , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoenzymes/metabolism , Mitomycin/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/physiology , STAT1 Transcription Factor , Trans-Activators/genetics , Transcription, Genetic , Tumor Cells, Cultured , bcl-2-Associated X Protein , fas Receptor
16.
Int J Cancer ; 77(5): 796-802, 1998 Aug 31.
Article in English | MEDLINE | ID: mdl-9688315

ABSTRACT

We have previously shown that growth of HT29 human colorectal cancer cells at confluence increased their resistance to the cytotoxic agent cisplatin. This study further explores the mechanisms of this resistance phenotype. DNA platination induced by cisplatin exposure is slightly reduced by confluence. However, at an equivalent DNA platination level, non-confluent cells accumulate in the G2/M phase of the cell cycle, demonstrate aberrant mitotic figures and die by apoptosis, while confluent cells progress slowly through the cell cycle, do not reach mitosis and are more resistant to drug-induced cell death. At a molecular level, cisplatin enhances cyclin B and p34cdc2 levels and histone H1 kinase activity in non-confluent, but not in confluent, cells. Furthermore, when HT29 cells reach confluence, expression of the cyclin-dependent kinase inhibitor p27Kip1 increases and cells accumulate in the G0/G1 phase of the cell cycle. Transfection-mediated over-expression of p27Kip1 in non-confluent HT29 cells decreases the cytotoxic activity of cisplatin as well as its ability to trigger apoptosis. Non-confluent HT29 cells over-expressing p27Kip1 are also more resistant to doxorubicin, etoposide and 5-fluorouracil. Our results suggest that p27Kip1 contributes to the confluence-dependent resistance phenotype.


Subject(s)
Cell Cycle Proteins , Cell Cycle/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm , Microtubule-Associated Proteins/metabolism , Tumor Suppressor Proteins , Apoptosis/drug effects , Cell Adhesion , Cell Cycle/drug effects , Cell Line , Cisplatin/toxicity , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Inhibitors/metabolism , G2 Phase , Genes, p53 , HT29 Cells , Humans , Microtubule-Associated Proteins/biosynthesis , Mitosis , Point Mutation , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Transfection , Tumor Suppressor Protein p53/genetics
17.
Cancer Res ; 57(13): 2661-7, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9205074

ABSTRACT

Resistance of colorectal cancer cells to chemotherapeutic drugs increases as cells reach confluence. Here we show that the small stress protein HSP27, which has been described to block necrotic and apoptotic cell death, accumulates in confluent human colorectal cancer cell lines HT-29 and Caco2. Cell confluence also induces HSP27 phosphorylation and changes in its intracellular distribution. We also show that overexpression of human HSP27 by transfection of HT-29 cells increased the resistance of cells to doxorubicin or cisplatin and prevented drug-induced apoptosis. Interestingly, nonconfluent HSP27-transfected cells and confluent control cells in which HSP27 is expressed at the same level displayed a similar drug resistance. HSP27-transfected cells did not exhibit an enhanced resistance when they reached confluence, nor was there an increased accumulation of HSP27. We have previously shown that HSP27 expression blocks tumor necrosis factor-induced cell death as a result of decreasing intracellular reactive oxygen species (ROS). Here we show that HSP27 overexpression in HT-29 cells, obtained either by transfection or by growing the cells at high density, correlated with a significant ROS decrease. We conclude that cell confluent-dependent HSP27 accumulation, probably due to its ability to decrease ROS levels, is essential for the establishment of the resistance of colorectal cancer cells when reaching confluence.


Subject(s)
Apoptosis , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm , Protein Serine-Threonine Kinases/metabolism , Cell Count , Cisplatin/pharmacology , Colorectal Neoplasms/pathology , Culture Media, Serum-Free/metabolism , Doxorubicin/pharmacology , HT29 Cells , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Subcellular Fractions/metabolism , Transfection , Tumor Cells, Cultured
18.
J Neurosci Methods ; 75(1): 49-54, 1997 Jul 18.
Article in English | MEDLINE | ID: mdl-9262143

ABSTRACT

Neurological dysfunction can be assessed by analysing footprint patterns and walking tracks. However, because such an analysis is very time consuming, we developed an MS-Windows program called FOOTPRINTS which facilitates the analysis of the commonly used measures and which is considerably quicker than manual scoring methods. The prints are scanned at a resolution of 75 dpi and stored as black and white bitmaps for further analysis. In order to validate the program, we analysed the footprint patterns of mice and rats, using both the program and the conventional manual scoring method. In the first study, the walking patterns of 3-, 14-, and 26-month-old Janvier Wistar rats were compared, and in the second the footprint patterns of C57BL mice were assessed. Comparison of the data obtained using the program and of the data obtained by manual scoring showed that the computer-based analysis gives reliable results. The program saves considerable time as the analysis took 1/8th of the time needed for manual evaluation.


Subject(s)
Cost Control , Foot/anatomy & histology , Software , Walking , Analysis of Variance , Animals , Evaluation Studies as Topic , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Reproducibility of Results
19.
J Natl Cancer Inst ; 89(11): 783-9, 1997 Jun 04.
Article in English | MEDLINE | ID: mdl-9182976

ABSTRACT

BACKGROUND: The transmembrane receptor Fas, together with its protein-binding partner (Fas ligand), is a key regulator of programmed cell death (i.e., apoptosis). Fas and Fas ligand also influence the ability of cytotoxic T lymphocytes and natural killer cells to eliminate tumor cells. However, by inducing apoptosis in activated T cells, the Fas/Fas ligand system may protect some tumor cells from clearance by the immune system. Anticancer drugs enhance Fas ligand expression on the surface of Fas receptor-expressing leukemia cells, thus suggesting that apoptosis caused by these drugs may be mediated via the Fas/Fas ligand system. PURPOSE: This study was conducted to further investigate the relationship between the modulation of Fas receptor gene and protein expression by treatment of cells with cytotoxic drugs and the immune clearance of tumor cells. METHODS: Fas expression on human HT29 colon carcinoma cells treated with a variety of anticancer drugs (cisplatin, doxorubicin, mitomycin C, fluorouracil, and camptothecin) was analyzed by use of quantitative flow cytometry. Human HCT8R and HCT116 colon carcinoma cells and human U937 leukemia cells were treated with cisplatin only and analyzed in the same way. Fas ligand messenger RNA and protein levels were studied by use of a reverse transcription-polymerase chain reaction assay and by flow cytometry. Fas gene expression and messenger RNA levels in cisplatin-treated HT29 cells were characterized by use of in vitro nuclear run-on and northern blot hybridization assays. The cytotoxic activities of agonistic anti-Fas antibodies, Fas ligand, and allogeneic peripheral blood leukocytes, in the absence or presence of Fas-blocking monoclonal antibodies, against tumor cells were assessed by methylene blue staining and chromium-51 release assays. RESULTS: Clinically relevant concentrations of cisplatin, doxorubicin, mitomycin C, fluorouracil, or camptothecin enhanced Fas receptor expression on the plasma membrane of HT29 cells. Cisplatin-mediated increases in Fas expression were confirmed in HCT8R, HCT116, and U937 cells. The enhancement of Fas protein expression was associated with an increased sensitivity of cisplatin-treated tumor cells to agonistic anti-Fas antibodies, to soluble Fas ligand, and to allogeneic peripheral blood leukocyte-mediated cytotoxicity. Each of these effects was blocked by co-treatment of the cells with antagonistic anti-Fas antibody. CONCLUSION AND IMPLICATIONS: In addition to their direct cytotoxic effects, chemotherapeutic drugs sensitize tumor cells to Fas-mediated cytotoxicity and Fas-dependent immune clearance. On the basis of these findings, new strategies might be developed to improve the efficacy of these drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Membrane Glycoproteins/physiology , fas Receptor/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cisplatin/pharmacology , Drug Screening Assays, Antitumor , Fas Ligand Protein , Flow Cytometry , HT29 Cells/drug effects , HT29 Cells/metabolism , Humans , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Mice , RNA, Messenger/metabolism , Tumor Cells, Cultured , fas Receptor/drug effects , fas Receptor/metabolism
20.
Eur J Biochem ; 237(3): 653-9, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8647109

ABSTRACT

To investigate the role of the small 27-kDa heat-shock protein (Hsp27) in the intrinsic resistance of colon cancer cells to doxorubicin, we modified Hsp27 expression either genetically by transfection or pharmacologically by cisplatin treatment. HT-29 cells were transfected with a full-length Hsp27 construct in the sense or antisense orientation. We found a good correlation between cell survival after doxorubicin treatment and Hsp27 content. A similar correlation was found for the thermoresistance of the Hsp27-transfected cells. In contrast, the sensitivity of the different transfected cells to 5-fluorouracil was not modified. cis-Platinum(II)diammine dichloride (cisplatin) treatment of HT-29 or Caco2 cells dramatically increased their Hsp27 mRNA and protein content. Accordingly, the cells became thermoresistant. Contrary to what has been previously assumed, however, cell resistance to doxorubicin was reduced. Our data suggest that the decreased resistance of the cells to doxorubicin may be due to a concomitant increase of topoisomerase II expression, the main target of anthracyclines. In conclusion, although Hsp27 seems to participate in the natural resistance of colon cancer cells to anthracyclines, its increase after cisplatin treatment is not associated with a decreased cytotoxicity to doxorubicin.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Doxorubicin/pharmacology , Heat-Shock Proteins/metabolism , Cell Survival/drug effects , Cisplatin/pharmacology , DNA Topoisomerases, Type II/metabolism , Drug Resistance/physiology , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Hot Temperature , Humans , Molecular Weight , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...