Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(8): 785, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381018

ABSTRACT

Pediatric gliomas comprise a broad range of brain tumors derived from glial cells. While high-grade gliomas are often resistant to therapy and associated with a poor outcome, children with low-grade gliomas face a better prognosis. However, the treatment of low-grade gliomas is often associated with severe long-term adverse effects. This shows that there is a strong need for improved treatment approaches. Here, we highlight the potential for repurposing disulfiram to treat pediatric gliomas. Disulfiram is a drug used to support the treatment of chronic alcoholism and was found to be effective against diverse cancer types in preclinical studies. Our results show that disulfiram efficiently kills pediatric glioma cell lines as well as patient-derived glioma stem cells. We propose a novel mechanism of action to explain disulfiram's anti-oncogenic activities by providing evidence that disulfiram induces the degradation of the oncoprotein MLL. Our results further reveal that disulfiram treatment and MLL downregulation induce similar responses at the level of histone modifications and gene expression, further strengthening that MLL is a key target of the drug and explaining its anti-oncogenic properties.


Subject(s)
Alcoholism/drug therapy , Disulfiram/therapeutic use , Glioma/drug therapy , Glioma/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Proteolysis , Auranofin/pharmacology , Auranofin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Disulfiram/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Repositioning , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/pathology , Histones/metabolism , Humans , Lysine/metabolism , Methylation/drug effects , Neoplasm Grading , Protein Processing, Post-Translational/drug effects , Proteolysis/drug effects , Transcription, Genetic/drug effects
2.
Cell Death Discov ; 7(1): 87, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33931592

ABSTRACT

To sustain their malignancy, tumour cells acquire several metabolic adaptations such as increased oxygen, glucose, glutamine, and lipids uptake. Other metabolic processes are also enhanced as part of tumour metabolic reprogramming, for example, increased serine metabolism. Serine is a non-essential amino acid that supports several metabolic processes that are crucial for the growth and survival of proliferating cells, including protein, DNA, and glutathione synthesis. Indeed, increased activity of D-3-phosphoglycerate dehydrogenase (PHGDH), the enzyme rate-limiting de novo serine synthesis, has been extensively reported in several tumours. Therefore, selective inhibition of PHGDH may represent a new therapeutic strategy for over-expressing PHGDH tumours, owing to its downstream inhibition of essential biomass production such as one-carbon units and nucleotides. This perspective article will discuss the current status of research into small molecular inhibitors against PHGDH in colorectal cancer, breast cancer, and Ewing's sarcoma. We will summarise recent studies on the development of PHGDH-inhibitors, highlighting their clinical potential as new therapeutics. It also wants to shed a light on some of the key limitations of the use of PHGDH-inhibitors in cancer treatment which are worth taking into account.

3.
Cell Death Dis ; 10(11): 785, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619667

ABSTRACT

Medulloblastoma (MB) is the most common malignant solid paediatric brain tumour. The standard treatment for MB is surgical resection of the tumour, radiation and chemotherapy. This therapy is associated with high morbidity and adverse side effects. Hence, more targeted and less toxic therapies are vitally needed to improve the quality of life of survivors. NPI-0052 is a novel proteasome inhibitor that irreversibly binds the 20S proteasome subunit. This compound has anti-tumour activity in metastatic solid tumours, glioblastoma and multiple myeloma with a good safety profile. Importantly, NPI-0052 has a lipophilic structure and can penetrate the blood-brain barrier, making it a suitable treatment for brain tumours. In the present study, we performed an in silico gene expression analysis to evaluate the proteasome subunit expression in MB. To evaluate the anticancer activity of NPI-0052, we used a range of MB patient-derived MB cells and cell lines. The synergistic cell death of NPI-0052 with γ-radiation was evaluated in tumour organoids derived from patient-derived MB cells. We show that high expression of proteasome subunits is a poor prognostic factor for MB patients. Also, our preclinical work demonstrated that NPI-0052 can inhibit proteasome activity and activate apoptosis in MB cells. Moreover, we observe that NPI-0052 has a synergistic apoptotic effect with γ-radiation, a component of the current MB therapy. Here, we present compelling preclinical evidence that NPI-0052 can be used as an adjuvant treatment for p53-family-expressing MB tumours.


Subject(s)
Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/radiotherapy , Gamma Rays/therapeutic use , Lactones/pharmacology , Medulloblastoma/drug therapy , Medulloblastoma/radiotherapy , Pyrroles/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cerebellar Neoplasms/pathology , Chemoradiotherapy , Humans , Medulloblastoma/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology
4.
Genes Dev ; 31(17): 1738-1753, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28971956

ABSTRACT

Medulloblastoma is the most common solid primary brain tumor in children. Remarkable advancements in the understanding of the genetic and epigenetic basis of these tumors have informed their recent molecular classification. However, the genotype/phenotype correlation of the subgroups remains largely uncharacterized. In particular, the metabolic phenotype is of great interest because of its druggability, which could lead to the development of novel and more tailored therapies for a subset of medulloblastoma. p73 plays a critical role in a range of cellular metabolic processes. We show overexpression of p73 in a proportion of non-WNT medulloblastoma. In these tumors, p73 sustains cell growth and proliferation via regulation of glutamine metabolism. We validated our results in a xenograft model in which we observed an increase in survival time in mice on a glutamine restriction diet. Notably, glutamine starvation has a synergistic effect with cisplatin, a component of the current medulloblastoma chemotherapy. These findings raise the possibility that glutamine depletion can be used as an adjuvant treatment for p73-expressing medulloblastoma.


Subject(s)
Cerebellar Neoplasms/diet therapy , Cerebellar Neoplasms/physiopathology , Glutamine/metabolism , Medulloblastoma/diet therapy , Medulloblastoma/physiopathology , Tumor Protein p73/genetics , Tumor Protein p73/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Glutaminase/genetics , Glutaminase/metabolism , Heterografts , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Tumor Cells, Cultured
5.
Cell Rep ; 20(2): 411-426, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28700942

ABSTRACT

Cell migration through the brain parenchyma underpins neurogenesis and glioblastoma (GBM) development. Since GBM cells and neuroblasts use the same migratory routes, mechanisms underlying migration during neurogenesis and brain cancer pathogenesis may be similar. Here, we identify a common pathway controlling cell migration in normal and neoplastic cells in the CNS. The nuclear scaffold protein promyelocytic leukemia (PML), a regulator of forebrain development, promotes neural progenitor/stem cell (NPC) and neuroblast migration in the adult mouse brain. The PML pro-migratory role is active also in transformed mouse NPCs and in human primary GBM cells. In both normal and neoplastic settings, PML controls cell migration via Polycomb repressive complex 2 (PRC2)-mediated repression of Slits, key regulators of axon guidance. Finally, a PML/SLIT1 axis regulates sensitivity to the PML-targeting drug arsenic trioxide in primary GBM cells. Taken together, these findings uncover a drug-targetable molecular axis controlling cell migration in both normal and neoplastic cells.


Subject(s)
Central Nervous System/metabolism , Promyelocytic Leukemia Protein/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Central Nervous System/cytology , Glioblastoma/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice , Neurogenesis/genetics , Neurogenesis/physiology , Nuclear Lamina/metabolism
6.
Neuron ; 74(1): 122-35, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22500635

ABSTRACT

Activity-dependent modifications of chromatin are believed to contribute to dramatic changes in neuronal circuitry. The mechanisms underlying these modifications are not fully understood. The histone variant H3.3 is incorporated in a replication-independent manner into different regions of the genome, including gene regulatory elements. It is presently unknown whether H3.3 deposition is involved in neuronal activity-dependent events. Here, we analyze the role of the histone chaperone DAXX in the regulation of H3.3 incorporation at activity-dependent gene loci. DAXX is found to be associated with regulatory regions of selected activity-regulated genes, where it promotes H3.3 loading upon membrane depolarization. DAXX loss not only affects H3.3 deposition but also impairs transcriptional induction of these genes. Calcineurin-mediated dephosphorylation of DAXX is a key molecular switch controlling its function upon neuronal activation. Overall, these findings implicate the H3.3 chaperone DAXX in the regulation of activity-dependent events, thus revealing a new mechanism underlying epigenetic modifications in neurons.


Subject(s)
Carrier Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Potentials/physiology , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Calcineurin/metabolism , Calcium/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Co-Repressor Proteins , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Genes, Immediate-Early , Mice , Mice, Knockout , Molecular Chaperones , Nerve Net/growth & development , Nerve Net/metabolism , Nerve Net/microbiology , Neural Conduction/physiology , Transcription, Genetic/physiology
7.
J Natl Cancer Inst ; 101(11): 828-32, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19470951

ABSTRACT

Peptides that interfere with the natural resistance of cancer cells to genotoxin-induced apoptosis may improve the efficacy of anticancer regimens. We have previously reported that a cell-permeable RasGAP-derived peptide (TAT-RasGAP(317-326)) specifically sensitizes tumor cells to genotoxin-induced apoptosis in vitro. Here, we examined the in vivo stability of a protease-resistant D-form of the peptide, RI.TAT-RasGAP(317-326), and its effect on tumor growth in nude mice bearing subcutaneous human colon cancer HCT116 xenograft tumors. After intraperitoneal injection, RI.TAT-RasGAP(317-326) persisted in the blood of nude mice for more than 1 hour and was detectable in various tissues and subcutaneous tumors. Tumor-bearing mice treated daily for 7 days with RI.TAT-RasGAP(317-326) (1.65 mg/kg body weight) and cisplatin (0.5 mg/kg body weight) or doxorubicin (0.25 mg/kg body weight) displayed reduced tumor growth compared with those treated with either genotoxin alone (n = 5-7 mice per group; P = .004 and P = .005, respectively; repeated measures analysis of variance [ANOVA, two-sided]). This ability of the RI.TAT-RasGAP(317-326) peptide to enhance the tumor growth inhibitory effect of cisplatin was still observed at peptide doses that were at least 150-fold lower than the dose lethal to 50% of mice. These findings provide the proof of principle that RI.TAT-RasGAP(317-326) may be useful for improving the efficacy of chemotherapy in patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , GTPase-Activating Proteins/pharmacology , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/administration & dosage , Cisplatin/pharmacology , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Drug Synergism , GTPase-Activating Proteins/administration & dosage , HCT116 Cells , Humans , Infusions, Parenteral , Mice , Mice, Nude , Molecular Sequence Data , Mutagens/pharmacology , Peptide Fragments/administration & dosage , Transplantation, Heterologous
8.
Cell Cycle ; 8(10): 1467-8, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19411830
9.
Exp Cell Res ; 315(12): 2081-91, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19328779

ABSTRACT

The specific sensitization of tumor cells to the apoptotic response induced by genotoxins is a promising way of increasing the efficacy of chemotherapies. The RasGAP-derived fragment N2, while not regulating apoptosis in normal cells, potently sensitizes tumor cells to cisplatin- and other genotoxin-induced cell death. Here we show that fragment N2 in living cells is mainly located in the cytoplasm and only minimally associated with specific organelles. The cytoplasmic localization of fragment N2 was required for its cisplatin-sensitization property because targeting it to the mitochondria or the ER abrogated its ability to increase the death of tumor cells in response to cisplatin. These results indicate that fragment N2 requires a spatially constrained cellular location to exert its anti-cancer activity.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Cytoplasm/metabolism , ras GTPase-Activating Proteins/metabolism , Amino Acid Motifs , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Cisplatin/pharmacology , Humans , Mice , ras GTPase-Activating Proteins/genetics
10.
J Photochem Photobiol B ; 88(1): 29-35, 2007 Jul 27.
Article in English | MEDLINE | ID: mdl-17560792

ABSTRACT

BACKGROUND: 5,10,15,20-Tetrakis(m-hydroxyphenyl)chlorin (mTHPC)-mediated photodynamic therapy (PDT) has shown insufficient tumor selectivity for the treatment of pleural mesothelioma. Tumor selectivity of mTHPC-PDT may be enhanced in the presence of the TAT-RasGAP(317-326) peptide which has the potential to specifically sensitize tumor cells to cytostatic agents. MATERIALS AND METHODS: H-meso-1 and human fibroblast cell cultures, respectively, were exposed to two different mTHPC doses followed by light delivery with and without TAT-RasGAP(317-326) administration. mTHPC was added to the cultures at a concentration of 0.04microg/ml and 0.10microg/ml, respectively, 24h before laser light illumination at 652nm (3J/cm(2), 40mW/cm(2)). TAT-RasGAP(317-326) was added to the cultures immediately after light delivery at a concentration of 20microM. The apoptosis rate was determined by scoring the cells displaying pycnotic nuclei. Cell viability was measured by using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. RESULTS: Light delivery associated with 0.04microg/ml mTHPC resulted in a significantly higher apoptosis rate in the presence of TAT-RasGAP(317-326) than without in H-meso-1 cells (p<0.05) but not in fibroblasts. In contrast, 1.0microg/ml mTHPC and light resulted in a significantly higher apoptosis rate in both H-meso-1 cells and fibroblasts as compared to controls (p<0.05) but the addition of TAT-RasGAP(317-326) did not lead to a further significant increase of the apoptosis rate of both H-meso-1 cells and fibroblasts as compared to mTHPC and light delivery alone. CONCLUSION: TAT-RasGAP(317-326) selectively enhanced the effect of mTHPC and light delivery on H-meso-1 cells but not on fibroblasts. However, this effect was mTHPC dose-dependent and occurred only at a low sensitizer dose.


Subject(s)
Apoptosis/drug effects , GTPase-Activating Proteins/therapeutic use , Mesoporphyrins/therapeutic use , Mesothelioma/drug therapy , Peptide Fragments/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Cell Survival , Fibroblasts/drug effects , GTPase-Activating Proteins/chemical synthesis , Humans , Mesothelioma/pathology , Peptide Fragments/chemical synthesis , Peptides/therapeutic use , Tumor Cells, Cultured
11.
Crit Rev Oncol Hematol ; 63(2): 160-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17544289

ABSTRACT

Agents that induce DNA damage in cells--the so-called genotoxins--have successfully been used for decades to treat patients with tumors. Genotoxins alter the DNA of cells, which is detected by DNA damage sensors and which leads to the activation of p53. Activation of p53 can lead to the death of cancer cells. The efficacy of genotoxins in humans is however limited by their toxicity to normal tissues. Specific sensitization of tumor cells to the action of genotoxins would reduce the efficacious doses of genotoxins to be used in patients, diminishing the detrimental side-effects of the drugs on normal tissues. A series of compounds able to sensitize cancer cells to DNA-damaging drugs have recently been identified that have the potential to increase the efficacy of currently used anti-cancer treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , DNA Damage/drug effects , Neoplasms/drug therapy , Drug Resistance, Neoplasm , Humans , Neoplasms/metabolism , Neoplasms/pathology
12.
Mol Cancer Res ; 5(5): 497-507, 2007 May.
Article in English | MEDLINE | ID: mdl-17510315

ABSTRACT

Although chemotherapy has revolutionized cancer treatment, the associated side effects induced by lack of specificity to tumor cells remain a challenging problem. We have previously shown that TAT-RasGAP(317-326),a cell-permeable peptide derived from RasGAP, specifically sensitizes cancer cells to the action of genotoxins. The underlying mechanisms of this sensitization were not defined however. Here, we report that TAT-RasGAP(317-326) requires p53, but not the Ras effectors Akt and extracellular signal-regulated kinase, to mediate its tumor sensitization abilities. The TAT-RasGAP(317-326) peptide, although not modulating the transcriptional activity of p53 or its phosphorylation and acetylation status, nevertheless requires a functional p53 cellular status to increase the sensitivity of tumor cells to genotoxins. Genes regulated by p53 encode proapoptotic proteins, such as PUMA, and cell cycle control proteins, such as p21. The ability of TAT-RasGAP(317-326) to sensitize cancer cells was found to require PUMA but not p21. TAT-RasGAP(317-326) did not affect PUMA levels, however, but increased genotoxin-induced mitochondrial depolarization and caspase-3 activation. These results indicate that TAT-RasGAP(317-326) sensitizes tumor cells by activating signals that intersect with the p53 pathway downstream of, or at the level of, proapoptotic p53 target gene products to increase the activation of the mitochondrial death pathway.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Gene Products, tat/pharmacology , Mutagens/pharmacology , Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , ras GTPase-Activating Proteins/pharmacology , Acetylation/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Products, tat/chemistry , HCT116 Cells , Humans , MAP Kinase Signaling System/drug effects , Neoplasms/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Thermodynamics , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/genetics , ras GTPase-Activating Proteins/chemistry
13.
Mol Biol Cell ; 16(8): 3511-20, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15901831

ABSTRACT

RasGAP bears two caspase-3 cleavage sites that are used sequentially as caspase activity increases in cells. When caspase-3 is mildly activated, RasGAP is first cleaved at position 455. This leads to the production of an N-terminal fragment, called fragment N, that activates the Ras-PI3K-Akt pathway and that promotes cell survival. At higher caspase activity, RasGAP is further cleaved at position 157 generating two small N-terminal fragments named N1 and N2. We have now determined the contribution of this second cleavage event in the regulation of apoptosis using cells in which the wild-type RasGAP gene has been replaced by a cDNA encoding a RasGAP mutant that cannot be cleaved at position 157. Our results show that cleavage of fragment N at position 157 leads to a marked reduction in Akt activity. This is accompanied by efficient processing of caspase-3 that favors cell death in response to various apoptotic stimuli. In nontumorigenic cells, fragments N1 and N2 do not modulate apoptosis. Therefore, the role of the second caspase-mediated cleavage of RasGAP is to allow the inactivation of the antiapoptotic function of fragment N so that caspases are no longer hampered in their ability to kill cells.


Subject(s)
Apoptosis , Caspases/metabolism , Mutation/genetics , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Caspase 3 , Cell Line , Down-Regulation , Enzyme Activation , Fibroblasts , Mice , Mice, Knockout , ras GTPase-Activating Proteins/deficiency
14.
Mol Cell Biol ; 24(23): 10425-36, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542850

ABSTRACT

Tight control of apoptosis is required for proper development and maintenance of homeostasis in multicellular organisms. Cells can protect themselves from potentially lethal stimuli by expressing antiapoptotic factors, such as inhibitors of apoptosis, FLICE (caspase 8)-inhibitory proteins, and members of the Bcl2 family. Here, we describe a mechanism that allows cells to survive once executioner caspases have been activated. This mechanism relies on the partial cleavage of RasGAP by caspase 3 into an amino-terminal fragment called fragment N. Generation of this fragment leads to the activation of the antiapoptotic Akt kinase, preventing further amplification of caspase activity. Partial cleavage of RasGAP is required for cell survival under stress conditions because cells expressing an uncleavable RasGAP mutant cannot activate Akt, cannot prevent amplification of caspase 3 activity, and eventually undergo apoptosis. Executioner caspases therefore control the extent of their own activation by a feedback regulatory mechanism initiated by the partial cleavage of RasGAP that is crucial for cell survival under adverse conditions.


Subject(s)
Caspases/metabolism , ras GTPase-Activating Proteins/chemistry , Animals , Apoptosis , Blotting, Western , Caspase 3 , Caspases/chemistry , Cell Line , Cell Survival , Cells, Cultured , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Electroporation , Enzyme Activation , Fibroblasts/metabolism , Humans , Image Processing, Computer-Assisted , Jurkat Cells , Lentivirus/genetics , Mice , Models, Genetic , Mutation , Plasmids/metabolism , Protein Structure, Tertiary , Stress, Physiological , Time Factors , Transfection , ras GTPase-Activating Proteins/metabolism
15.
Oncogene ; 23(55): 8971-8, 2004 Nov 25.
Article in English | MEDLINE | ID: mdl-15467750

ABSTRACT

Treatment of many cancers relies on the combined action of several genotoxins, but the detrimental effect of these drugs on normal cells can cause severe side effects. One major challenge in anticancer therapy is therefore to increase the selectivity of current treatments toward cancer cells in order to spare normal cells. We have recently demonstrated that a RasGAP caspase cleavage fragment is able to sensitize HeLa cells towards cisplatin-induced apoptosis. Here, we extend this observation by showing that this fragment also enhances cell death induced by adriamycin and mitoxantrone, two other widely used genotoxins. Furthermore, we have delineated a short sequence within this fragment that still bears the genotoxin-sensitization property. The peptide encoded by this sequence, when fused to the TAT cell permeation sequence, potently sensitized a number of tumors cells, but not normal cells, towards apoptosis induced by cisplatin, adriamycin and mitoxantrone. This sensitization effect was not mediated through modulation of NFkappaB activity or activation of the JNK and p38 MAPK pathways. Our results demonstrate the feasibility in enhancing the efficacy of currently used drugs to selectively kill cancer cells using peptides derived from pro-apoptotic caspase substrate fragments.


Subject(s)
GTPase-Activating Proteins/chemistry , Mutagens/metabolism , Peptide Fragments/chemistry , Peptides/chemistry , ras GTPase-Activating Proteins/metabolism , Apoptosis , Blotting, Western , Cell Line, Tumor , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Gene Products, tat/chemistry , Genes, Reporter , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Luciferases/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitoxantrone/pharmacology , NF-kappa B/metabolism , Plasmids/metabolism , Protein Structure, Tertiary , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism , src Homology Domains
16.
Biochem Pharmacol ; 68(6): 1027-31, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15313397

ABSTRACT

Executioner caspases induce the biochemical and cellular changes characteristic of apoptosis. Activation of caspases is therefore regarded as "the kiss of death" resulting in the cell's demise. Recent reports indicate however that in some situations, caspase activation may induce other responses than apoptosis. These findings raise the question of how cells manage to counteract the killing activities of executioner caspases. Experiments performed in our laboratory have unraveled a mechanism that allows cells to survive in the presence of activated executioner caspases. This mechanism is based on the partial cleavage of RasGAP into an N-terminal fragment that activates the Ras-PI3K-Akt survival pathway. This protective pathway may be activated to allow cells to use executioner caspases for other purposes than inducing apoptosis.


Subject(s)
Apoptosis/physiology , Caspases/physiology , ras GTPase-Activating Proteins/physiology , Animals , Caspases/metabolism , Cell Survival/physiology , Enzyme Activation , Humans , Phosphatidylinositol 3-Kinases/metabolism , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/therapeutic use , ras Proteins/metabolism
17.
Carcinogenesis ; 25(6): 909-21, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14742324

ABSTRACT

p120 RasGTPase-activating protein (RasGAP), the main regulator of Ras GTPase family members, is cleaved at low caspase activity into an N-terminal fragment that triggers potent anti-apoptotic signals via activation of the Ras/PI-3 kinase/Akt pathway. When caspase activity is increased, RasGAP fragment N is further processed into two fragments that effectively potentiate apoptosis. Expression of RasGAP protein and its cleavage was assessed in human lung cancer cells with different histology and responsiveness to anticancer drug-induced apoptosis. Here we show that therapy-sensitive small lung carcinoma cell (SCLC) lines have lower RasGAP expression levels and higher spontaneous cleavage with formation of fragment N compared to therapy-resistant non-small cell lung carcinoma cell (NSCLC) lines. The first RasGAP cleavage event strongly correlated with the increased level of spontaneous apoptosis in SCLC. However, generation of protective RasGAP fragment N also related to the potency of SCLC to develop secondary therapy-resistance. In response to etoposide (ET), RasGAP fragment N was further cleaved in direct dependence on caspase-3 activity, which was more pronounced in NSCLC cells. Caspase inhibition, while effectively preventing the second cleavage of RasGAP, barely affected the first cleavage of RasGAP into fragment N that was always detectable in NSCLC and SCLC cells. These findings suggest that different levels of RasGAP and fragment N might play a significant role in the biology and different clinical course of both subtypes of lung neoplasms. Furthermore, constitutive formation of RasGAP fragment N can potentially contribute to primary resistance of NSCLC to anticancer therapy by ET but also to secondary therapy-resistance in SCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/metabolism , Caspases/metabolism , Etoposide/pharmacology , Lung Neoplasms/metabolism , ras GTPase-Activating Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/enzymology , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Enzyme Activation , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...