Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(45): 73292-73308, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27689335

ABSTRACT

The moderate anticancer effect of arginine deprivation in clinical trials has been linked to an induced argininosuccinate synthetase (ASS1) expression in initially ASS1-negative tumors, and ASS1-positive cancers are anticipated as non-responders. Our previous studies indicated that arginine deprivation and low doses of the natural arginine analog canavanine can enhance radioresponse. However, the efficacy of the proposed combination in the presence of extracellular citrulline, the substrate for arginine synthesis by ASS1, remains to be elucidated, in particular for malignant cells with positive and/or inducible ASS1 as in colorectal cancer (CRC). Here, the physiological citrulline concentration of 0.05 mM was insufficient to overcome cell cycle arrest and radiosensitization triggered by arginine deficiency. Hyperphysiological citrulline (0.4 mM) did not entirely compensate for the absence of arginine and significantly decelerated cell cycling. Similar levels of canavanine-induced apoptosis were detected in the absence of arginine regardless of citrulline supplementation both in 2-D and advanced 3-D assays, while normal colon epithelial cells in organoid/colonosphere culture were unaffected. Notably, canavanine tremendously enhanced radiosensitivity of arginine-starved 3-D CRC spheroids even in the presence of hyperphysiological citrulline. We conclude that the novel combinatorial targeting strategy of metabolic-chemo-radiotherapy has great potential for the treatment of malignancies with inducible ASS1 expression.


Subject(s)
Arginine/metabolism , Canavanine/administration & dosage , Citrulline/metabolism , Radiation, Ionizing , Apoptosis/drug effects , Apoptosis/radiation effects , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , DNA Methylation/drug effects , DNA Methylation/radiation effects , Gene Expression , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Radiation Tolerance , Spheroids, Cellular , Tumor Cells, Cultured
2.
Exp Cell Res ; 341(1): 67-74, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26751966

ABSTRACT

Tumor cells rely on a continued exogenous nutrient supply in order to maintain a high proliferative activity. Although a strong dependence of some tumor types on exogenous arginine sources has been reported, the mechanisms of arginine sensing by tumor cells and the impact of changes in arginine availability on translation and cell cycle regulation are not fully understood. The results presented herein state that human colorectal carcinoma cells rapidly exhaust the internal arginine sources in the absence of exogenous arginine and repress global translation by activation of the GCN2-mediated pathway and inhibition of mTOR signaling. Tumor suppressor protein p53 activation and G1/G0 cell cycle arrest support cell survival upon prolonged arginine starvation. Cells with the mutant or deleted TP53 fail to stop cell cycle progression at defined cell cycle checkpoints which appears to be associated with reduced recovery after durable metabolic stress triggered by arginine withdrawal.


Subject(s)
Arginine/metabolism , Cell Cycle , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Protein Biosynthesis , HCT116 Cells , HT29 Cells , Humans , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
3.
Int J Biochem Cell Biol ; 70: 29-38, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26546743

ABSTRACT

Deprivation for the single amino acid arginine is a rapidly developing metabolic anticancer therapy, which allows growth control in a number of highly malignant tumors. Here we report that one of the responses of human solid cancer cells to arginine starvation is the induction of prolonged endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR). Systematic study of two colorectal carcinoma HCT-116 and HT29, glioblastoma U251 MG and ovarian carcinoma SKOV3 cell lines revealed, however, that the ER stress triggered by the absence of arginine does not result in massive apoptosis despite a profound upregulation of the proapoptotic gene CHOP. Instead, Akt- and MAPK-dependent pathways were activated which may counteract proapoptotic signaling. Treatment with DMSO as a disaggregating agent or with cycloheximide to block protein synthesis reduced ER stress evoked by arginine deprivation. On the other hand, ER stress and apoptosis induction in arginine-starved cells could be critically augmented by the arginine analog of plant origin canavanine, but not by the classic ER stress inducer tunicamycin. Our data suggest that canavanine treatment applied under the lack of arginine may enhance the efficacy of arginine deprivation-based anticancer therapy.


Subject(s)
Arginine/deficiency , Culture Media/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/drug effects , Gene Expression Regulation, Neoplastic , Canavanine/pharmacology , Cell Line, Tumor , Cycloheximide/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimethyl Sulfoxide/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , HCT116 Cells , HT29 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organ Specificity , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects
4.
Amino Acids ; 45(5): 1221-30, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036986

ABSTRACT

Increased amino acid requirement of malignant cells is exploited in metabolic antitumor therapy, e.g., enzymotherapies based on arginine or methionine deprivation. However, studies on animal models and clinical trials revealed that solid tumors are much less susceptible to single amino acid starvation than could be expected from the in vitro data. We conducted a comparative analysis of the response of several tumor cell lines to single amino acid starvation in 2-D monolayer versus 3-D spheroid culture. We revealed for the first time that in comparison with monolayer culture tumor cells, spheroids are much less susceptible to the deprivation of individual amino acids (i.e., arginine, leucine, lysine or methionine). Accordingly, even after prolonged (up to 10 days) starvation, spheroid cells could readily resume proliferation when appropriate amino acid was resupplemented. In the case of arginine deprivation, similar apoptosis induction was detected both in 2-D and 3-D culture, suggesting that this process does not determine the level of tumor cell sensitivity to this kind of treatment. It was also observed that spheroids much better mimic the in vivo ability of tumor cells to utilize citrulline as arginine precursor for growth in amino acid deficient environment. We conclude that 3-D spheroid culture better reflects in vivo tumor cell response to single amino acid starvation than 2-D monolayer culture and should be used as an integral model in the studies of this type of antitumor metabolic targeting.


Subject(s)
Amino Acids/metabolism , Cell Culture Techniques/methods , Neoplasms/metabolism , Apoptosis , Cell Line, Tumor , Humans , Neoplasms/physiopathology
5.
Anticancer Drugs ; 22(2): 148-57, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20717004

ABSTRACT

Arginine deprivation achieved by means of recombinant arginine-degrading enzymes is currently being developed as a novel anticancer enzymotherapy. In this study, we showed that arginine deprivation in vitro profoundly and selectively sensitized human cancer cells of different organ origin to low doses of canavanine, an arginine analogue of plant origin. In sensitive cancer cells arginine starvation led to the activation of caspase-9, caspase-3 and caspase-7, cleavage of reparation enzyme, polyADP ribosyl polymerase, and DNA fragmentation, which are the typical hallmarks of intrinsic apoptosis realized by the mitochondrial pathway. Co-administration of canavanine significantly accelerated and enhanced apoptotic manifestations induced by arginine deprivation. The augmentation of canavanine toxicity for cancer cells was observed when either a formulated arginine-free medium or complete medium supplemented with bovine arginase preparation was used. Cycloheximide efficiently rescued malignant cells from canavanine-induced cytotoxicity under arginine deprivation, suggesting that it results mainly from canavanine incorporation into newly synthesized proteins. Cancer cells sensitive or resistant to arginine deprivation alone were not capable of restoring their proliferation after 24 h of combined treatment, whereas pseudonormal cells retained such ability. Our data suggest that the incorporation of canavanine into anticancer treatment schemes based on artificially created arginine starvation could be a novel strategy in tumor enzymochemotherapy.


Subject(s)
Apoptosis/drug effects , Arginine/deficiency , Canavanine/pharmacology , Neoplasms/therapy , Arginine/analogs & derivatives , Arginine/metabolism , Canavanine/pharmacokinetics , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Drug Therapy, Combination , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding
6.
Cell Biol Int ; 34(11): 1085-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20653567

ABSTRACT

Single amino acid Arg (arginine) deprivation is currently considered as a therapeutic approach to treat certain types of tumours; the molecular mechanisms that underlie tumour cell sensitivity or resistance to Arg restriction are still little understood. Here, we address the question of whether endogenous levels of key Arg metabolic enzymes [catabolic: arginases, ARG1 (arginase type 1) and ARG2 (arginase type 2), and anabolic: OTC (ornithine transcarbamylase) and ASS (argininosuccinate synthetase)] affect cellular responses to arginine deprivation in vitro. Human epithelial cancer cells of different organs of origin exhibiting variable sensitivity to Arg deprivation provided the experimental models. Neither the basal expression status of the analysed enzymes, nor their changes upon arginine withdrawal correlated with cancer cell sensitivity to arginine deprivation. However, the ability to utilize exogenous Arg precursors (ornithine and citrulline) for growth in Arg-deficient medium strongly correlated with expression of the corresponding enzymes, OTC and ASS. We also observed that OTC expression was below the level of detection in all the types of tumour cells analysed, suggesting that in vitro, at least for them, Arg is an essential amino acid.


Subject(s)
Arginase/genetics , Arginine/deficiency , Neoplasms/enzymology , Arginase/metabolism , Arginine/metabolism , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Citrulline/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Neoplasms/metabolism , Ornithine/metabolism , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...