Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Biomolecules ; 12(8)2022 08 02.
Article in English | MEDLINE | ID: mdl-36008963

ABSTRACT

Alternative splicing allows the synthesis of different protein variants starting from a single gene. Human Beclin 1 (BECN1) is a key autophagy regulator that acts as haploinsufficient tumor suppressor since its decreased expression correlates with tumorigenesis and poor prognosis in cancer patients. Recent studies show that BECN1 mRNA undergoes alternative splicing. Here, we report on the isolation and molecular and functional characterization of three BECN1 transcript variants (named BECN1-α, -ß and -γ) in human cancer cells. In ovarian cancer NIHOVCAR3, these splicing variants were found along with the canonical wild-type. BECN1-α lacks 143 nucleotides at its C-terminus and corresponds to a variant previously described. BECN1-ß and -γ lack the BCL2 homology 3 domain and other regions at their C-termini. Following overexpression in breast cancer cells MDA-MB231, we found that BECN1-α stimulates autophagy. Specifically, BECN1-α binds to Parkin and stimulates mitophagy. On the contrary, BECN1-ß reduces autophagy with a dominant negative effect over the endogenous wild-type isoform. BECN1-γ maintains its ability to interact with the vacuolar protein sorting 34 and only has a slight effect on autophagy. It is possible that cancer cells utilize the alternative splicing of BECN1 for modulating autophagy and mitophagy in response to environmental stresses.


Subject(s)
Apoptosis Regulatory Proteins , Beclin-1/metabolism , Neoplasms , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy , Beclin-1/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Isoforms/genetics
2.
Int J Mol Sci ; 23(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35563171

ABSTRACT

Neuroblastoma is a malignant extracranial solid tumor arising from the sympathoadrenal lineage of the neural crest and is often associated with N-MYC amplification. Cathepsin D has been associated with chemoresistance in N-MYC-overexpressing neuroblastomas. Increased EGFR expression also has been associated with the aggressive behavior of neuroblastomas. This work aimed to understand the mechanisms linking EGFR stimulation and cathepsin D expression with neuroblastoma progression and prognosis. Gene correlation analysis in pediatric neuroblastoma patients revealed that individuals bearing a high EGFR transcript level have a good prognosis only when CTSD (the gene coding for the lysosomal protease Cathepsin D, CD) is highly expressed. Low CTSD expression was associated with poor clinical outcome. CTSD expression was negatively correlated with CCNB2, CCNA2, CDK1 and CDK6 genes involved in cell cycle division. We investigated the biochemical pathways downstream to EGFR stimulation in human SH-SY5Y neuroblastoma cells engineered for overexpressing or silencing of CD expression. Cathepsin D overexpression decreased the proliferative potential of neuroblastoma cells through downregulation of the pro-oncogenic MAPK signaling pathway. EGFR stimulation downregulated cathepsin D expression, thus favoring cell cycle division. Our data suggest that chemotherapeutics that inhibit the EGFR pathway, along with stimulators of cathepsin D synthesis and activity, could benefit neuroblastoma prognosis.


Subject(s)
Cathepsin D , Neuroblastoma , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Cycle/genetics , Child , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lysosomes/metabolism , Neuroblastoma/metabolism , Peptide Hydrolases/metabolism
3.
Mol Carcinog ; 58(10): 1754-1769, 2019 10.
Article in English | MEDLINE | ID: mdl-31215708

ABSTRACT

We have previously shown that nearly half of mesothelioma patients have tumors with low autophagy and that these patients have a significantly worse outcome than those with high autophagy. We hypothesized that autophagy may be beneficial by facilitating immunogenic cell death (ICD) of tumor cells following chemotherapy. An important hallmark of ICD is that death of tumor cells is preceded or accompanied by the release of damage-associated molecular pattern molecules (DAMPs), which then can stimulate an antitumor immune response. Therefore, we measured how autophagy affected the release of three major DAMPs: high mobility group box 1 (HMGB1), ATP, and calreticulin following chemotherapy. We found that autophagy in three-dimensional (3D) models with low autophagy at baseline could be upregulated with the cell-permeant Tat-BECN1 peptide and confirmed that autophagy in 3D models with high autophagy at baseline could be inhibited with MRT 68921 or ATG7 RNAi, as we have previously shown. In in vitro 3D spheroids, we found that, when autophagy was high or upregulated, DAMPs were released following chemotherapy; however, when autophagy was low or inhibited, DAMPs release was significantly impaired. Similarly, in ex vivo tumors, when autophagy was high or upregulated, HMGB1 was released following chemotherapy but, when autophagy was low, HMGB1 release was not seen. We conclude that autophagy can be upregulated in at least some tumors with low autophagy and that upregulation of autophagy can restore the release of DAMPs following chemotherapy. Autophagy may be necessary for ICD in this tumor.


Subject(s)
Autophagy/genetics , HMGB1 Protein/genetics , Immunogenic Cell Death/genetics , Mesothelioma/drug therapy , Adenosine Triphosphate/genetics , Alarmins/genetics , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy-Related Protein 7/antagonists & inhibitors , Autophagy-Related Proteins/genetics , Beclin-1/genetics , Calreticulin/genetics , Cell Culture Techniques , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity, Cellular/genetics , Mesothelioma/genetics , Mesothelioma/pathology , RNA Interference , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology
4.
Cell Commun Signal ; 17(1): 39, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31046771

ABSTRACT

BACKGROUND: In the event of amino acid starvation, the cell activates two main protective pathways: Amino Acid starvation Response (AAR), to inhibit global translation, and autophagy, to recover the essential substrates from degradation of redundant self-components. Whether and how AAR and autophagy (ATG) are cross-regulated and at which point the two regulatory pathways intersect remain unknown. Here, we provide experimental evidence that the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) specifically located at the lysosome level links the AAR with the autophagy pathway. METHODS: As an inducer of the AAR, we used halofuginone (HF), an alkaloid that binds to the prolyl-tRNA synthetase thus mimicking the unavailability of proline (PRO). Induction of AAR was determined assessing the phosphorylation of the eukaryotic translation initiation factor (eIF) 2α. Autophagy was monitored by assessing the processing and accumulation of microtubule-associated protein 1 light chain 3 isoform B (LC3B) and sequestosome-1 (p62/SQSTM1) levels. The activity of mTORC1 was monitored through assessment of the phosphorylation of mTOR, (rp)S6 and 4E-BP1. Global protein synthesis was determined by puromycin incorporation assay. mTORC1 presence on the membrane of the lysosomes was monitored by cell fractionation and mTOR expression was determined by immunoblotting. RESULTS: In three different types of human cancer cells (thyroid cancer WRO cells, ovarian cancer OAW-42 cells, and breast cancer MCF-7 cells), HF induced both the AAR and the autophagy pathways time-dependently. In WRO cells, which showed the strongest induction of autophagy and of AAR, global protein synthesis was little if any affected. Consistently, 4E-BP1 and (rp)S6 were phosphorylated. Concomitantly, mTOR expression and activation declined along with its detachment from the lysosomes and its degradation by the proteasome, and with the nuclear translocation of transcription factor EB (TFEB), a transcription factor of many ATG genes. The extra supplementation of proline rescued all these effects. CONCLUSIONS: We demonstrate that the AAR and autophagy are mechanistically linked at the level of mTORC1, and that the lysosome is the central hub of the cross-talk between these two metabolic stress responses.


Subject(s)
Autophagy/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Piperidines/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Synthesis Inhibitors/pharmacology , Quinazolinones/pharmacology , Amino Acids/deficiency , Amino Acids/metabolism , Eukaryotic Initiation Factor-2/metabolism , Humans , MCF-7 Cells , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism
5.
Methods Mol Biol ; 1880: 491-510, 2019.
Article in English | MEDLINE | ID: mdl-30610717

ABSTRACT

Three-dimensional (3D) models are acquiring importance in cancer research due to their ability to mimic multiple features of the tumor microenvironment more accurately than standard monolayer two-dimensional (2D) cultures. Several groups, including our laboratory, are now accumulating evidence that autophagy in solid tumors is also better represented in 3D than in 2D. Here we detail how we generate 3D models, both in vitro multicellular spheroids generated from cell lines and ex vivo tumor fragment spheroids generated from tumor samples, and how autophagy can be measured in 3D cultures.


Subject(s)
Autophagy/physiology , Cell Culture Techniques/methods , Mesothelioma/pathology , Spheroids, Cellular/pathology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Proteins/analysis , Autophagy-Related Proteins/metabolism , Cell Culture Techniques/instrumentation , Cell Line, Tumor , Humans , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/metabolism , Tubulin/analysis , Tubulin/metabolism
6.
Mol Carcinog ; 57(3): 319-332, 2018 03.
Article in English | MEDLINE | ID: mdl-29073722

ABSTRACT

The benefits of inhibiting autophagy in cancer are still controversial, with differences in outcome based on the type of tumor, the context and the particular stage of inhibition. Here, we investigated the impact of inhibiting autophagy at different stages on chemosensitivity using 3-dimensional (3D) models of mesothelioma, including ex vivo human tumor fragment spheroids. As shown by LC3B accumulation, we successfully inhibited autophagy using either an early stage ULK1/2 inhibitor (MRT 68921) or a late stage inhibitor (hydroxychloroquine). We found that inhibition of autophagy at the early stage, but not at late stage, potentiated chemosensitivity. This effect was seen only in those spheroids with high autophagy and active initiation at steady state. Inhibition of autophagy alone, at either early or late stage, did not cause cell death, showing that the inhibitors were non-toxic and that mesothelioma did not depend on autophagy at baseline, at least over 24 h. Using ATG13 puncta analysis, we found that autophagy initiation identified tumors that are more chemosensitive at baseline and after autophagy inhibition. Our results highlight a potential role of autophagy initiation in supporting mesothelioma cells during chemotherapy. Our work also highlights the importance of testing the inhibition of different stages in order to uncover the role of autophagy and the potential of its modulation in the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Autophagy/drug effects , Hydroxychloroquine/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mesothelioma/drug therapy , Protein Kinase Inhibitors/pharmacology , Autophagy-Related Protein-1 Homolog/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mesothelioma/metabolism , Mesothelioma/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Spheroids, Cellular , Tumor Cells, Cultured
7.
Mol Carcinog ; 56(12): 2681-2691, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28856729

ABSTRACT

The potential benefit of nutrient starvation in the prevention and treatment of cancer is presently under consideration. Resveratrol (RV), a dietary polyphenol acting as a protein (caloric) restriction mimetic, could substitute for amino acid starvation. The effects of starvation and of caloric restriction are mediated, among others, by autophagy, a process that contributes to cell homeostasis by promoting the lysosomal degradation of damaged and redundant self-constituents. Up-regulation of autophagy favors cell survival under nutrient shortage situation, and may drive cancer cells into a non-replicative, dormant state. Both RV and amino acid starvation effectively induced the aminoacid response and autophagy. These processes were associated with inhibition of the mTOR pathway and disruption of the BECLIN1-BCL-2 complex. The number of transcripts positively impinging on the autophagy pathway was higher in RV-treated than in starved cancer cells. Consistent with our data, it appears that RV treatment is more effective than and can substitute for starvation for inducing autophagy in cancer cells. The present findings are clinically relevant because of the potential therapeutic implications.


Subject(s)
Amino Acids/metabolism , Autophagy/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/genetics , Blotting, Western , Cell Line, Tumor , Cluster Analysis , Female , Humans , Microscopy, Fluorescence , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Resveratrol , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Oncotarget ; 7(51): 84999-85020, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27829222

ABSTRACT

GLUT1 is the facilitative transporter playing the major role in the internalization of glucose. Basally, GLUT1 resides on vesicles located in a para-golgian area, and is translocated onto the plasmamembrane upon activation of the PI3KC1-AKT pathway. In proliferating cancer cells, which demand a high quantity of glucose for their metabolism, GLUT1 is permanently expressed on the plasmamembrane. This is associated with the abnormal activation of the PI3KC1-AKT pathway, consequent to the mutational activation of PI3KC1 and/or the loss of PTEN. The latter, in fact, could antagonize the phosphorylation of AKT by limiting the availability of Phosphatidylinositol (3,4,5)-trisphosphate. Here, we asked whether PTEN could control the plasmamembrane expression of GLUT1 also through its protein-phosphatase activity on AKT. Experiments of co-immunoprecipitation and in vitro de-phosphorylation assay with homogenates of cells transgenically expressing the wild type or knocked-down mutants (lipid-phosphatase, protein-phosphatase, or both) isoforms demonstrated that indeed PTEN physically interacts with AKT and drives its dephosphorylation, and so limiting the expression of GLUT1 at the plasmamembrane. We also show that growth factors limit the ability of PTEN to dephosphorylate AKT. Our data emphasize the fact that PTEN acts in two distinct steps of the PI3k/AKT pathway to control the expression of GLUT1 at the plasmamembrane and, further, add AKT to the list of the protein substrates of PTEN.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Oncogene Protein v-akt/metabolism , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Cell Extracts , Cell Line, Tumor , Female , Humans , PTEN Phosphohydrolase/genetics , Phosphorylation , Protein Binding , RNA, Small Interfering/genetics , Signal Transduction
9.
Autophagy ; 12(7): 1180-94, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27097020

ABSTRACT

Understanding the role of autophagy in cancer has been limited by the inability to measure this dynamic process in formalin-fixed tissue. We considered that 3-dimensional models including ex vivo tumor, such as we have developed for studying mesothelioma, would provide valuable insights. Using these models, in which we could use lysosomal inhibitors to measure the autophagic flux, we sought a marker of autophagy that would be valid in formalin-fixed tumor and be used to assess the role of autophagy in patient outcome. Autophagy was studied in mesothelioma cell lines, as 2-dimensional (2D) monolayers and 3-dimensional (3D) multicellular spheroids (MCS), and in tumor from 25 chemonaive patients, both as ex vivo 3D tumor fragment spheroids (TFS) and as formalin-fixed tissue. Autophagy was evaluated as autophagic flux by detection of the accumulation of LC3 after lysosomal inhibition and as autophagy initiation by detection of ATG13 puncta. We found that autophagic flux in 3D, but not in 2D, correlated with ATG13 positivity. In each TFS, ATG13 positivity was similar to that of the original tumor. When tested in tissue microarrays of 109 chemonaive patients, higher ATG13 positivity correlated with better prognosis and provided information independent of known prognostic factors. Our results show that ATG13 is a static marker of the autophagic flux in 3D models of mesothelioma and may also reflect autophagy levels in formalin-fixed tumor. If confirmed, this marker would represent a novel prognostic factor for mesothelioma, supporting the notion that autophagy plays an important role in this cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Proteins/metabolism , Autophagy/physiology , Mesothelioma/metabolism , Mesothelioma/therapy , Animals , Cell Culture Techniques , Cell Line , Formaldehyde , Humans , Treatment Outcome
10.
Med Res Rev ; 36(5): 845-70, 2016 09.
Article in English | MEDLINE | ID: mdl-27114232

ABSTRACT

In familial neurodegenerative disorders, protein aggregates form continuously because of genetic mutations that drive the synthesis of truncated or unfolded proteins. The oxidative stress imposed by neurotransmitters and environmental neurotoxins constitutes an additional threat to the folding of the proteins and the integrity of organelle membranes in neurons. Failure in degrading such altered materials compromises the function of neurons and eventually leads to neurodegeneration. The lysosomal proteolytic enzyme Cathepsin D is the only aspartic-type protease ubiquitously expressed in all the cells of the human body, and it is expressed at high level in the brain. In general, cathepsin D mediated proteolysis is essential to neuronal cell homeostasis through the degradation of unfolded or oxidized protein aggregates delivered to lysosomes via autophagy or endocytosis. More specifically, many altered neuronal proteins that hallmark neurodegenerative diseases (e.g., the amyloid precursor, α-synuclein, and huntingtin) are physiologic substrates of cathepsin D and would abnormally accumulate if not efficiently degraded by this enzyme. Furthermore, experimental evidence indicates that cathepsin D activity is linked to the metabolism of cholesterol and of glycosaminoglycans, which accounts for its involvement in neuronal plasticity. This review focuses on the unique role of cathepsin D mediated proteolysis in the pathogenesis of human neurodegenerative diseases.


Subject(s)
Cathepsin D/metabolism , Neurodegenerative Diseases/enzymology , Animals , Humans , Neurodegenerative Diseases/etiology
11.
PLoS One ; 11(3): e0150044, 2016.
Article in English | MEDLINE | ID: mdl-26982031

ABSTRACT

To investigate the underlying causes of chemoresistance in malignant pleural mesothelioma, we have studied mesothelioma cell lines as 3D spheroids, which acquire increased chemoresistance compared to 2D monolayers. We asked whether the gene expression of 3D spheroids would reveal mechanisms of resistance. To address this, we measured gene expression of three mesothelioma cell lines, M28, REN and VAMT, grown as 2D monolayers and 3D spheroids. A total of 209 genes were differentially expressed in common by the three cell lines in 3D (138 upregulated and 71 downregulated), although a clear resistance pathway was not apparent. We then compared the list of 3D genes with two publicly available datasets of gene expression of 56 pleural mesotheliomas compared to normal tissues. Interestingly, only three genes were increased in both 3D spheroids and human tumors: argininosuccinate synthase 1 (ASS1), annexin A4 (ANXA4) and major vault protein (MVP); of these, ASS1 was the only consistently upregulated of the three genes by qRT-PCR. To measure ASS1 protein expression, we stained 2 sets of tissue microarrays (TMA): one with 88 pleural mesothelioma samples and the other with additional 88 pleural mesotheliomas paired with matched normal tissues. Of the 176 tumors represented on the two TMAs, ASS1 was expressed in 87 (50%; staining greater than 1 up to 3+). For the paired samples, ASS1 expression in mesothelioma was significantly greater than in the normal tissues. Reduction of ASS1 expression by siRNA significantly sensitized mesothelioma spheroids to the pro-apoptotic effects of bortezomib and of cisplatin plus pemetrexed. Although mesothelioma is considered by many to be an ASS1-deficient tumor, our results show that ASS1 is elevated at the mRNA and protein levels in mesothelioma 3D spheroids and in human pleural mesotheliomas. We also have uncovered a survival role for ASS1, which may be amenable to targeting to undermine mesothelioma multicellular resistance.


Subject(s)
Argininosuccinate Synthase/metabolism , Cell Survival , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Mesothelioma/genetics , Spheroids, Cellular , Annexin A4/metabolism , Cell Line, Tumor , Humans , Mesothelioma/pathology , Vault Ribonucleoprotein Particles/metabolism
12.
Sci Rep ; 6: 19311, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26776680

ABSTRACT

Protein aggregation is a common biological phenomenon, observed in different physiological and pathological conditions. Decreased protein solubility and a tendency to aggregate is also observed during physiological aging but the causes are currently unknown. Herein we performed a biophysical separation of aging-related high molecular weight aggregates, isolated from the bone marrow and splenic cells of aging mice and followed by biochemical and mass spectrometric analysis. The analysis indicated that compared to younger mice an increase in protein post-translational carbonylation was observed. The causative role of these modifications in inducing protein misfolding and aggregation was determined by inducing carbonyl stress in young mice, which recapitulated the increased protein aggregation observed in old mice. Altogether our analysis indicates that oxidative stress-related post-translational modifications accumulate in the aging proteome and are responsible for increased protein aggregation and altered cell proteostasis.


Subject(s)
Aging/metabolism , Protein Aggregates , Protein Carbonylation , Proteins/metabolism , Aging/pathology , Animals , Bone Marrow Cells/metabolism , Female , Mice , Oxidative Stress , Protein Aggregation, Pathological , Reactive Oxygen Species/metabolism , Spleen/metabolism
13.
PLoS One ; 10(8): e0134825, 2015.
Article in English | MEDLINE | ID: mdl-26284517

ABSTRACT

Malignant pleural mesothelioma is a highly chemoresistant solid tumor. We have studied this apoptotic resistance using in vitro and ex vivo three-dimensional models, which acquire a high level of chemoresistance that can be reduced by PI3K/mTOR inhibitors. Here, we investigate the activity of GDC-0980, a novel dual PI3K/mTOR inhibitor, which has been proposed to be effective in mesothelioma. In this work, we aimed to identify mechanisms and markers of efficacy for GDC-0980 by utilizing 3D models of mesothelioma, both in vitro multicellular spheroids and ex vivo tumor fragment spheroids grown from patient tumor samples. We found that a subset of mesothelioma spheroids is sensitive to GDC-0980 alone and to its combination with chemotherapy. Unexpectedly, this sensitivity did not correlate with the activation of the Akt/mTOR pathway. Instead, sensitivity to GDC-0980 correlated with the presence of constitutive ATG13 puncta, a feature of autophagy, a cellular program that supports cells under stress. In tumor fragment spheroids grown from 21 tumors, we also found a subset (n = 11) that was sensitive to GDC-0980, a sensitivity that also correlated with the presence of ATG13 puncta. Interference with autophagy by siRNA of ATG7, an essential autophagic protein, increased the response to chemotherapy, but only in the sensitive multicellular spheroids. In the spheroids resistant to GDC-0980, autophagy appeared to play no role. In summary, we show that GDC-0980 is effective in mesothelioma 3D models that display ATG13 puncta, and that blockade of autophagy increases their response to chemotherapy. For the first time, we show a role for autophagy in the response to chemotherapy of 3D models of mesothelioma and propose ATG13 as a potential biomarker of the therapeutic responsiveness of mesothelioma.


Subject(s)
Autophagy/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Pyrimidines/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Biomarkers/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Activating Enzymes/metabolism
14.
Genes Cancer ; 5(7-8): 226-39, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25221641

ABSTRACT

Proliferating cancer cells oxidize glucose through the glycolytic pathway. Since this metabolism is less profitable in terms of ATP production, cancer cells consume large quantity of glucose, and those that experience insufficient blood supply become glucose-addicted. We have analyzed the response to glucose depletion in WRO and FTC133 follicular thyroid cancer cells, which differ in the expression of two key regulators of the glucose metabolism. WRO cells, which express wild type p53 and PTEN, showed a higher rate of cell proliferation and were much less sensitive to glucose-depletion than FTC133 cells, which are PTEN null and express mutant p53. Glucose depletion slowed-down the autophagy flux in FTC133 cells, not in WRO cells. In a wound-healing assay, WRO cells were shown to migrate faster than FTC133 cells. Glucose depletion slowed down the cell migration rate, and these effects were more evident in FTC133 cells. Genetic silencing of either wild-type PTEN or p53 in WRO cells resulted in increased uptake of glucose, whereas the ectopic expression of PTEN in FTC133 cells resulted in diminished glucose uptake. In conclusion, compared to WRO, FTC133 cells were higher glucose up-taker and consumer. These data do not support the general contention that cancer cells lacking PTEN or expressing the mutant p53R273H are more aggressive and prone to better face glucose depletion. We propose that concurrent PTEN deficiency and mutant p53 leads to a glucose-addiction state that renders the cancer cell more sensitive to glucose restriction. The present observation substantiates the view that glucose-restriction may be an adjuvant strategy to combat these tumours.

15.
J Mol Endocrinol ; 53(2): 247-58, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25125078

ABSTRACT

Glucose represents an important source of energy for the cells. Proliferating cancer cells consume elevated quantity of glucose, which is converted into lactate regardless of the presence of oxygen. This phenomenon, known as the Warburg effect, has been proven to be useful for imaging metabolically active tumours in cancer patients by (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET). Glucose is internalised in the cells by glucose transporters (GLUTs) belonging to the GLUT family. GLUT1 (SLC2A1) is the most prevalent isoform in more aggressive and less differentiated thyroid cancer histotypes. In a previous work, we found that loss of expression of PTEN was associated with increased expression of GLUT1 on the plasma membrane (PM) and probability of detecting thyroid incidentalomas by FDG-PET. Herein, we investigated the molecular pathways that govern the expression of GLUT1 on the PM and the glucose uptake in WRO (expressing WT PTEN) and FTC133 (PTEN null) follicular thyroid cancer cells cultured under glucose-depleted conditions. The membrane expression of GLUT1 was enhanced in glucose-deprived cells. Through genetic manipulations of PTEN expression, we could demonstrate that the lack of this oncosuppressor has a dominant effect on the membrane expression of GLUT1 and glucose uptake. We conclude that loss of function of PTEN increases the probability of cancer detection by FDG-PET or other glucose-based imaging diagnosis.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 1/metabolism , Glucose/metabolism , PTEN Phosphohydrolase/metabolism , Thyroid Neoplasms/metabolism , Cell Line, Tumor , Gene Expression , Humans , Intracellular Space/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Thyroid Neoplasms/genetics
16.
Phytother Res ; 28(12): 1761-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25044209

ABSTRACT

The keratinocyte-derived A431 Squamous Cell Carcinoma cells express the p53R273H mutant, which has been reported to inhibit apoptosis and autophagy. Here, we show that the crude extract of turmeric (Curcuma longa), similarly to its bioactive component Curcumin, could induce both apoptosis and autophagy in A431 cells, and these effects were concomitant with degradation of p53. Turmeric and curcumin also stimulated the activity of mTOR, which notoriously promotes cell growth and acts negatively on basal autophagy. Rapamycin-mediated inhibition of mTOR synergized with turmeric and curcumin in causing p53 degradation, increased the production of autophagosomes and exacerbated cell toxicity leading to cell necrosis. Small-interference mediated silencing of the autophagy proteins BECLIN 1 or ATG7 abrogated the induction of autophagy and largely rescued p53 stability in Turmeric-treated or Curcumin-treated cells, indicating that macroautophagy was mainly responsible for mutant p53 degradation. These data uncover a novel mechanism of turmeric and curcumin toxicity in chemoresistant cancer cells bearing mutant p53.


Subject(s)
Autophagy/drug effects , Carcinoma, Squamous Cell/pathology , Curcuma/chemistry , Plant Extracts/pharmacology , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Humans , Membrane Proteins/metabolism , Mutant Proteins/metabolism , RNA Interference , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Activating Enzymes/metabolism
17.
Biomed Res Int ; 2014: 343542, 2014.
Article in English | MEDLINE | ID: mdl-24877083

ABSTRACT

Autophagy is a lysosomal-driven catabolic process that contributes to the preservation of cell homeostasis through the regular elimination of cellular damaged, aged, and redundant molecules and organelles. Autophagy plays dual opposite roles in cancer: on one hand it prevents carcinogenesis; on the other hand it confers an advantage to cancer cells to survive under prohibitive conditions. Autophagy has been implicated in ovarian cancer aggressiveness and in ovarian cancer cell chemoresistance and dormancy. Small noncoding microRNAs (miRNAs) regulate gene expression at posttranscriptional level, thus playing an important role in many aspects of cell pathophysiology, including cancerogenesis and cancer progression. Certain miRNAs have recently emerged as important epigenetic modulators of autophagy in cancer cells. The mRNA of several autophagy-related genes contains, in fact, the target sequence for miRNAs belonging to different families, with either oncosuppressive or oncogenic activities. MiRNA profiling studies have identified some miRNAs aberrantly expressed in ovarian cancer tissues that can impact autophagy. In addition, plasma and stroma cell-derived miRNAs in tumour-bearing patients can regulate the expression of relevant autophagy genes in cancer cells. The present review focuses on the potential implications of miRNAs regulating autophagy in ovarian cancer pathogenesis and progression.


Subject(s)
Autophagy , Epigenesis, Genetic , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , RNA, Neoplasm/metabolism , Animals , Female , Gene Expression Regulation, Neoplastic , Humans
18.
Biosci Rep ; 33(2): e00034, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23464837

ABSTRACT

CD (cathepsin D) is a ubiquitous lysosomal hydrolase involved in a variety of pathophysiological functions, including protein turnover, activation of pro-hormones, cell death and embryo development. CD-mediated proteolysis plays a pivotal role in tissue and organ homoeostasis. Altered expression and compartmentalization of CD have been observed in diseased muscle fibres. Whether CD is actively involved in muscle development, homoeostasis and dystrophy remains to be demonstrated. Zebrafish (Danio rerio) is emerging as a valuable 'in vivo' vertebrate model for muscular degeneration and congenital myopathies. In this work, we report on the perturbance of the somitic musculature development in zebrafish larvae caused by MPO (morpholino)-mediated silencing of CD in oocytes at the time of fertilization. Restoring CD expression, using an MPO-non-matching mutated mRNA, partially rescued the normal phenotype, confirming the indispensable role of CD in the correct development and integrity of the somitic musculature. This is the first report showing a congenital myopathy caused by CD deficiency in a vertebrate experimental animal model.


Subject(s)
Cathepsin D/genetics , Myopathies, Structural, Congenital/genetics , Zebrafish/genetics , Zygote/metabolism , Animals , Cathepsin D/biosynthesis , Disease Models, Animal , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Gene Silencing , Humans , Myopathies, Structural, Congenital/etiology , Myopathies, Structural, Congenital/pathology , RNA, Messenger/genetics , Zebrafish/growth & development , Zygote/pathology
19.
Int J Biochem Cell Biol ; 45(2): 273-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23107604

ABSTRACT

The lysosomal protease Cathepsin D (CD) plays a role in neurodegenerative diseases, cancer, and embryo-fetus abnormalities. It is therefore of interest to know how this protein is synthesized in animal species used for modeling human diseases. Zebrafish (Danio rerio) is emerging as a valuable 'in vivo' vertebrate model for several human diseases. We have characterized the biogenetic pathways of zebrafish and human CD transgenically expressed in both human SH-SY5Y cells and zebrafish PAC2 cells. Differently from human CD, zebrafish CD was synthesized as a mono-glycosylated precursor (pro-CD) that was eventually processed into a single-chain mature polypeptide. In PAC2 cells, ammonium chloride and chloroquine impaired the N-glycosylation, and greatly stimulated the secretion, of pro-CD; still, a portion of un-glycosylated pro-CD reached the lysosomes and was processed to mature CD. The treatment with tunicamycin, which abrogates N-glycosylation, resulted in a similar effect. Zebrafish pro-CD was correctly processed when expressed in human cells, and its glycosylation, transport and maturation were not impaired by ammonium chloride. On the contrary, the transport and processing of human pro-CD expressed in zebrafish cells were profoundly altered: while the intermediate single-chain was not detectable, a small amount of double-chain mature CD still formed. This fact indicates that the enzyme machinery for single- to double-chain processing of mammal CD is present in zebrafish. Our data highlight the respective impact of the information imparted by the primary sequence and of the cellular transport and processing machineries in the biogenesis of lysosomal CD.


Subject(s)
Cathepsin D/metabolism , Enzyme Precursors/metabolism , Lysosomes/metabolism , Protein Processing, Post-Translational , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Cathepsin D/chemistry , Cell Line , Enzyme Precursors/chemistry , Glycosylation , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Sequence Homology, Amino Acid , Zebrafish , Zebrafish Proteins/chemistry
20.
Int J Nanomedicine ; 7: 4147-58, 2012.
Article in English | MEDLINE | ID: mdl-22904626

ABSTRACT

BACKGROUND AND METHODS: Nanoparticles engineered to carry both a chemotherapeutic drug and a sensitive imaging probe are valid tools for early detection of cancer cells and to monitor the cytotoxic effects of anticancer treatment simultaneously. Here we report on the effect of size (10-30 nm versus 50 nm), type of material (mesoporous silica versus polystyrene), and surface charge functionalization (none, amine groups, or carboxyl groups) on biocompatibility, uptake, compartmentalization, and intracellular retention of fluorescently labeled nanoparticles in cultured human ovarian cancer cells. We also investigated the involvement of caveolae in the mechanism of uptake of nanoparticles. RESULTS: We found that mesoporous silica nanoparticles entered via caveolae-mediated endocytosis and reached the lysosomes; however, while the 50 nm nanoparticles permanently resided within these organelles, the 10 nm nanoparticles soon relocated in the cytoplasm. Naked 10 nm mesoporous silica nanoparticles showed the highest and 50 nm carboxyl-modified mesoporous silica nanoparticles the lowest uptake rates, respectively. Polystyrene nanoparticle uptake also occurred via a caveolae-independent pathway, and was negatively affected by serum. The 30 nm carboxyl-modified polystyrene nanoparticles did not localize in lysosomes and were not toxic, while the 50 nm amine-modified polystyrene nanoparticles accumulated within lysosomes and eventually caused cell death. Ovarian cancer cells expressing caveolin-1 were more likely to endocytose these nanoparticles. CONCLUSION: These data highlight the importance of considering both the physicochemical characteristics (ie, material, size and surface charge on chemical groups) of nanoparticles and the biochemical composition of the cell membrane when choosing the most suitable nanotheranostics for targeting cancer cells.


Subject(s)
Biocompatible Materials/pharmacokinetics , Nanoparticles/chemistry , Ovarian Neoplasms/metabolism , Polystyrenes/pharmacokinetics , Silicon Dioxide/pharmacokinetics , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Caveolin 1/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Endocytosis , Female , Humans , Lysosomes/metabolism , Ovarian Neoplasms/drug therapy , Particle Size , Polystyrenes/chemistry , Polystyrenes/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Structure-Activity Relationship , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...