Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Theranostics ; 10(11): 5120-5136, 2020.
Article in English | MEDLINE | ID: mdl-32308772

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive forms of cancer. It has been proposed that the presence within these tumors of a population of cells with stem-like features termed Glioma Initiating Cells (GICs) is responsible for the relapses that take place in the patients with this disease. Targeting this cell population is therefore an issue of great therapeutic interest in neuro-oncology. We had previously found that the neurotrophic factor MIDKINE (MDK) promotes resistance to glioma cell death. The main objective of this work is therefore investigating the role of MDK in the regulation of GICs. Methods: Assays of gene and protein expression, self-renewal capacity, autophagy and apoptosis in cultures of GICs derived from GBM samples subjected to different treatments. Analysis of the growth of GICs-derived xenografts generated in mice upon blockade of the MDK and its receptor the ALK receptor tyrosine kinase (ALK) upon exposure to different treatments. Results: Genetic or pharmacological inhibition of MDK or ALK decreases the self-renewal and tumorigenic capacity of GICs via the autophagic degradation of the transcription factor SOX9. Blockade of the MDK/ALK axis in combination with temozolomide depletes the population of GICs in vitro and has a potent anticancer activity in xenografts derived from GICs. Conclusions: The MDK/ALK axis regulates the self-renewal capacity of GICs by controlling the autophagic degradation of the transcription factor SOX9. Inhibition of the MDK/ALK axis may be a therapeutic strategy to target GICs in GBM patients.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Midkine/metabolism , Neoplastic Stem Cells/metabolism , Temozolomide/pharmacology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/pharmacology , Autophagy/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line , Female , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Mice, Nude , Midkine/antagonists & inhibitors , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Biochem Pharmacol ; 157: 266-274, 2018 11.
Article in English | MEDLINE | ID: mdl-30195736

ABSTRACT

Glioblastoma multiforme (GBM) is the most frequent and aggressive type of brain tumor due, at least in part, to its poor response to current anticancer treatments. These features could be explained, at least partially, by the presence within the tumor mass of a small population of cells termed Glioma Initiating Cells (GICs) that has been proposed to be responsible for the relapses occurring in this disease. Thus, the development of novel therapeutic approaches (and specifically those targeting the population of GICs) is urgently needed to improve the survival of the patients suffering this devastating disease. Previous observations by our group and others have shown that Δ9-Tetrahydrocannabinol (THC, the main active ingredient of marijuana) and other cannabinoids including cannabidiol (CBD) exert antitumoral actions in several animal models of cancer, including gliomas. We also found that the administration of THC (or of THC + CBD at a 1:1 ratio) in combination with temozolomide (TMZ), the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. In this work we investigated the effect of the combination of TMZ and THC:CBD mixtures containing different ratios of the two cannabinoids in preclinical glioma models, including those derived from GICs. Our findings show that TMZ + THC:CBD combinations containing a higher proportion of CDB (but not TMZ + CBD alone) produce a similar antitumoral effect as the administration of TMZ together with THC and CBD at a 1:1 ratio in xenografts generated with glioma cell lines. In addition, we also found that the administration of TMZ + THC:CBD at a 1:1 ratio reduced the growth of orthotopic xenografts generated with GICs derived from GBM patients and enhanced the survival of the animals bearing these intracranial xenografts. Remarkably, the antitumoral effect observed in GICs-derived xenografts was stronger when TMZ was administered together with cannabinoid combinations containing a higher proportion of CBD. These findings support the notion that the administration of TMZ together with THC:CBD combinations - and specifically those containing a higher proportion of CBD - may be therapeutically explored to target the population of GICs in GBM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Cannabidiol/therapeutic use , Dronabinol/therapeutic use , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Temozolomide/therapeutic use , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/pathology , Humans , Male , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Biochem Pharmacol ; 157: 275-284, 2018 11.
Article in English | MEDLINE | ID: mdl-30125556

ABSTRACT

Glioblastoma multiforme (GBM) is the most frequent and aggressive form of brain cancer. These features are explained at least in part by the high resistance exhibited by these tumors to current anticancer therapies. Thus, the development of novel therapeutic approaches is urgently needed to improve the survival of the patients suffering this devastating disease. Δ9-Tetrahydrocannabinol (THC, the major active ingredient of marijuana), and other cannabinoids have been shown to exert antitumoral actions in animal models of cancer, including glioma. The mechanism of these anticancer actions relies, at least in part, on the ability of these compounds to stimulate autophagy-mediated apoptosis in tumor cells. Previous observations from our group demonstrated that local administration of THC (or of THC + CBD at a 1:1 ratio, a mixture that resembles the composition of the cannabinoid-based medicine Sativex®) in combination with Temozolomide, the benchmark agent for the treatment of GBM, synergistically reduces the growth of glioma xenografts. With the aim of optimizing the possible clinical utilization of cannabinoids in anti-GBM therapies, in this work we explored the anticancer efficacy of the systemic administration of cannabinoids in combination with TMZ in preclinical models of glioma. Our results show that oral administration of Sativex-like extracts (containing THC and CBD at a 1:1 ratio) in combination with TMZ produces a strong antitumoral effect in both subcutaneous and intracranial glioma cell-derived tumor xenografts. In contrast, combined administration of Sativex-like and BCNU (another alkylating agent used for the treatment of GBM which share structural similarities with the TMZ) did not show a stronger effect than individual treatments. Altogether, our findings support the notion that the combined administration of TMZ and oral cannabinoids could be therapeutically exploited for the management of GBM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Cannabidiol/therapeutic use , Dronabinol/therapeutic use , Glioma/drug therapy , Temozolomide/therapeutic use , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/pathology , Cannabidiol/administration & dosage , Carmustine/therapeutic use , Cell Line, Tumor , Dronabinol/administration & dosage , Glioma/pathology , Heterografts , Humans , Male , Mice, Nude , Temozolomide/administration & dosage
5.
Autophagy ; 12(12): 2386-2403, 2016 12.
Article in English | MEDLINE | ID: mdl-27764541

ABSTRACT

The phospholipid cardiolipin (CL) has been proposed to play a role in selective mitochondrial autophagy, or mitophagy. CL externalization to the outer mitochondrial membrane would act as a signal for the human Atg8 ortholog subfamily, MAP1LC3 (LC3). The latter would mediate both mitochondrial recognition and autophagosome formation, ultimately leading to removal of damaged mitochondria. We have applied quantitative biophysical techniques to the study of CL interaction with various Atg8 human orthologs, namely LC3B, GABARAPL2 and GABARAP. We have found that LC3B interacts preferentially with CL over other di-anionic lipids, that CL-LC3B binding occurs with positive cooperativity, and that the CL-LC3B interaction relies only partially on electrostatic forces. CL-induced increased membrane fluidity appears also as an important factor helping LC3B to bind CL. The LC3B C terminus remains exposed to the hydrophilic environment after protein binding to CL-enriched membranes. In intact U87MG human glioblastoma cells rotenone-induced autophagy leads to LC3B translocation to mitochondria and subsequent delivery of mitochondria to lysosomes. We have also observed that GABARAP, but not GABARAPL2, interacts with CL in vitro. However neither GABARAP nor GABARAPL2 were translocated to mitochondria in rotenone-treated U87MG cells. Thus the various human Atg8 orthologs might play specific roles in different autophagic processes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy-Related Protein 8 Family/metabolism , Autophagy , Cardiolipins/metabolism , Microtubule-Associated Proteins/metabolism , Mitophagy , Amino Acid Sequence , Apoptosis Regulatory Proteins , Autophagy/drug effects , Autophagy-Related Protein 8 Family/chemistry , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Dronabinol/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Microtubule-Associated Proteins/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitophagy/drug effects , Pressure , Protein Binding/drug effects , Rotenone/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid
6.
Autophagy ; 12(11): 2213-2229, 2016 11.
Article in English | MEDLINE | ID: mdl-27635674

ABSTRACT

Autophagy is considered primarily a cell survival process, although it can also lead to cell death. However, the factors that dictate the shift between these 2 opposite outcomes remain largely unknown. In this work, we used Δ9-tetrahydrocannabinol (THC, the main active component of marijuana, a compound that triggers autophagy-mediated cancer cell death) and nutrient deprivation (an autophagic stimulus that triggers cytoprotective autophagy) to investigate the precise molecular mechanisms responsible for the activation of cytotoxic autophagy in cancer cells. By using a wide array of experimental approaches we show that THC (but not nutrient deprivation) increases the dihydroceramide:ceramide ratio in the endoplasmic reticulum of glioma cells, and this alteration is directed to autophagosomes and autolysosomes to promote lysosomal membrane permeabilization, cathepsin release and the subsequent activation of apoptotic cell death. These findings pave the way to clarify the regulatory mechanisms that determine the selective activation of autophagy-mediated cancer cell death.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Ceramides/pharmacology , Lysosomes/metabolism , Neoplasms/pathology , Biological Transport/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dronabinol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Lysosomes/drug effects , Lysosomes/ultrastructure , Models, Biological , Permeability , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Sphingolipids/biosynthesis
7.
Article in English | MEDLINE | ID: mdl-26071989

ABSTRACT

It is well-established that cannabinoids exert palliative effects on some cancer-associated symptoms. In addition evidences obtained during the last fifteen years support that these compounds can reduce tumor growth in animal models of cancer. Cannabinoids have been shown to activate an ER-stress related pathway that leads to the stimulation of autophagy-mediated cancer cell death. In addition, cannabinoids inhibit tumor angiogenesis and decrease cancer cell migration. The mechanisms of resistance to cannabinoid anticancer action as well as the possible strategies to develop cannabinoid-based combinational therapies to fight cancer have also started to be explored. In this review we will summarize these observations (that have already helped to set the bases for the development of the first clinical studies to investigate the potential clinical benefit of using cannabinoids in anticancer therapies) and will discuss the possible future avenues of research in this area.


Subject(s)
Antineoplastic Agents/therapeutic use , Cannabinoids/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cannabinoids/pharmacology , Clinical Trials as Topic , Endocannabinoids/metabolism , Humans , Neoplasms/metabolism
8.
J Invest Dermatol ; 135(6): 1629-1637, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25674907

ABSTRACT

Although the global incidence of cutaneous melanoma is increasing, survival rates for patients with metastatic disease remain <10%. Novel treatment strategies are therefore urgently required, particularly for patients bearing BRAF/NRAS wild-type tumors. Targeting autophagy is a means to promote cancer cell death in chemotherapy-resistant tumors, and the aim of this study was to test the hypothesis that cannabinoids promote autophagy-dependent apoptosis in melanoma. Treatment with Δ(9)-Tetrahydrocannabinol (THC) resulted in the activation of autophagy, loss of cell viability, and activation of apoptosis, whereas cotreatment with chloroquine or knockdown of Atg7, but not Beclin-1 or Ambra1, prevented THC-induced autophagy and cell death in vitro. Administration of Sativex-like (a laboratory preparation comprising equal amounts of THC and cannabidiol (CBD)) to mice bearing BRAF wild-type melanoma xenografts substantially inhibited melanoma viability, proliferation, and tumor growth paralleled by an increase in autophagy and apoptosis compared with standard single-agent temozolomide. Collectively, our findings suggest that THC activates noncanonical autophagy-mediated apoptosis of melanoma cells, suggesting that cytotoxic autophagy induction with Sativex warrants clinical evaluation for metastatic disease.


Subject(s)
Autophagy , Cannabinoids/chemistry , Melanoma/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cannabidiol , Cannabinol/chemistry , Cell Death , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dacarbazine/analogs & derivatives , Dacarbazine/chemistry , Dronabinol/chemistry , Drug Combinations , Humans , Male , Melanoma/metabolism , Membrane Proteins/metabolism , Mice , Mice, Nude , Microscopy, Confocal , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/metabolism , Plant Extracts/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/metabolism , Temozolomide , ras Proteins/metabolism
9.
Mol Cell Oncol ; 2(3): e980134, 2015.
Article in English | MEDLINE | ID: mdl-27308456

ABSTRACT

In a recent article, we found that Tribbles pseudokinase 3 (TRIB3) plays a tumor suppressor role and that this effect relies on the dysregulation of the phosphorylation of v-akt murine thymoma viral oncogene homolog (AKT) by the mammalian target of rapamycin complex 2 (mTORC2 complex), and the subsequent hyperphosphorylation and inactivation of the transcription factor Forkhead box O3 (FOXO3).

10.
Traffic ; 15(8): 839-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24831241

ABSTRACT

In the liver, the P-type ATPase and membrane pump ATP7B plays a crucial role in Cu(+) donation to cuproenzymes and in the elimination of excess Cu(+). ATP7B is endowed with a COOH-cytoplasmic (DE)XXXLL-type traffic signal. We find that accessory (Lys -3, Trp -2, Ser -1 and Leu +2) and canonical (D -4, Leu 0 and Leu +1) residues confer the DKWSLLL signal with the versatility required for the Cu(+)-regulated cycling of ATP7B between the trans-Golgi network (TGN) and the plasma membrane (PM). The separate mutation of these residues caused a disruption of the signal, resulting in different ATP7B distribution phenotypes. These phenotypes indicate the key roles of specific residues at separate steps of ATP7B trafficking, including sorting at the TGN, transport from the TGN to the PM and its endocytosis, and recycling to the TGN and PM. The distinct roles of ATP7B in the TGN and PM and the variety of phenotypes caused by the mutation of the canonical and accessory residues of the DKWSLLL signal can explain the separate or joined presentation of Wilson's cuprotoxicosis and the dysfunction of the cuproenzymes that accept Cu(+) at the TGN.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/metabolism , Protein Sorting Signals , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Animals , COS Cells , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Copper-Transporting ATPases , Hep G2 Cells , Humans , Mice , Protein Transport , trans-Golgi Network/metabolism
11.
Biochim Biophys Acta ; 1831(10): 1573-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23567453

ABSTRACT

Δ(9)-Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoids inhibit tumor growth in animal models of cancer. This effect relies, at least in part, on the up-regulation of several endoplasmic reticulum stress-related proteins including the pseudokinase tribbles homologue-3 (TRIB3), which leads in turn to the inhibition of the AKT/mTORC1 axis and the subsequent stimulation of autophagy-mediated apoptosis in tumor cells. Here, we took advantage of the use of cells derived from Trib3-deficient mice to investigate the precise mechanisms by which TRIB3 regulates the anti-cancer action of THC. Our data show that RasV(12)/E1A-transformed embryonic fibroblasts derived from Trib3-deficient mice are resistant to THC-induced cell death. We also show that genetic inactivation of this protein abolishes the ability of THC to inhibit the phosphorylation of AKT and several of its downstream targets, including those involved in the regulation of the AKT/mammalian target of rapamycin complex 1 (mTORC1) axis. Our data support the idea that THC-induced TRIB3 up-regulation inhibits AKT phosphorylation by regulating the accessibility of AKT to its upstream activatory kinase (the mammalian target of rapamycin complex 2; mTORC2). Finally, we found that tumors generated by inoculation of Trib3-deficient cells in nude mice are resistant to THC anticancer action. Altogether, the observations presented here strongly support that TRIB3 plays a crucial role on THC anti-neoplastic activity. This article is part of a Special Issue entitled Lipid Metabolism in Cancer.


Subject(s)
Cell Cycle Proteins/physiology , Dronabinol/pharmacology , Neoplasms, Experimental/prevention & control , Animals , Autophagy , Cell Death/drug effects , Cells, Cultured , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Mice, Nude , Multiprotein Complexes/metabolism , Neoplasms, Experimental/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
12.
Autophagy ; 7(9): 1071-3, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21593591

ABSTRACT

Δ9-tetrahydrocannabinol (THC), the main active component of marijuana, promotes cancer cell death via autophagy stimulation. We find that activation of the tyrosine kinase receptor ALK by its ligand midkine interferes with the signaling mechanism by which THC promotes autophagy-mediated glioma cell death.


Subject(s)
Autophagy/drug effects , Cytokines/pharmacology , Glioma/enzymology , Glioma/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Dronabinol/pharmacology , Humans , Midkine , Models, Biological
13.
Methods Enzymol ; 489: 297-317, 2011.
Article in English | MEDLINE | ID: mdl-21266237

ABSTRACT

Different physiological and pathological situations that produce alterations in the endoplasmic reticulum, lead to a condition known as ER stress. ER stress activates a complex intracellular signal transduction pathway, called unfolded protein response (UPR). UPR is tailored essentially to reestablish ER homeostasis. However, when persistent, ER stress can switch the cytoprotective functions of UPR into cell death promoting mechanisms. One of the cellular mechanisms that are regulated by ER stress is autophagy. Autophagy is a cellular process by which different cytoplasmic components including organelles are targeted for degradation to the autophagosomes. Interestingly, like ER stress, autophagy can be a protective or a cell death promoting mechanism. Recently, a variety of anticancer therapies (including those that stimulate ER stress) have been shown to activate autophagy in tumor cells, which has been proposed to either enhance cancer cell death or act as a mechanism of resistance to chemotherapy. In this chapter, we will describe some of the procedures that are currently used to analyze autophagy as well as some of the experimental approaches that can be undertaken to investigate the connection between ER stress and autophagy in cancer.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/physiology , Neoplasms/pathology , Signal Transduction/physiology , Unfolded Protein Response , Animals , Cells, Cultured , Endoplasmic Reticulum/pathology , Fluorescent Antibody Technique , Humans , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/physiology , Multiprotein Complexes , Neoplasm Transplantation , Proteins/metabolism , RNA Interference , Stress, Physiological , TOR Serine-Threonine Kinases , Transplantation, Heterologous
14.
Mol Cancer Ther ; 10(1): 90-103, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21220494

ABSTRACT

Glioblastoma multiforme (GBM) is highly resistant to current anticancer treatments, which makes it crucial to find new therapeutic strategies aimed at improving the poor prognosis of patients suffering from this disease. Δ(9)-Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoid receptor agonists inhibit tumor growth in animal models of cancer, including glioma, an effect that relies, at least in part, on the stimulation of autophagy-mediated apoptosis in tumor cells. Here, we show that the combined administration of THC and temozolomide (TMZ; the benchmark agent for the management of GBM) exerts a strong antitumoral action in glioma xenografts, an effect that is also observed in tumors that are resistant to TMZ treatment. Combined administration of THC and TMZ enhanced autophagy, whereas pharmacologic or genetic inhibition of this process prevented TMZ + THC-induced cell death, supporting that activation of autophagy plays a crucial role on the mechanism of action of this drug combination. Administration of submaximal doses of THC and cannabidiol (CBD; another plant-derived cannabinoid that also induces glioma cell death through a mechanism of action different from that of THC) remarkably reduces the growth of glioma xenografts. Moreover, treatment with TMZ and submaximal doses of THC and CBD produced a strong antitumoral action in both TMZ-sensitive and TMZ-resistant tumors. Altogether, our findings support that the combined administration of TMZ and cannabinoids could be therapeutically exploited for the management of GBM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Dronabinol/pharmacology , Glioblastoma/drug therapy , Animals , Autophagy/drug effects , Brain Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Dronabinol/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Random Allocation , Temozolomide , Transfection , Xenograft Model Antitumor Assays
16.
J Clin Invest ; 119(5): 1359-72, 2009 May.
Article in English | MEDLINE | ID: mdl-19425170

ABSTRACT

Autophagy can promote cell survival or cell death, but the molecular basis underlying its dual role in cancer remains obscure. Here we demonstrate that delta(9)-tetrahydrocannabinol (THC), the main active component of marijuana, induces human glioma cell death through stimulation of autophagy. Our data indicate that THC induced ceramide accumulation and eukaryotic translation initiation factor 2alpha (eIF2alpha) phosphorylation and thereby activated an ER stress response that promoted autophagy via tribbles homolog 3-dependent (TRB3-dependent) inhibition of the Akt/mammalian target of rapamycin complex 1 (mTORC1) axis. We also showed that autophagy is upstream of apoptosis in cannabinoid-induced human and mouse cancer cell death and that activation of this pathway was necessary for the antitumor action of cannabinoids in vivo. These findings describe a mechanism by which THC can promote the autophagic death of human and mouse cancer cells and provide evidence that cannabinoid administration may be an effective therapeutic strategy for targeting human cancers.


Subject(s)
Autophagy/drug effects , Cannabinoids/pharmacology , Endoplasmic Reticulum/drug effects , Glioma/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cannabinoids/therapeutic use , Caspase 3/metabolism , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Line, Transformed , Cell Line, Tumor , Dronabinol/pharmacology , Dronabinol/therapeutic use , Endoplasmic Reticulum/pathology , Enzyme Inhibitors/pharmacology , Eukaryotic Initiation Factor-2/metabolism , Glioma/drug therapy , Glioma/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Microtubule-Associated Proteins/metabolism , Models, Biological , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
17.
Glia ; 57(13): 1374-85, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19229996

ABSTRACT

Gliomas, one of the most malignant forms of cancer, exhibit high resistance to conventional therapies. Identification of the molecular mechanisms responsible for this resistance is therefore of great interest to improve the efficacy of the treatments against these tumors. Delta9-Tetrahydrocannabinol (THC), the major active ingredient of marijuana, and other cannabinoids inhibit tumor growth in animal models of cancer, including glioma, an effect that relies, at least in part, on the ability of these compounds to induce apoptosis of tumor cells. By analyzing the gene expression profile of two sub-clones of C6 glioma cells with different sensitivity to cannabinoid-induced apoptosis, we found a subset of genes with a marked differential expression in the two sub-clones. Furthermore, we identified the epidermal growth factor receptor ligand amphiregulin as a candidate factor to mediate the resistance of glioma cells to cannabinoid treatment. Amphiregulin was highly overexpressed in the cannabinoid-resistant cell line, both in culture and in tumor xenografts. Moreover, in vivo silencing of amphiregulin rendered the resistant tumors xenografts sensitive to cannabinoid antitumoral action. Amphiregulin expression was associated with increased extracellular signal-regulated kinase (ERK) activation, which mediated the resistance to THC by blunting the expression of p8 and TRB3-two genes involved in cannabinoid-induced apoptosis of glioma cells. Our findings therefore identify Amphirregulin as a factor for resistance of glioma cells to THC-induced apoptosis and contribute to unraveling the molecular bases underlying the emerging notion that targeted inhibition of the EGFR pathway can improve the efficacy of antitumoral therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dronabinol/pharmacology , Glioma/drug therapy , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Amphiregulin , Animals , Apoptosis/genetics , Apoptosis/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , EGF Family of Proteins , Enzyme Activation/genetics , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Glioma/genetics , Glioma/pathology , Glioma/physiopathology , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Xenograft Model Antitumor Assays
18.
J Cell Physiol ; 218(2): 416-26, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18932217

ABSTRACT

We have previously reported the physical interaction between Daxx, the adaptor protein that mediates activation of the Jun amino-terminal kinase (JNK), and GLUT4, the insulin-dependent glucose transporter, interaction that involves their C-domains. Co-immunoprecipitation and two-hybrid-based protein-protein interaction studies show now that Daxx and GLUT4 interact with JNK1 through D-sites in their NH(2)-(aa 1-501) and large endofacial loop, respectively. Serum deprivation strongly enhances the association of JNK1 with Daxx and dissociates the kinase from GLUT4. SP600125, a potent JNK1 inhibitor, reduces the JNK1 activity associated with GLUT4 and the phosphorylation of two minor GLUT4 species in serum-starved 3T3-L1 adipocytes. In addition, Daxx interacts with kinesin KIF5B through the 6xTPR domain of the kinesin light chain, a domain engaged in the grab hold of protein cargo by kinesin motors that codistribute with JNK. Depletion of Daxx in 3T3-L1 adipocytes provokes the partial translocation of the GLUT4 retained in the GLUT4 storage compartment to endosomes.


Subject(s)
Carrier Proteins/metabolism , Glucose Transporter Type 4/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kinesins/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Nuclear Proteins/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/enzymology , Animals , Anthracenes/pharmacology , Carrier Proteins/chemistry , Co-Repressor Proteins , Glucose Transporter Type 4/chemistry , Humans , Immunoprecipitation , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Mice , Microtubules/drug effects , Microtubules/metabolism , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/chemistry , Molecular Chaperones , Nuclear Proteins/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Serum
SELECTION OF CITATIONS
SEARCH DETAIL
...