Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Biol Chem ; 287(28): 23852-63, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22532565

ABSTRACT

Lipid droplets (LDs) are ubiquitous organelles storing neutral lipids, including triacylglycerol (TAG) and cholesterol ester. The properties of LDs vary greatly among tissues, and LD-binding proteins, the perilipin family in particular, play critical roles in determining such diversity. Overaccumulation of TAG in LDs of non-adipose tissues may cause lipotoxicity, leading to diseases such as diabetes and cardiomyopathy. However, the physiological significance of non-adipose LDs in a normal state is poorly understood. To address this issue, we generated and characterized mice deficient in perilipin 5 (Plin5), a member of the perilipin family particularly abundant in the heart. The mutant mice lacked detectable LDs, containing significantly less TAG in the heart. Particulate structures containing another LD-binding protein, Plin2, but negative for lipid staining, remained in mutant mice hearts. LDs were recovered by perfusing the heart with an inhibitor of lipase. Cultured cardiomyocytes from Plin5-null mice more actively oxidized fatty acid than those of wild-type mice. Production of reactive oxygen species was increased in the mutant mice hearts, leading to a greater decline in heart function with age. This was, however, reduced by the administration of N-acetylcysteine, a precursor of an antioxidant, glutathione. Thus, we conclude that Plin5 is essential for maintaining LDs at detectable sizes in the heart, by antagonizing lipase(s). LDs in turn prevent excess reactive oxygen species production by sequestering fatty acid from oxidation and hence suppress oxidative burden to the heart.


Subject(s)
Fatty Acids/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Female , Free Radical Scavengers/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Lipase/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Muscle Proteins/genetics , Myocardium/cytology , Myocardium/ultrastructure , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress , Triglycerides/metabolism
2.
Cell Metab ; 35(4): 620-632.e5, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36812915

ABSTRACT

How exercise elicits systemic metabolic benefits in both muscles and non-contractile tissues is unclear. Autophagy is a stress-induced lysosomal degradation pathway that mediates protein and organelle turnover and metabolic adaptation. Exercise activates autophagy in not only contracting muscles but also non-contractile tissues including the liver. However, the role and mechanism of exercise-activated autophagy in non-contractile tissues remain mysterious. Here, we show that hepatic autophagy activation is essential for exercise-induced metabolic benefits. Plasma or serum from exercised mice is sufficient to activate autophagy in cells. By proteomic studies, we identify fibronectin (FN1), which was previously considered as an extracellular matrix protein, as an exercise-induced, muscle-secreted, autophagy-inducing circulating factor. Muscle-secreted FN1 mediates exercise-induced hepatic autophagy and systemic insulin sensitization via the hepatic receptor α5ß1 integrin and the downstream IKKα/ß-JNK1-BECN1 pathway. Thus, we demonstrate that hepatic autophagy activation drives exercise-induced metabolic benefits against diabetes via muscle-secreted soluble FN1 and hepatic α5ß1 integrin signaling.


Subject(s)
Fibronectins , Proteomics , Mice , Animals , Fibronectins/metabolism , Liver/metabolism , Autophagy , Integrins
4.
Front Cell Dev Biol ; 10: 844481, 2022.
Article in English | MEDLINE | ID: mdl-35646940

ABSTRACT

Autophagy is a stress-induced lysosomal degradation pathway regulated by evolutionarily conserved autophagy-related (ATG) genes. Recent research has revealed that autophagy plays an important role in the regulation of energy metabolism, development of metabolic tissues, and pathogenesis of metabolic disorders. Bulk and selective degradation by autophagy helps maintain protein homeostasis and physiological function of cells. Aside from classical degradative roles, ATG proteins also carry out non-classical secretory functions of metabolic tissues. In this review, we summarize recent progresses and unanswered questions on the mechanisms of autophagy and ATG proteins in metabolic regulation, with a focus on organelle and nutrient storage degradation, as well as vesicular and hormonal secretion. Such knowledge broadens our understanding on the cause, pathophysiology, and prevention of metabolic diseases including obesity and diabetes.

5.
Autophagy ; 17(10): 3262-3263, 2021 10.
Article in English | MEDLINE | ID: mdl-34281478

ABSTRACT

Macroautophagy/autophagy is primarily considered as a degradative pathway via the lysosome, yet the secretory functions of autophagy proteins have recently been unveiled. Autophagy proteins have been implicated in metabolic organ development, homeostasis and function, and deficiency in autophagy is associated with metabolic disorders. However, the molecular mechanisms by which autophagy proteins regulate energy metabolism and insulin sensitivity were unclear. We previously showed that systemic activation of autophagy by a hyperactive BECN1F121A mutant reduces insulin storage in islets but improves insulin sensitivity systemically. In our recent study, we found that BECN1 functions in adipose tissue to systemically regulate energy metabolism. Adipose-specific expression of BECN1F121A is sufficient to improve systemic insulin sensitivity without negatively affecting pancreatic insulin storage. We demonstrated that BECN1 interacts with exocyst subunit proteins and facilitates the secretion of an adipokine, ADIPOQ (adiponectin, C1Q and collagen domain containing), in adipose tissue. Thus, our findings suggest that BECN1 regulates insulin sensitivity in a non-degradative and non-cell autonomous manner by facilitating ADIPOQ secretion. Our study also highlighted the distinct functions of autophagy proteins in different metabolic tissues.


Subject(s)
Autophagy , Insulin Resistance , Adiponectin , Animals , Beclin-1/metabolism , Insulin/metabolism , Lysosomes/metabolism , Mice
6.
Cell Rep ; 35(8): 109184, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34038729

ABSTRACT

Autophagy dysregulation is implicated in metabolic diseases, including type 2 diabetes. However, the mechanism by which the autophagy machinery regulates metabolism is largely unknown. Autophagy is generally considered a degradation process via lysosomes. Here, we unveil a metabolically important non-cell-autonomous, non-degradative mechanism regulated by the essential autophagy protein Becn1 in adipose tissue. Upon high-fat diet challenge, autophagy-hyperactive Becn1F121A mice show systemically improved insulin sensitivity and enhanced activation of AMP-activated protein kinase (AMPK), a central regulator of energy homeostasis, via a non-cell-autonomous mechanism mediated by adiponectin, an adipose-derived metabolic hormone. Adipose-specific Becn1F121A expression is sufficient to activate AMPK in non-adipose tissues and improve systemic insulin sensitivity by increasing adiponectin secretion. Further, Becn1 enhances adiponectin secretion by interacting with components of the exocyst complex via the coiled-coil domain. Together, our study demonstrates that Becn1 improves insulin sensitivity by facilitating adiponectin secretion through binding the exocyst in adipose tissue.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Beclin-1/metabolism , Insulin/metabolism , Lysosomes/metabolism , Animals , Autophagy , Humans , Mice , Transfection
7.
Genes (Basel) ; 12(1)2021 01 14.
Article in English | MEDLINE | ID: mdl-33466690

ABSTRACT

Cancer stem cells (CSCs), having both self-renewal and tumorigenic capacity, utilize an energy metabolism system different from that of non-CSCs. Lipid droplets (LDs) are organelles that store neutral lipids, including triacylglycerol. Previous studies demonstrated that LDs are formed and store lipids as an energy source in some CSCs. LDs play central roles not only in lipid storage, but also as a source of endogenous lipid ligands, which are involved in numerous signaling pathways, including the peroxisome proliferator-activated receptor (PPAR) signaling pathway. However, it remains unclear whether LD-derived signal transduction is involved in the maintenance of the properties of CSCs. We investigated the roles of LDs in cancer stemness using pancreatic and colorectal CSCs and isogenic non-CSCs. PPARα was activated in CSCs in which LDs accumulated, but not in non-CSCs, and pharmacological and genetic inhibition of PPARα suppressed cancer stemness. In addition, inhibition of both re-esterification and lipolysis pathways suppressed cancer stemness. Our study suggested that LD metabolic turnover accompanying PPARα activation is a promising anti-CSC therapeutic target.


Subject(s)
Colorectal Neoplasms/metabolism , Lipid Droplets/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , PPAR alpha/metabolism , Pancreatic Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Energy Metabolism , HT29 Cells , Humans , Lipid Droplets/pathology , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , PPAR alpha/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction
8.
Sci Adv ; 7(8)2021 02.
Article in English | MEDLINE | ID: mdl-33608268

ABSTRACT

Drug abuse is a foremost public health problem. Cocaine is a widely abused drug worldwide that produces various reward-related behaviors. The mechanisms that underlie cocaine-induced disorders are unresolved, and effective treatments are lacking. Here, we found that an autophagy-related protein Becn2 is a previously unidentified regulator of cocaine reward behaviors. Becn2 deletion protects mice from cocaine-stimulated locomotion and reward behaviors, as well as cocaine-induced dopamine accumulation and signaling, by increasing presynaptic dopamine receptor 2 (D2R) autoreceptors in dopamine neurons. Becn2 regulates D2R endolysosomal trafficking, degradation, and cocaine-induced behaviors via interacting with a D2R-bound adaptor GASP1. Inactivating Becn2 by upstream autophagy inhibitors stabilizes striatal presynaptic D2R, reduces dopamine release and signaling, and prevents cocaine reward in normal mice. Thus, the autophagy protein Becn2 is essential for cocaine psychomotor stimulation and reward through regulating dopamine neurotransmission, and targeting Becn2 by autophagy inhibitors is a potential strategy to prevent cocaine-induced behaviors.


Subject(s)
Cocaine , Animals , Autophagy-Related Proteins , Cocaine/pharmacology , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Reward
9.
FEBS J ; 287(10): 2023-2036, 2020 05.
Article in English | MEDLINE | ID: mdl-31868973

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant primary brain tumour in adults. Since glioma stem cells (GSCs) are associated with therapeutic resistance as well as the initiation and recurrence in GBM, therapies targeting GSCs are considered to be effective for long-term survival in GBM. Several reports suggested that oxidative phosphorylation (OXPHOS) of cancer stem cells is important for their survival; however, the requirement of OXPHOS in GSCs remains unclear. Few effective and safe agents that target GSC mitochondria are available in clinical settings. In this study, we demonstrated that GSCs had high OXPHOS activity compared with isogenic differentiated GSCs and that GSC survival depended on their OXPHOS activity. Remarkably, we showed that complexes III and IV had broad therapeutic windows and that the expression levels of mitochondrial DNA-coded components of complexes III and IV were elevated in GSCs compared with differentiated GSCs. Moreover, our search of the Food and Drug Administration-approved drugs for those targeting GSC mitochondria revealed that verteporfin (Visudyne® ), a drug approved for macular degeneration, was a novel GSC-specific cytotoxic compound that reduced OXPHOS activity. Importantly, the cytotoxic effect of verteporfin was specific to GSCs without any toxicity to normal cells, and the IC50 of approximately 200 nm was ten times less than its maximum blood concentration in humans. Overall, these findings indicated that high mitochondrial OXPHOS of GSCs is a potential GSC-specific vulnerability and that clinically available drugs, such as verteporfin, might become novel GSC-specific cytotoxic agents.


Subject(s)
Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Oxidative Phosphorylation/drug effects , Verteporfin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Xenograft Model Antitumor Assays
10.
Autophagy ; 14(11): 2026-2028, 2018.
Article in English | MEDLINE | ID: mdl-30081744

ABSTRACT

Macroautophagy/autophagy abnormality has been recently associated with metabolic disorders, such as type 2 diabetes (T2D). However, the effect of autophagy activation in systemic energy metabolism was poorly understood. In our recent study, we demonstrated that autophagy plays different roles in distinct metabolic tissues, using an autophagy-hyperactive mouse model. In insulin-producing ß cells, excess autophagy degrades insulin-containing vesicles (a process termed vesicophagy), resulting in decreased insulin contents and systemic glucose intolerance; whereas in insulin-responsive cells, activating autophagy decreases endoplasmic reticulum (ER) stress and improves insulin sensitivity.


Subject(s)
Autophagy , Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Beclin-1 , Endoplasmic Reticulum Stress , Insulin , Insulin Secretion , Mice , Proto-Oncogene Proteins c-bcl-2
11.
Anticancer Res ; 38(9): 5093-5099, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30194154

ABSTRACT

BACKGROUND/AIM: Cancer stem cells (CSCs) are associated with tumorigenesis, recurrence, and metastasis. Cell-cell communication via gap junctions (GJs) between metastatic cancer cells and astrocytes is necessary for brain metastasis. Agents targeting communication between CSCs and astrocytes are expected to suppress brain metastasis. MATERIALS AND METHODS: Using the A549 CSC, a cancer stem-like cell derived from A549, we examined the effect of AS602801, an anti-cancer stem cell agent whose safety has been confirmed in a phase 2 clinical trial, on GJ communication and connexin expression using a dye-transfer assay and immunoblot analysis, respectively. RESULTS: AS602801 specifically suppressed cell-cell communication in A549 CSCs without any suppression of GJ communication in astrocytes; it also decreased the expression of connexin 43, a constituent of GJs, in A549 CSCs. CONCLUSION: The anti-cancer stem cell agent, AS602801, is a potential drug candidate against brain metastasis.


Subject(s)
Astrocytes/cytology , Benzothiazoles/pharmacology , Connexin 43/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Pyrimidines/pharmacology , A549 Cells , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain Neoplasms/prevention & control , Cell Communication/drug effects , Down-Regulation , Gap Junctions/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Rats
12.
Oncotarget ; 9(66): 32667-32679, 2018 Aug 24.
Article in English | MEDLINE | ID: mdl-30220973

ABSTRACT

Use of epidermal growth factor receptor (EGFR) inhibitors represented by gefitinib and erlotinib has become the standard of treatment for non-small-cell lung cancers (NSCLCs) with activating EGFR mutations. However, the majority of NSCLCs, which overexpress EGFR without such mutations, are resistant to EGFR inhibitors, and the mechanism(s) behind such primary resistance of NSCLCs without activating EGFR mutations to EGFR inhibitors still remains poorly understood. Here in this study, we show that glucose metabolism mediated by GLUT1, a facilitative glucose transporter, is involved in gefitinib resistance of NSCLC cells. We found that GLUT1 expression and glucose uptake were increased in resistant NSCLC cells after gefitinib treatment and that genetic as well as pharmacological inhibition of GLUT1 sensitized not only NSCLC cells with primary resistance but also those with acquired resistance to gefitinib. In vivo, the combination of systemic gefitinib and a GLUT1 inhibitor, both of which failed to inhibit tumor growth when administered alone, significantly inhibited the growth of xenograft tumors formed by the implantation of NSCLC cells with wild-type EGFR (wt-EGFR). Since our data indicated that GLUT1 was similarly involved in erlotinib resistance, our findings suggest that the activity of GLUT1-mediated glucose metabolism could be a critical determinant for the sensitivity of NSCLC cells to EGFR inhibitors and that concurrent GLUT1 inhibition may therefore be a mechanism-based approach to treating NSCLCs resistant to EGFR inhibitors, including those with wt-EGFR.

13.
Cell Rep ; 23(11): 3286-3299, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898399

ABSTRACT

Autophagy, a stress-induced lysosomal degradative pathway, has been assumed to exert similar metabolic effects in different organs. Here, we establish a model where autophagy plays different roles in insulin-producing ß cells versus insulin-responsive cells, utilizing knockin (Becn1F121A) mice manifesting constitutively active autophagy. With a high-fat-diet challenge, the autophagy-hyperactive mice unexpectedly show impaired glucose tolerance, but improved insulin sensitivity, compared to mice with normal autophagy. Autophagy hyperactivation enhances insulin signaling, via suppressing ER stress in insulin-responsive cells, but decreases insulin secretion by selectively sequestrating and degrading insulin granule vesicles in ß cells, a process we term "vesicophagy." The reduction in insulin storage, insulin secretion, and glucose tolerance is reversed by transient treatment of autophagy inhibitors. Thus, ß cells and insulin-responsive tissues require different autophagy levels for optimal function. To improve insulin sensitivity without hampering secretion, acute or intermittent, rather than chronic, activation of autophagy should be considered in diabetic therapy development.


Subject(s)
Autophagy , Beclin-1/metabolism , Insulin/metabolism , Animals , Autophagosomes/metabolism , Autophagy/drug effects , Beclin-1/genetics , Benzamides/pharmacology , Diet, High-Fat , Endoplasmic Reticulum Stress , Gene Knock-In Techniques , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Pyrimidines/pharmacology , Signal Transduction
14.
Oncotarget ; 8(55): 94872-94882, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29212273

ABSTRACT

CEP-1347 is a mixed lineage kinase inhibitor tested in a large-scale phase 2/3 clinical trial in early Parkinson's disease, in which its safety and tolerability, but nevertheless not efficacy, was demonstrated. Here we identify by drug repositioning CEP-1347 as a potential anti-cancer stem cell drug. In vitro, CEP-1347 efficiently induced differentiation and inhibited the self-renewal and tumor-initiating capacities of human cancer stem cells from glioblastoma as well as from pancreatic and ovarian cancers at clinically-relevant concentrations, without impairing the viability of normal fibroblasts and neural stem cells. In vivo, a 10-day systemic administration of CEP-1347 at a dose that was less than 1/10 the mouse equivalent of the dose safely given to humans for 2 years was sufficient to effectively reduce tumor-initiating cancer stem cells within established tumors in mice. Furthermore, the same treatment protocol significantly extended the survival of mice receiving orthotopic implantation of glioma stem cells. Together, our findings suggest that CEP-1347 is a promising candidate for cancer stem cell-targeting therapy and that further clinical and preclinical studies are warranted to evaluate its efficacy in cancer treatment.

15.
Anticancer Res ; 37(11): 6177-6188, 2017 11.
Article in English | MEDLINE | ID: mdl-29061799

ABSTRACT

BACKGROUND/AIM: Olanzapine, an atypical antipsychotic, is now increasingly used as an off-label indication for the management of cancer patients with chemotherapy-induced nausea and vomiting (CINV). However, how olanzapine affects cancer cells per se remains poorly understood. MATERIALS AND METHODS: The effects of olanzapine treatment and survivin knockdown, alone or in combination with chemotherapeutic agents, on survivin expression and cell viability were investigated in human cancer cell lines. RESULTS: Olanzapine reduced survivin expression in lung and pancreatic cancer stem cell (CSC) lines and sensitized them to chemotherapeutic agents such as 5-fluorouracil, gemcitabine, and cisplatin in a survivin expression-dependent manner. Olanzapine also reduced survivin expression and chemosensitized serum-cultured, non-CSC ovarian cancer cells that expressed survivin. CONCLUSION: Olanzapine may benefit cancer patients not only as an antiemetic for CINV, but also by enhancing the effects of chemotherapeutic agents through down-regulation of survivin, which has been implicated in multidrug chemoresistance.


Subject(s)
Antineoplastic Agents/pharmacology , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Drug Resistance, Neoplasm/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pancreatic Neoplasms/drug therapy , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Olanzapine , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Survivin , Tumor Cells, Cultured
16.
Oncotarget ; 8(53): 90996-91008, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29207619

ABSTRACT

Currently, there is no established therapeutic option for high-grade meningioma recurring after surgery and radiotherapy, and few chemotherapeutic agents are in development for the treatment of high-grade meningioma. Here in this study, we screened a panel of chemotherapeutic agents for their possible antitumor activity in high-grade meningioma and discovered that high-grade meningioma cells show a preferential sensitivity to antimetabolites, in particular, to gemcitabine. In vitro, gemcitabine inhibited the growth of high-grade meningioma cells effectively by inducing S-phase arrest and apoptotic cell death. In vivo, systemic gemcitabine chemotherapy suppressed not only tumor initiation but also inhibited the growth and achieved a long-term control of established tumors in xenograft models of high-grade meningioma. Histological analysis indicated that systemic gemcitabine blocks cell cycle progression and promotes apoptotic cell death in tumor cells in vivo. Together, our data demonstrate that gemcitabine exerts potent antitumor activity against high-grade meningioma through cytostatic and cytotoxic mechanisms. We therefore propose gemcitabine is a promising chemotherapeutic agent that warrants further investigation as a treatment option for high-grade meningioma.

17.
FEBS Open Bio ; 7(6): 835-844, 2017 06.
Article in English | MEDLINE | ID: mdl-28593138

ABSTRACT

Glioblastoma multiforme is the most malignant primary intrinsic brain tumor. Glioma stem cells (GSCs) are associated with chemoradiotherapy resistance and the recurrence of glioblastomas after conventional therapy. The targeting of GSCs is potentially an effective treatment for the long-term survival of glioblastoma patients. Licochalcone A, a natural chalconoid from licorice root, exerts anticancer effects; however, its effect on GSCs remains unknown. We found that Licochalcone A induced massive caspase-dependent death in GSCs but not in differentiated GSCs nor normal somatic and neural stem cells. Prior to cell death, Licochalcone A caused mitochondrial fragmentation and reduced the membrane potential and ATP production in GSCs. Thus, Licochalcone A induces mitochondrial dysfunction and shows promise as an anticancer stem cell drug.

18.
Oncotarget ; 7(19): 27021-32, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27027242

ABSTRACT

A phase 2 clinical trial investigating the efficacy and safety of AS602801, a newly developed JNK inhibitor, in the treatment of inflammatory endometriosis is complete. We are now examining whether AS602801 acts against human cancer cells in vitro and in vivo. In vitro, AS602801 exhibited cytotoxicity against both serum-cultured non-stem cancer cells and cancer stem cells derived from human pancreatic cancer, non-small cell lung cancer, ovarian cancer and glioblastoma at concentrations that did not decrease the viability of normal human fibroblasts. AS602801 also inhibited the self-renewal and tumor-initiating capacity of cancer stem cells surviving AS602801 treatment. Cancer stem cells in established xenograft tumors were reduced by systemic administration of AS602801 at a dose and schedule that did not adversely affect the health of the tumor-bearing mice. These findings suggest AS602801 is a promising anti-cancer stem cell agent, and further investigation of the utility of AS602801 in the treatment of cancer seems warranted.


Subject(s)
Benzothiazoles/pharmacology , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays/methods , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Tumor Burden/drug effects
19.
Oncol Rep ; 35(1): 593-601, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26534836

ABSTRACT

Ovarian cancer is the most lethal gynecological malignancy, for which platinum- and taxane-based chemotherapy plays a major role. Chemoresistance of ovarian cancer poses a major obstacle to the successful management of this devastating disease; however, effective measures to overcome platinum and taxane resistance are yet to be established. In the present study, while investigating the mechanism underlying the chemoresistance of ovarian cancer, we found that JNK may have a key role in the resistance of ovarian cancer cells to cisplatin and paclitaxel. Importantly, whereas simultaneous application of a JNK inhibitor and either of the chemotherapeutic agents had contrasting effects for cisplatin (enhanced cytotoxicity) and paclitaxel (decreased cytotoxicity), JNK inhibitor treatment prior to chemotherapeutic agent application invariably enhanced the cytotoxicity of both drugs, suggesting that the basal JNK activity is commonly involved in the chemoresistance of ovarian cancer cells to cisplatin and paclitaxel in contrast to drug­induced JNK activity which may have different roles for these two drugs. Furthermore, we confirmed using non-transformed human and rodent fibroblasts that sequential application of the JNK inhibitor and the chemotherapeutic agents did not augment their toxicity. Thus, our findings highlight for the first time the possible differential roles of the basal and induced JNK activities in the chemoresistance of ovarian cancer cells and also suggest that time­staggered JNK inhibition may be a rational and promising strategy to overcome the resistance of ovarian cancer to platinum- and taxane-based chemotherapy.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , MAP Kinase Signaling System/drug effects , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Paclitaxel/pharmacology
20.
Autophagy ; 12(9): 1460-71, 2016 09.
Article in English | MEDLINE | ID: mdl-27305347

ABSTRACT

Cannabinoids and related drugs generate profound behavioral effects (such as analgesic effects) through activating CNR1 (cannabinoid receptor 1 [brain]). However, repeated cannabinoid administration triggers lysosomal degradation of the receptor and rapid development of drug tolerance, limiting the medical use of marijuana in chronic diseases. The pathogenic mechanisms of cannabinoid tolerance are not fully understood, and little is known about its prevention. Here we show that a protein involved in macroautophagy/autophagy (a conserved lysosomal degradation pathway), BECN2 (beclin 2), mediates cannabinoid tolerance by preventing CNR1 recycling and resensitization after prolonged agonist exposure, and deletion of Becn2 rescues CNR1 activity in mouse brain and conveys resistance to analgesic tolerance to chronic cannabinoids. To target BECN2 therapeutically, we established a competitive recruitment model of BECN2 and identified novel synthetic, natural or physiological stimuli of autophagy that sequester BECN2 from its binding with GPRASP1, a receptor protein for CNR1 degradation. Co-administration of these autophagy inducers effectively restores the level and signaling of brain CNR1 and protects mice from developing tolerance to repeated cannabinoid usage. Overall, our findings demonstrate the functional link among autophagy, receptor signaling and animal behavior regulated by psychoactive drugs, and develop a new strategy to prevent tolerance and improve medical efficacy of cannabinoids by modulating the BECN2 interactome and autophagy activity.


Subject(s)
Autophagy/drug effects , Cannabinoids/metabolism , Drug Tolerance , Intracellular Signaling Peptides and Proteins/metabolism , Receptor, Cannabinoid, CB1/metabolism , Analgesics/chemistry , Animals , Behavior, Animal , Brain/metabolism , Gene Deletion , HEK293 Cells , HeLa Cells , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Management , Protein Transport , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL