Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Trends Endocrinol Metab ; 34(3): 170-180, 2023 03.
Article in English | MEDLINE | ID: mdl-36732094

ABSTRACT

mTORC1, the mammalian target of rapamycin complex 1, is a key regulator of cellular physiology. The lipid metabolite phosphatidic acid (PA) binds to and activates mTORC1 in response to nutrients and growth factors. We review structural findings and propose a model for PA activation of mTORC1. PA binds to a highly conserved sequence in the α4 helix of the FK506 binding protein 12 (FKBP12)/rapamycin-binding (FRB) domain of mTOR. It is proposed that PA binding to two adjacent positively charged amino acids breaks and shortens the C-terminal region of helix α4. This has profound consequences for both substrate binding and the catalytic activity of mTORC1.


Subject(s)
Phosphatidic Acids , TOR Serine-Threonine Kinases , Humans , Phosphatidic Acids/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Amino Acids/metabolism
2.
PLoS One ; 17(10): e0276579, 2022.
Article in English | MEDLINE | ID: mdl-36269753

ABSTRACT

Metabolic reprogramming is now considered a hallmark of cancer cells. KRas-driven cancer cells use glutaminolysis to generate the tricarboxylic acid cycle intermediate α-ketoglutarate via a transamination reaction between glutamate and oxaloacetate. We reported previously that exogenously supplied unsaturated fatty acids could be used to synthesize phosphatidic acid-a lipid second messenger that activates both mammalian target of rapamycin (mTOR) complex 1 (mTORC1) and mTOR complex 2 (mTORC2). A key target of mTORC2 is Akt-a kinase that promotes survival and regulates cell metabolism. We report here that mono-unsaturated oleic acid stimulates the phosphorylation of ATP citrate lyase (ACLY) at the Akt phosphorylation site at S455 in an mTORC2 dependent manner. Inhibition of ACLY in KRas-driven cancer cells in the absence of serum resulted in loss of cell viability. We examined the impact of glutamine (Gln) deprivation in combination with inhibition of ACLY on the viability of KRas-driven cancer cells. While Gln deprivation was somewhat toxic to KRas-driven cancer cells by itself, addition of the ACLY inhibitor SB-204990 increased the loss of cell viability. However, the transaminase inhibitor aminooxyacetate was minimally toxic and the combination of SB-204990 and aminooxtacetate led to significant loss of cell viability and strong cleavage of poly-ADP ribose polymerase-indicating apoptotic cell death. This effect was not observed in MCF7 breast cancer cells that do not have a KRas mutation or in BJ-hTERT human fibroblasts which have no oncogenic mutation. These data reveal a synthetic lethality between inhibition of glutamate oxaloacetate transaminase and ACLY inhibition that is specific for KRas-driven cancer cells and the apparent metabolic reprogramming induced by activating mutations to KRas.


Subject(s)
ATP Citrate (pro-S)-Lyase , Glutamine , Neoplasms , Humans , Adenosine Diphosphate Ribose , Aminooxyacetic Acid , ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Glutamates/genetics , Glutamine/antagonists & inhibitors , Glutamine/metabolism , Ketoglutaric Acids , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oleic Acids , Oxaloacetates , Phosphatidic Acids , Proto-Oncogene Proteins c-akt/metabolism , Transaminases/genetics
4.
Cancer Lett ; 522: 164-170, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34563639

ABSTRACT

Inhibition of mammalian target of rapamycin complex 1 (mTORC1) with rapamycin in the absence of transforming growth factor-ß (TGFß) signaling induces apoptosis in many cancer cell lines. In the presence of TGFß, rapamycin induces G1 cell cycle arrest; however, in the absence of TGFß, cells do not arrest in G1 and progress into S-phase where rapamycin is cytotoxic rather than cytostatic. However, we observed that DU145 prostate and NCI-H2228 lung cancer cells were resistant to the cytotoxic effect of rapamycin. Of interest, the rapamycin-resistant DU145 and NCI-H2228 cells have mutations in the RB and CDKN2A tumor suppressor genes. The gene products of RB and CDKN2A (pRb and p14ARF) suppress E2F family transcription factors that promote cell cycle progression from G1 into S. Restoration of wild type RB or inhibition of E2F activity in DU145 and NCI-H2228 cells led to rapamycin sensitivity. These data provide evidence that the combination of mutant RB and mutant CDKN2A in cancer cells leads to rapamycin resistance, which has implications for precision medicine approaches to anti-cancer therapies.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Lung Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Retinoblastoma Protein/genetics , Transforming Growth Factor beta/genetics , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , E2F Transcription Factors/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mutation/genetics , Phosphorylation/drug effects , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Sirolimus/adverse effects , Sirolimus/pharmacology
5.
Cell Cycle ; 19(17): 2158-2167, 2020 09.
Article in English | MEDLINE | ID: mdl-32715871

ABSTRACT

G1 cell cycle progression is controlled largely by growth factors in early G1 indicating that it is appropriate to divide and by nutrients in late G1 indicating sufficient raw material for cell division. We previously mapped a late G1 cell cycle checkpoint for lipids upstream from a mammalian target of rapamycin complex 1 (mTORC1)-mediated checkpoint and downstream from a mid-G1 checkpoint known as the Restriction point. We therefore investigated a role for lipids in progression through late G1 into S-phase. Quiescent BJ-hTERT human fibroblasts were primed with 10% fetal bovine serum (FBS) for 3.5 h at which time, cells were treated with a mixture of lipids and carrier bovine serum albumin (BSA) along with [3 H]-thymidine deoxyribose ([3 H]-TdR) to monitor progression into S-phase. Surprisingly, BSA by itself was more effective than FBS in promoting progression to S-phase - the lipids had no impact on progression. While insulin strongly stimulated mTORC1 activity, it did not impact on [3 H]-TdR incorporation. Although BSA modestly elevated mTORC1 activity, rapamycin strongly inhibited BSA-induced progression to S-phase. BSA treatment promoted mitosis, but not progression through a second G1. Thus, after priming quiescent cells with FBS, albumin was sufficient to promote progression into S-phase. The BSA was not simply a source of amino acids in that amino acids were present in the culture media. We propose that the presence of albumin - the most abundant protein in serum - reflects a broader availability of essential amino acids needed for cell growth.


Subject(s)
Fibroblasts/cytology , G1 Phase , Intercellular Signaling Peptides and Proteins/pharmacology , S Phase , Serum Albumin, Bovine/pharmacology , Amino Acids/pharmacology , Animals , Cattle , Cell Death/drug effects , Cell Line , Fibroblasts/drug effects , G1 Phase/drug effects , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pinocytosis/drug effects , S Phase/drug effects
6.
J Biol Chem ; 295(1): 263-274, 2020 01 03.
Article in English | MEDLINE | ID: mdl-31767684

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) promotes cell growth and proliferation in response to nutrients and growth factors. Amino acids induce lysosomal translocation of mTORC1 via the Rag GTPases. Growth factors activate Ras homolog enriched in brain (Rheb), which in turn activates mTORC1 at the lysosome. Amino acids and growth factors also induce the phospholipase D (PLD)-phosphatidic acid (PA) pathway, required for mTORC1 signaling through mechanisms that are not fully understood. Here, using human and murine cell lines, along with immunofluorescence, confocal microscopy, endocytosis, PLD activity, and cell viability assays, we show that exogenously supplied PA vesicles deliver mTORC1 to the lysosome in the absence of amino acids, Rag GTPases, growth factors, and Rheb. Of note, pharmacological or genetic inhibition of endogenous PLD prevented mTORC1 lysosomal translocation. We observed that precancerous cells with constitutive Rheb activation through loss of tuberous sclerosis complex subunit 2 (TSC2) exploit the PLD-PA pathway and thereby sustain mTORC1 activation at the lysosome in the absence of amino acids. Our findings indicate that sequential inputs from amino acids and growth factors trigger PA production required for mTORC1 translocation and activation at the lysosome.


Subject(s)
Amino Acids/deficiency , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidic Acids/metabolism , Amino Acids/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Endocytosis , Humans , Mice , Phospholipase D/metabolism , Protein Transport , Ras Homolog Enriched in Brain Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism
7.
PLoS One ; 14(8): e0216024, 2019.
Article in English | MEDLINE | ID: mdl-31412041

ABSTRACT

(-)-Oleocanthal (oleocanthal) is a phenolic compound found in varying concentrations in extra virgin olive oil oleocanthal has been shown to be active physiologically, benefiting several diseased states by conferring anti-inflammatory and neuroprotective benefits. Recently, we and other groups have demonstrated its specific and selective toxicity toward cancer cells; however, the mechanism leading to cancer cell death is still disputed. The current study demonstrates that oleocanthal, as well as naturally oleocanthal-rich extra virgin olive oils, induced damage to cancer cells' lysosomes leading to cellular toxicity in vitro and in vivo. Lysosomal membrane permeabilization following oleocanthal treatment in various cell lines was assayed via three complementary methods. Additionally, we found oleocanthal treatment reduced tumor burden and extended lifespan of mice engineered to develop pancreatic neuroendocrine tumors. Finally, following-up on numerous correlative studies demonstrating consumption of olive oil reduces cancer incidence and morbidity, we observed that extra virgin olive oils naturally rich in oleocanthal sharply reduced cancer cell viability and induced lysosomal membrane permeabilization while oleocanthal-poor oils did not. Our results are especially encouraging since tumor cells often have larger and more numerous lysosomes, making them especially vulnerable to lysosomotropic agents such as oleocanthal.


Subject(s)
Aldehydes/administration & dosage , Brain Neoplasms/drug therapy , Cell Membrane Permeability/drug effects , Cyclopentane Monoterpenes/administration & dosage , Lysosomes/drug effects , Neuroectodermal Tumors, Primitive/drug therapy , Olive Oil/administration & dosage , Phenols/administration & dosage , Plant Oils/administration & dosage , Animals , Apoptosis , Brain Neoplasms/pathology , Lysosomes/metabolism , Mice , Necrosis , Neuroectodermal Tumors, Primitive/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Trends Endocrinol Metab ; 30(6): 357-368, 2019 06.
Article in English | MEDLINE | ID: mdl-31040047

ABSTRACT

Cancer cells consume glutamine, a nonessential amino acid (NEAA), at exceedingly high rates to fulfill their energetic and biosynthetic requirements for proliferation. Glutamine plays distinct roles from essential amino acids in cell cycle progression and in the activation of mammalian target of rapamycin (mTOR). Furthermore, the need of cancer cells for glutamine can be exploited therapeutically - especially those driven by KRas. In this review we explore several distinct cellular roles for glutamine that contribute to glutamine addiction in KRas-driven cancer cells and discuss opportunities for therapeutic intervention created by glutamine addiction.


Subject(s)
Amino Acids, Essential/metabolism , Glutamine/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Humans , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
9.
Am J Bot ; 105(11): 1929-1937, 2018 11.
Article in English | MEDLINE | ID: mdl-30418663

ABSTRACT

PREMISE OF THE STUDY: The global climate during the early Miocene was warmer than the present and preceded the even warmer middle Miocene climatic optimum. The paleo-CO2 records for this interval suggest paradoxically low concentrations (<450 ppm) that are difficult to reconcile with a warmer-than-present global climate. METHODS: In this study, we use a leaf gas-exchange model to estimate CO2 concentrations using stomatal characteristics of fossil leaves from a late early Miocene Neotropical assemblage from Panama that we date to 18.01 ± 0.17 Ma via 238 U/206 Pb zircon geochronology. We first validated the model for Neotropical environments by estimating CO2 from canopy leaves of 21 extant species in a natural Panamanian forest and from leaves of seven Neotropical species in greenhouse experiments at 400 and 700 ppm. KEY RESULTS: The results showed that the most probable combined CO2 estimate from the natural forests and 400 ppm experiments is 475 ppm, and for the 700 ppm experiments is 665 ppm. CO2 estimates from the five fossil species exhibit bimodality, with two species most consistent with a low mode (528 ppm) and three with a high mode (912 ppm). CONCLUSIONS: Despite uncertainties, it is very likely (at >95% confidence) that CO2 during the late early Miocene exceeded 400 ppm. These results revise upwards the likely CO2 concentration at this time, more in keeping with a CO2 -forced greenhouse climate.


Subject(s)
Atmosphere/chemistry , Carbon Dioxide , Climate , Fossils , Plant Stomata/physiology , Models, Biological
10.
J Biol Chem ; 293(42): 16390-16401, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30194281

ABSTRACT

Glutamine is a key nutrient required for sustaining cell proliferation, contributing to nucleotide, protein, and lipid synthesis. The mTOR complex 1 (mTORC1) is a highly conserved protein complex that acts as a sensor of nutrients, relaying signals for the shift from catabolic to anabolic metabolism. Although glutamine plays an important role in mTORC1 activation, the mechanism is not clear. Here we describe a leucine- and Rag-independent mechanism of mTORC1 activation by glutamine that depends on phospholipase D and the production of phosphatidic acid, which is required for the stability and activity of mTORC1. The phospholipase D-dependent activation of mTORC1 by glutamine depended on the GTPases ADP ribosylation factor 1 (Arf1), RalA, and Rheb. Glutamine deprivation could be rescued by α-ketoglutarate, a downstream metabolite of glutamine. This mechanism represents a distinct nutrient input to mTORC1 that is independent of Rag GTPases and leucine.


Subject(s)
Glutamine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phospholipase D/metabolism , Cell Line , Humans , Mechanistic Target of Rapamycin Complex 1/chemistry , Nutrients/metabolism , Phosphatidic Acids/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , ral GTP-Binding Proteins/metabolism
11.
Cancer Lett ; 423: 28-35, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29524555

ABSTRACT

Prostate cells are hormonally driven to grow and divide. Typical treatments for prostate cancer involve blocking activation of the androgen receptor by androgens. Androgen deprivation therapy can lead to the selection of cancer cells that grow and divide independently of androgen receptor activation. Prostate cancer cells that are insensitive to androgens commonly display metastatic phenotypes and reduced long-term survival of patients. In this study we provide evidence that androgen-insensitive prostate cancer cells have elevated PLD activity relative to the androgen-sensitive prostate cancer cells. PLD activity has been linked with promoting survival in many human cancer cell lines; and consistent with the previous studies, suppression of PLD activity in the prostate cancer cells resulted in apoptotic cell death. Of significance, suppressing the elevated PLD activity in androgen resistant prostate cancer lines also blocked the ability of these cells to migrate and invade Matrigel™. Since survival signals are generally an early event in tumorigenesis, the apparent coupling of survival and metastatic phenotypes implies that metastasis is an earlier event in malignant prostate cancer than generally thought. This finding has implications for screening strategies designed to identify prostate cancers before dissemination.


Subject(s)
Drug Resistance, Neoplasm , Phospholipase D/metabolism , Prostatic Neoplasms/metabolism , Up-Regulation , Androgens/pharmacology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Metastasis
12.
J Biol Chem ; 292(15): 6303-6311, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28223357

ABSTRACT

mTOR, the mammalian target of rapamycin, integrates growth factor and nutrient signals to promote a transformation from catabolic to anabolic metabolism, cell growth, and cell cycle progression. Phosphatidic acid (PA) interacts with the FK506-binding protein-12-rapamycin-binding (FRB) domain of mTOR, which stabilizes both mTOR complexes: mTORC1 and mTORC2. We report here that mTORC1 and mTORC2 are activated in response to exogenously supplied fatty acids via the de novo synthesis of PA, a central metabolite for membrane phospholipid biosynthesis. We examined the impact of exogenously supplied fatty acids on mTOR in KRas-driven cancer cells, which are programmed to utilize exogenous lipids. The induction of mTOR by oleic acid was dependent upon the enzymes responsible for de novo synthesis of PA. Suppression of the de novo synthesis of PA resulted in G1 cell cycle arrest. Although it has long been appreciated that mTOR is a sensor of amino acids and glucose, this study reveals that mTOR also senses the presence of lipids via production of PA.


Subject(s)
Multiprotein Complexes/metabolism , Phosphatidic Acids/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , MCF-7 Cells , Male , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/genetics , Oleic Acid/pharmacology , Phosphatidic Acids/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , TOR Serine-Threonine Kinases/genetics
13.
J Biol Chem ; 292(3): 936-944, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27956548

ABSTRACT

Lipids are important nutrients that proliferating cells require to maintain energy homeostasis as well as to build plasma membranes for newly synthesized cells. Previously, we identified nutrient-sensing checkpoints that exist in the latter part of the G1 phase of the cell cycle that are dependent upon essential amino acids, Gln, and finally, a checkpoint mediated by mammalian target of rapamycin (mTOR), which integrates signals from both nutrients and growth factors. In this study, we have identified and temporally mapped a lipid-mediated G1 checkpoint. This checkpoint is located after the Gln checkpoint and before the mTOR-mediated cell cycle checkpoint. Intriguingly, clear cell renal cell carcinoma cells (ccRCC) have a dysregulated lipid-mediated checkpoint due in part to defective phosphatase and tensin homologue (PTEN). When deprived of lipids, instead of arresting in G1, these cells continue to cycle and utilize lipid droplets as a source of lipids. Lipid droplets have been known to maintain endoplasmic reticulum homeostasis and prevent cytotoxic endoplasmic reticulum stress in ccRCC. Dysregulation of the lipid-mediated checkpoint forces these cells to utilize lipid droplets, which could potentially lead to therapeutic opportunities that exploit this property of ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Membrane/metabolism , G1 Phase Cell Cycle Checkpoints , Lipid Metabolism , Carcinoma, Renal Cell/pathology , Cell Membrane/pathology , Endoplasmic Reticulum Stress , Glutamine/metabolism , Humans , Kidney Neoplasms , MCF-7 Cells , Neoplasm Proteins/metabolism , PTEN Phosphohydrolase/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Nature ; 533(7602): 243-6, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27096364

ABSTRACT

New World monkeys (platyrrhines) are a diverse part of modern tropical ecosystems in North and South America, yet their early evolutionary history in the tropics is largely unknown. Molecular divergence estimates suggest that primates arrived in tropical Central America, the southern-most extent of the North American landmass, with several dispersals from South America starting with the emergence of the Isthmus of Panama 3-4 million years ago (Ma). The complete absence of primate fossils from Central America has, however, limited our understanding of their history in the New World. Here we present the first description of a fossil monkey recovered from the North American landmass, the oldest known crown platyrrhine, from a precisely dated 20.9-Ma layer in the Las Cascadas Formation in the Panama Canal Basin, Panama. This discovery suggests that family-level diversification of extant New World monkeys occurred in the tropics, with new divergence estimates for Cebidae between 22 and 25 Ma, and provides the oldest fossil evidence for mammalian interchange between South and North America. The timing is consistent with recent tectonic reconstructions of a relatively narrow Central American Seaway in the early Miocene epoch, coincident with over-water dispersals inferred for many other groups of animals and plants. Discovery of an early Miocene primate in Panama provides evidence for a circum-Caribbean tropical distribution of New World monkeys by this time, with ocean barriers not wholly restricting their northward movements, requiring a complex set of ecological factors to explain their absence in well-sampled similarly aged localities at higher latitudes of North America.


Subject(s)
Animal Migration , Fossils , Platyrrhini , Tropical Climate , Animals , Caribbean Region , Cebidae , Forests , History, Ancient , North America , Oceans and Seas , Panama , Phylogeny , Platyrrhini/anatomy & histology , Platyrrhini/classification
15.
Mol Cancer Ther ; 15(3): 347-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26916116

ABSTRACT

The mTOR pathway is a critical regulator of cell growth, proliferation, metabolism, and survival. Dysregulation of mTOR signaling has been observed in most cancers and, thus, the mTOR pathway has been extensively studied for therapeutic intervention. Rapamycin is a natural product that inhibits mTOR with high specificity. However, its efficacy varies by dose in several contexts. First, different doses of rapamycin are needed to suppress mTOR in different cell lines; second, different doses of rapamycin are needed to suppress the phosphorylation of different mTOR substrates; and third, there is a differential sensitivity of the two mTOR complexes mTORC1 and mTORC2 to rapamycin. Intriguingly, the enigmatic properties of rapamycin dosage can be explained in large part by the competition between rapamycin and phosphatidic acid (PA) for mTOR. Rapamycin and PA have opposite effects on mTOR whereby rapamycin destabilizes and PA stabilizes both mTOR complexes. In this review, we discuss the properties of rapamycin dosage in the context of anticancer therapeutics.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Sirolimus/administration & dosage , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism
16.
J Biol Chem ; 291(17): 9322-9, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26921316

ABSTRACT

During G1-phase of the cell cycle, normal cells respond first to growth factors that indicate that it is appropriate to divide and then later in G1 to the presence of nutrients that indicate sufficient raw material to generate two daughter cells. Dividing cells rely on the "conditionally essential" amino acid glutamine (Q) as an anaplerotic carbon source for TCA cycle intermediates and as a nitrogen source for nucleotide biosynthesis. We previously reported that while non-transformed cells arrest in the latter portion of G1 upon Q deprivation, mutant KRas-driven cancer cells bypass the G1 checkpoint, and instead, arrest in S-phase. In this study, we report that the arrest of KRas-driven cancer cells in S-phase upon Q deprivation is due to the lack of deoxynucleotides needed for DNA synthesis. The lack of deoxynucleotides causes replicative stress leading to activation of the ataxia telangiectasia and Rad3-related protein (ATR)-mediated DNA damage pathway, which arrests cells in S-phase. The key metabolite generated from Q utilization was aspartate, which is generated from a transaminase reaction whereby Q-derived glutamate is converted to α-ketoglutarate with the concomitant conversion of oxaloacetate to aspartate. Aspartate is a critical metabolite for both purine and pyrimidine nucleotide biosynthesis. This study identifies the molecular basis for the S-phase arrest caused by Q deprivation in KRas-driven cancer cells that arrest in S-phase in response to Q deprivation. Given that arresting cells in S-phase sensitizes cells to apoptotic insult, this study suggests novel therapeutic approaches to KRas-driven cancers.


Subject(s)
Aspartic Acid/metabolism , Citric Acid Cycle , Glutamic Acid/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , S Phase Cell Cycle Checkpoints , Aspartic Acid/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Glutamic Acid/genetics , Humans , MCF-7 Cells , Proto-Oncogene Proteins p21(ras)/genetics , Purine Nucleotides/biosynthesis , Purine Nucleotides/genetics , Pyrimidine Nucleotides/biosynthesis , Pyrimidine Nucleotides/genetics
18.
Cell Cycle ; 14(20): 3331-9, 2015.
Article in English | MEDLINE | ID: mdl-26323019

ABSTRACT

mTOR - the mammalian/mechanistic target of rapamycin - has been implicated as a key signaling node for promoting survival of cancer cells. However, clinical trials that have targeted mTOR with rapamycin or rapamycin analogs have had minimal impact. In spite of the high specificity of rapamycin for mTOR, the doses needed to suppress key mTOR substrates have proved toxic. We report here that rapamycin when combined with AICAR - a compound that activates AMP-activated protein kinase makes rapamycin cytotoxic rather than cytostatic at doses that are tolerated clinically. AICAR by itself is able to suppress mTOR complex 1 (mTORC1), but also stimulates a feedback activation of mTORC2, which activates the survival kinase Akt. However, AICAR also suppresses production of phosphatidic acid (PA), which interacts with mTOR in a manner that is competitive with rapamycin. The reduced level of PA sensitizes mTORC2 to rapamycin at tolerable nano-molar doses leading reduced Akt phosphorylation and apoptosis. This study reveals how the use of AICAR enhances the efficacy of rapamycin such that rapamycin at low nano-molar doses can suppress mTORC2 and induce apoptosis in human cancer cells at doses that are clinically tolerable.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Neoplasms/metabolism , Neoplasms/pathology , Ribonucleotides/administration & dosage , Sirolimus/administration & dosage , Aminoimidazole Carboxamide/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , MCF-7 Cells , Neoplasms/drug therapy , Treatment Outcome
19.
Mol Cell Oncol ; 2(4): e1006077, 2015.
Article in English | MEDLINE | ID: mdl-26380379

ABSTRACT

(-)-Oleocanthal (OC), a phenolic compound present in extra-virgin olive oil (EVOO), has been implicated in the health benefits associated with diets rich in EVOO. We investigated the effect of OC on human cancer cell lines in culture and found that OC induced cell death in all cancer cells examined as rapidly as 30 minutes after treatment in the absence of serum. OC treatment of non-transformed cells suppressed their proliferation but did not cause cell death. OC induced both primary necrotic and apoptotic cell death via induction of lysosomal membrane permeabilization (LMP). We provide evidence that OC promotes LMP by inhibiting acid sphingomyelinase (ASM) activity, which destabilizes the interaction between proteins required for lysosomal membrane stability. The data presented here indicate that cancer cells, which tend to have fragile lysosomal membranes compared to non-cancerous cells, are susceptible to cell death induced by lysosomotropic agents. Therefore, targeting lysosomal membrane stability represents a novel approach for the induction of cancer-specific cell death.

20.
Cell Cycle ; 14(14): 2285-92, 2015.
Article in English | MEDLINE | ID: mdl-25945415

ABSTRACT

Mutations in genes encoding regulators of mTOR, the mammalian target of rapamycin, commonly provide survival signals in cancer cells. Rapamycin and analogs of rapamycin have been used with limited success in clinical trials to target mTOR-dependent survival signals in a variety of human cancers. Suppression of mTOR predominantly causes G1 cell cycle arrest, which likely contributes to the ineffectiveness of rapamycin-based therapeutic strategies. While rapamycin causes the accumulation of cells in G1, its effect in other cell cycle phases remains largely unexplored. We report here that when synchronized MDA-MB-231 breast cancer cells are allowed to progress into S-phase from G1, rapamycin activates the apoptotic machinery with a concomitant increase in cell death. In Calu-1 lung cancer cells, rapamycin induced a feedback increase in Akt phosphorylation at Ser473 in S-phase that mitigated rapamycin-induced apoptosis. However, sensitivity to rapamycin in S-phase could be reestablished if Akt phosphorylation was suppressed. We recently reported that glutamine (Gln) deprivation causes K-Ras mutant cancer cells to aberrantly arrest primarily in S-phase. Consistent with observed sensitivity of S-phase cells to rapamycin, interfering with Gln utilization sensitized both MDA-MB-231 and Calu-1 K-Ras mutant cancer cells to the apoptotic effect of rapamycin. Importantly, rapamycin induced substantially higher levels of cell death upon Gln depletion than that observed in cancer cells that were allowed to progress through S-phase after being synchronized in G1. We postulate that exploiting metabolic vulnerabilities in cancer cells such as S-phase arrest observed with K-Ras-driven cancer cells deprived of Gln, could be of great therapeutic potential.


Subject(s)
Apoptosis/drug effects , Sirolimus/pharmacology , Cell Line, Tumor , Chromones/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , MCF-7 Cells , Morpholines/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , S Phase , S Phase Cell Cycle Checkpoints/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...