Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 533(7602): 243-6, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-27096364

RESUMO

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.


Assuntos
Migração Animal , Fósseis , Platirrinos , Clima Tropical , Animais , Região do Caribe , Cebidae , Florestas , História Antiga , América do Norte , Oceanos e Mares , Panamá , Filogenia , Platirrinos/anatomia & histologia , Platirrinos/classificação
2.
J Biol Chem ; 295(1): 263-274, 2020 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-31767684

RESUMO

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.


Assuntos
Aminoácidos/deficiência , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Ácidos Fosfatídicos/metabolismo , Aminoácidos/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Endocitose , Humanos , Camundongos , Fosfolipase D/metabolismo , Transporte Proteico , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
3.
J Biol Chem ; 293(42): 16390-16401, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30194281

RESUMO

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.


Assuntos
Glutamina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fosfolipase D/metabolismo , Linhagem Celular , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/química , Nutrientes/metabolismo , Ácidos Fosfatídicos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Proteínas ral de Ligação ao GTP/metabolismo
4.
J Biol Chem ; 292(3): 936-944, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-27956548

RESUMO

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.


Assuntos
Carcinoma de Células Renais/metabolismo , Membrana Celular/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Metabolismo dos Lipídeos , Carcinoma de Células Renais/patologia , Membrana Celular/patologia , Estresse do Retículo Endoplasmático , Glutamina/metabolismo , Humanos , Neoplasias Renais , Células MCF-7 , Proteínas de Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Serina-Treonina Quinases TOR/metabolismo
5.
J Biol Chem ; 292(15): 6303-6311, 2017 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-28223357

RESUMO

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.


Assuntos
Complexos Multiproteicos/metabolismo , Ácidos Fosfatídicos/biossíntese , Serina-Treonina Quinases TOR/metabolismo , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Células MCF-7 , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos/genética , Ácido Oleico/farmacologia , Ácidos Fosfatídicos/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Serina-Treonina Quinases TOR/genética
6.
Am J Bot ; 105(11): 1929-1937, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30418663

RESUMO

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.


Assuntos
Atmosfera/química , Dióxido de Carbono , Clima , Fósseis , Estômatos de Plantas/fisiologia , Modelos Biológicos
7.
J Biol Chem ; 291(17): 9322-9, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26921316

RESUMO

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.


Assuntos
Ácido Aspártico/metabolismo , Ciclo do Ácido Cítrico , Ácido Glutâmico/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular , Ácido Aspártico/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Ácido Glutâmico/genética , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas p21(ras)/genética , Nucleotídeos de Purina/biossíntese , Nucleotídeos de Purina/genética , Nucleotídeos de Pirimidina/biossíntese , Nucleotídeos de Pirimidina/genética
8.
J Biol Chem ; 290(11): 6986-93, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25632961

RESUMO

AMP-activated protein kinase (AMPK), a critical sensor of energy sufficiency, acts as central metabolic switch in cell metabolism. Once activated by low energy status, AMPK phosphorylates key regulatory substrates and turns off anabolic biosynthetic pathways. In contrast, the mammalian/mechanistic target of rapamycin (mTOR) is active when there are sufficient nutrients for anabolic reactions. A critical factor regulating mTOR is phosphatidic acid (PA), a central metabolite of membrane lipid biosynthesis and the product of the phospholipase D (PLD)-catalyzed hydrolysis of phosphatidylcholine. PLD is a downstream target of the GTPase Rheb, which is turned off in response to AMPK via the tuberous sclerosis complex. Although many studies have linked AMPK with mTOR, very little is known about the connection between AMPK and PLD. In this report, we provide evidence for reciprocal regulation of PLD by AMPK and regulation of AMPK by PLD and PA. Suppression of AMPK activity led to an increase in PLD activity, and conversely, activation of AMPK suppressed PLD activity. Suppression of PLD activity resulted in elevated AMPK activity. Exogenously supplied PA abolished the inhibitory effects of elevated AMPK activity on mTOR signaling. In contrast, exogenously supplied PA could not overcome the effect AMPK activation if either mTOR or Raptor was suppressed, indicating that the inhibitory effects of PLD and PA on AMPK activity are mediated by mTOR. These data suggest a reciprocal feedback mechanism involving AMPK and the PLD/mTOR signaling node in cancer cells with therapeutic implications.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Neoplasias/enzimologia , Fosfolipase D/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Neoplasias/metabolismo , Ácidos Fosfatídicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo
9.
J Biol Chem ; 289(33): 22583-22588, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24990952

RESUMO

Phosphatidic acid (PA) is a critical metabolite at the heart of membrane phospholipid biosynthesis. However, PA also serves as a critical lipid second messenger that regulates several proteins implicated in the control of cell cycle progression and cell growth. Three major metabolic pathways generate PA: phospholipase D (PLD), diacylglycerol kinase (DGK), and lysophosphatidic acid acyltransferase (LPAAT). The LPAAT pathway is integral to de novo membrane phospholipid biosynthesis, whereas the PLD and DGK pathways are activated in response to growth factors and stress. The PLD pathway is also responsive to nutrients. A key target for the lipid second messenger function of PA is mTOR, the mammalian/mechanistic target of rapamycin, which integrates both nutrient and growth factor signals to control cell growth and proliferation. Although PLD has been widely implicated in the generation of PA needed for mTOR activation, it is becoming clear that PA generated via the LPAAT and DGK pathways is also involved in the regulation of mTOR. In this minireview, we highlight the coordinated maintenance of intracellular PA levels that regulate mTOR signals stimulated by growth factors and nutrients, including amino acids, lipids, glucose, and Gln. Emerging evidence indicates compensatory increases in one source of PA when another source is compromised, highlighting the importance of being able to adapt to stressful conditions that interfere with PA production. The regulation of PA levels has important implications for cancer cells that depend on PA and mTOR activity for survival.


Assuntos
Ácidos Fosfatídicos/metabolismo , Fosfolipase D/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Serina-Treonina Quinases TOR/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Animais , Diacilglicerol Quinase/genética , Diacilglicerol Quinase/metabolismo , Glucose/genética , Glucose/metabolismo , Glutamina/genética , Glutamina/metabolismo , Humanos , Ácidos Fosfatídicos/genética , Fosfolipase D/genética , Serina-Treonina Quinases TOR/genética
10.
Nat Cell Biol ; 9(6): 713-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17486117

RESUMO

Ras activation as a consequence of antigen receptor (T-cell receptor; TCR) engagement on T lymphocytes is required for T-cell development, selection and function. Lymphocyte function-associated antigen-1 (LFA-1) mediates lymphocyte adhesion, stabilization of the immune synapse and bidirectional signalling. Using a fluorescent biosensor we found that TCR activation with or without costimulation of CD28 led to activation of Ras only on the Golgi apparatus, whereas costimulation with LFA-1 induced Ras activation on both the Golgi and the plasma membrane. Ras activation on both compartments required RasGRP1, an exchange factor regulated by calcium and diacylglycerol (DAG), but phospholipase C (PLC) activity was required only for activation on the Golgi. Engagement of LFA-1 increased DAG levels at the plasma membrane by stimulating phospholipase D (PLD). PLD2 and phosphatidic acid phosphatase (PAP) were required for Ras activation on the plasma membrane. Thus, LFA-1 acts through PLD2 to reshape the pattern of Ras activation downstream of the TCR.


Assuntos
Membrana Celular/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Fosfolipase D/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Proteínas ras/metabolismo , Animais , Antígenos CD28/metabolismo , Comunicação Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Diglicerídeos/metabolismo , Ativação Enzimática/fisiologia , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia , Fosfolipases Tipo C/metabolismo
12.
Trends Endocrinol Metab ; 34(3): 170-180, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36732094

RESUMO

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.


Assuntos
Ácidos Fosfatídicos , Serina-Treonina Quinases TOR , Humanos , Ácidos Fosfatídicos/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Aminoácidos/metabolismo
13.
J Biol Chem ; 286(29): 25477-86, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21622984

RESUMO

The mammalian target of rapamycin (mTOR) is a critical sensor of nutritional sufficiency. Although much is known about the regulation of mTOR in response to growth factors, much less is known about the regulation of mTOR in response to nutrients. Amino acids have no impact on the signals that regulate Rheb, a GTPase required for the activation of mTOR complex 1 (mTORC1). Phospholipase D (PLD) generates a metabolite, phosphatidic acid, that facilitates association between mTOR and the mTORC1 co-factor Raptor. We report here that elevated PLD activity in human cancer cells is dependent on both amino acids and glucose and that amino acid- and glucose-induced increases in mTORC1 activity are dependent on PLD. Amino acid- and glucose-induced PLD and mTORC1 activity were also dependent on the GTPases RalA and ARF6 and the type III phosphatidylinositol-3-kinase hVps34. Thus, a key stimulatory event for mTORC1 activation in response to nutrients is the generation of phosphatidic acid by PLD.


Assuntos
Alimentos , Fosfolipase D/metabolismo , Proteínas/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Aminoácidos/farmacologia , Linhagem Celular Tumoral , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Glucose/farmacologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos , Neuropeptídeos/metabolismo , Ácidos Fosfatídicos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Proteínas ral de Ligação ao GTP/metabolismo
14.
PLoS One ; 17(10): e0276579, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36269753

RESUMO

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.


Assuntos
ATP Citrato (pro-S)-Liase , Glutamina , Neoplasias , Humanos , Adenosina Difosfato Ribose , Ácido Amino-Oxiacético , ATP Citrato (pro-S)-Liase/genética , ATP Citrato (pro-S)-Liase/metabolismo , Glutamatos/genética , Glutamina/antagonistas & inibidores , Glutamina/metabolismo , Ácidos Cetoglutáricos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 2 de Rapamicina/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Ácidos Oleicos , Oxaloacetatos , Ácidos Fosfatídicos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transaminases/genética
16.
Cancer Lett ; 522: 164-170, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34563639

RESUMO

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.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias da Próstata/tratamento farmacológico , Proteína do Retinoblastoma/genética , Fator de Crescimento Transformador beta/genética , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fatores de Transcrição E2F/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Mutação/genética , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/efeitos adversos , Sirolimo/farmacologia
17.
Biochim Biophys Acta ; 1791(9): 949-55, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19264150

RESUMO

During the past decade elevated phospholipase D (PLD) activity has been reported in virtually all cancers where it has been examined. PLD catalyzes the hydrolysis of phosphatidylcholine to generate the lipid second messenger phosphatidic acid (PA). While many targets of PA signaling have been identified, the most critical target of PA in cancer cells is likely to be mTOR - the mammalian target of rapamycin. mTOR has been widely implicated in signals that suppress apoptotic programs in cancer cells - frequently referred to as survival signals. mTOR exists as two multi-component complexes known as mTORC1 and mTORC2. Recent data has revealed that PA is required for the stability of both mTORC1 and mTORC2 complexes - and therefore also required for the kinase activity of both mTORC1 and mTORC2. PA interacts with mTOR in a manner that is competitive with rapamycin, and as a consequence, elevated PLD activity confers rapamycin resistance - a point that has been largely overlooked in clinical trials involving rapamycin-based strategies. The earliest genetic changes occurring in an emerging tumor are generally ones that suppress default apoptotic programs that likely represent the first line of defense of cancer. Targeting survival signals in human cancers represents a rational anti-cancer therapeutic strategy. Therefore, understanding the signals that regulate PA levels and how PA impacts upon mTOR could be important for developing strategies to de-repress the survival signals that suppress apoptosis. This review summarizes the role of PA in regulating the mTOR-mediated signals that promote cancer cell survival.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Ácidos Fosfatídicos/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Sobrevivência Celular , Humanos , Neoplasias/enzimologia , Fosfolipase D/metabolismo , Serina-Treonina Quinases TOR
18.
Biochem Biophys Res Commun ; 396(2): 562-5, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20438709

RESUMO

Phosphatidic acid (PA), the primary metabolite of the phospholipase D (PLD)-mediated hydrolysis of phosphatidylcholine, has been shown to act as a tumor promoting second messenger in many cancer cell lines. A key target of PA is the mammalian target of rapamycin (mTOR), a serine-threonine kinase that has been widely implicated in cancer cell survival signals. In agreement with its ability to relay survival signals, it has been reported that both PLD and mTOR are required for the stabilization of the p53 E3 ubiquitin ligase human double minute 2 (HDM2) protein. Thus, by stabilizing HDM2, PLD and mTOR are able to counter the pro-apoptotic signaling mediated by p53 and promote survival. mTOR exists in at least two distinct complexes-mTORC1 and mTORC2-that are both dependent on PLD-generated PA. Although PLD and its metabolite PA are clearly implicated in the transduction of survival signals to mTOR, it is not yet apparent which of the two mTOR complexes is critical for the stabilization of HDM2. We report here that the PLD/mTOR-dependent stabilization of HDM2 involves mTORC2 and the AGC family kinase serum- and glucocorticoid-inducible kinase 1 (SGK1). This study reveals that mTORC2 is a critical target of PLD-mediated survival signals and identifies SGK1 as a downstream target of mTORC2 for the stabilization of HDM2.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Fosfolipase D/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Estabilidade Enzimática , Humanos , Neoplasias Renais/enzimologia , Ácidos Fosfatídicos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/genética
20.
Cell Cycle ; 19(17): 2158-2167, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32715871

RESUMO

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.


Assuntos
Fibroblastos/citologia , Fase G1 , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Fase S , Soroalbumina Bovina/farmacologia , Aminoácidos/farmacologia , Animais , Bovinos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Humanos , Insulina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Pinocitose/efeitos dos fármacos , Fase S/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA