Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Open Biol ; 13(4): 230021, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37042113

RESUMEN

Expression and activity of the AMP-activated protein kinase (AMPK) α1 catalytic subunit of the heterotrimeric kinase significantly correlates with poor outcome for colorectal cancer patients. Hence there is considerable interest in uncovering signalling vulnerabilities arising from this oncogenic elevation of AMPKα1 signalling. We have therefore attenuated mammalian target of rapamycin (mTOR) control of AMPKα1 to generate a mutant colorectal cancer in which AMPKα1 signalling is elevated because AMPKα1 serine 347 cannot be phosphorylated by mTORC1. The elevated AMPKα1 signalling in this HCT116 α1.S347A cell line confers hypersensitivity to growth inhibition by metformin. Complementary chemical approaches confirmed this relationship in both HCT116 and the genetically distinct HT29 colorectal cells, as AMPK activators imposed vulnerability to growth inhibition by metformin in both lines. Growth inhibition by metformin was abolished when AMPKα1 kinase was deleted. We conclude that elevated AMPKα1 activity modifies the signalling architecture in such a way that metformin treatment compromises cell proliferation. Not only does this mutant HCT116 AMPKα1-S347A line offer an invaluable resource for future studies, but our findings suggest that a robust biomarker for chronic AMPKα1 activation for patient stratification could herald a place for the well-tolerated drug metformin in colorectal cancer therapy.


Asunto(s)
Neoplasias Colorrectales , Metformina , Humanos , Metformina/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Fosforilación , Transducción de Señal
2.
Cancers (Basel) ; 14(18)2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36139550

RESUMEN

The long noncoding RNA NEAT1 is known to be heavily dysregulated in many cancers. A single exon gene produces two isoforms, NEAT1_1 and NEAT1_2, through alternative 3'-end processing. As the longer isoform, NEAT1_2 is an essential scaffold for nuclear paraspeckle formation. It was previously thought that the short NEAT1_1 isoform only exists to keep the NEAT1 locus active for rapid paraspeckle formation. However, a recent glycolysis-enhancing function for NEAT1_1, contributing to cancer cell proliferation and the Warburg effect, has been demonstrated. Previous studies have mainly focused on quantifying total NEAT1 and NEAT1_2 expression levels. However, in light of the NEAT1_1 role in cancer cell metabolism, the contribution from specific NEAT1 isoforms is no longer clear. Here, the roles of NEAT1_1 and NEAT1_2 in metabolism and cancer progression are discussed.

3.
Sci Rep ; 12(1): 2889, 2022 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-35190587

RESUMEN

Metformin inhibits oxidative phosphorylation and can be used to dissect metabolic pathways in colorectal cancer (CRC) cells. CRC cell proliferation is inhibited by metformin in a dose dependent manner. MicroRNAs that regulate metabolism could be identified by their ability to alter the effect of metformin on CRC cell proliferation. An unbiased high throughput functional screen of a synthetic micoRNA (miRNA) library was used to identify miRNAs that impact the metformin response in CRC cells. Experimental validation of selected hits identified miRNAs that sensitize CRC cells to metformin through modulation of proliferation, apoptosis, cell-cycle and direct metabolic disruption. Among eight metformin sensitizing miRNAs identified by functional screening, miR-676-3p had both pro-apoptotic and cell cycle arrest activity in combination with metformin, whereas other miRNAs (miR-18b-5p, miR-145-3p miR-376b-5p, and miR-718) resulted primarily in cell cycle arrest when combined with metformin. Investigation of the combined effect of miRNAs and metformin on CRC cell metabolism showed that miR-18b-5p, miR-145-3p, miR-376b-5p, miR-676-3p and miR-718 affected glycolysis only, while miR-1181 only regulated CRC respiration. MicroRNAs can sensitize CRC cells to the anti-proliferative effects of metformin. Identifying relevant miRNA targets may enable the design of innovative therapeutic strategies.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Metformina/farmacología , MicroARNs/fisiología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Glucólisis/efectos de los fármacos , Glucólisis/genética , Humanos
4.
Cell Rep ; 38(7): 110365, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35172150

RESUMEN

AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) are metabolic kinases that co-ordinate nutrient supply with cell growth. AMPK negatively regulates mTORC1, and mTORC1 reciprocally phosphorylates S345/7 in both AMPK α-isoforms. We report that genetic or torin1-induced loss of α2-S345 phosphorylation relieves suppression of AMPK signaling; however, the regulatory effect does not translate to α1-S347 in HEK293T or MEF cells. Dephosphorylation of α2-S345, but not α1-S347, transiently targets AMPK to lysosomes, a cellular site for activation by LKB1. By mass spectrometry, we find that α2-S345 is basally phosphorylated at 2.5-fold higher stoichiometry than α1-S347 in HEK293T cells and, unlike α1, phosphorylation is partially retained after prolonged mTORC1 inhibition. Loss of α2-S345 phosphorylation in endogenous AMPK fails to sustain growth of MEFs under amino acid starvation conditions. These findings uncover an α2-specific mechanism by which AMPK can be activated at lysosomes in the absence of changes in cellular energy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Lisosomas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP/metabolismo , Secuencia de Aminoácidos , Animales , Activación Enzimática , Femenino , Glucógeno Sintasa Quinasa 3/química , Glucógeno Sintasa Quinasa 3/metabolismo , Células HEK293 , Células HeLa , Humanos , Isoenzimas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Fosforilación , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo
5.
Life Sci Alliance ; 5(5)2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35121625

RESUMEN

Cells respond to changing nutrient environments by adjusting the abundance of surface nutrient transporters and receptors. This can be achieved by modulating ubiquitin-dependent endocytosis, which in part is regulated by the NEDD4 family of E3 ligases. Here we report novel regulation of Pub1, a fission yeast Schizosaccharomyces pombe member of the NEDD4-family of E3 ligases. We show that nitrogen stress inhibits Pub1 function, thereby increasing the abundance of the amino acid transporter Aat1 at the plasma membrane and enhancing sensitivity to the toxic arginine analogue canavanine. We show that TOR complex 2 (TORC2) signalling negatively regulates Pub1, thus TORC2 mutants under nutrient stress have decreased Aat1 at the plasma membrane and are resistant to canavanine. Inhibition of TORC2 signalling increases Pub1 phosphorylation, and this is dependent on Gsk3 activity. Addition of the Tor inhibitor Torin1 increases phosphorylation of Pub1 at serine 199 (S199) by 2.5-fold, and Pub1 protein levels in S199A phospho-ablated mutants are reduced. S199 is conserved in NEDD4 and is located immediately upstream of a WW domain required for protein interaction. Together, we describe how the major TORC2 nutrient-sensing signalling network regulates environmental control of Pub1 to modulate the abundance of nutrient transporters.


Asunto(s)
Ligasas de Carbono-Nitrógeno/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Glucógeno Sintasa Quinasa 3/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
6.
Nat Biotechnol ; 40(4): 576-584, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34857927

RESUMEN

Protein phosphorylation dynamically integrates environmental and cellular information to control biological processes. Identifying functional phosphorylation amongst the thousands of phosphosites regulated by a perturbation at a global scale is a major challenge. Here we introduce 'personalized phosphoproteomics', a combination of experimental and computational analyses to link signaling with biological function by utilizing human phenotypic variance. We measure individual subject phosphoproteome responses to interventions with corresponding phenotypes measured in parallel. Applying this approach to investigate how exercise potentiates insulin signaling in human skeletal muscle, we identify both known and previously unidentified phosphosites on proteins involved in glucose metabolism. This includes a cooperative relationship between mTOR and AMPK whereby the former directly phosphorylates the latter on S377, for which we find a role in metabolic regulation. These results establish personalized phosphoproteomics as a general approach for investigating the signal transduction underlying complex biology.


Asunto(s)
Fenómenos Biológicos , Fosfoproteínas , Fosfoproteínas/genética , Fosforilación , Proteómica/métodos , Transducción de Señal/fisiología
7.
Open Biol ; 11(4): 200405, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33823663

RESUMEN

Fluctuations in TOR, AMPK and MAP-kinase signalling maintain cellular homeostasis and coordinate growth and division with environmental context. We have applied quantitative, SILAC mass spectrometry to map TOR and nutrient-controlled signalling in the fission yeast Schizosaccharomyces pombe. Phosphorylation levels at more than 1000 sites were altered following nitrogen stress or Torin1 inhibition of the TORC1 and TORC2 networks that comprise TOR signalling. One hundred and thirty of these sites were regulated by both perturbations, and the majority of these (119) new targets have not previously been linked to either nutritional or TOR control in either yeasts or humans. Elimination of AMPK inhibition of TORC1, by removal of AMPKα (ssp2::ura4+), identified phosphosites where nitrogen stress-induced changes were independent of TOR control. Using a yeast strain with an ATP analogue-sensitized Cdc2 kinase, we excluded sites that were changed as an indirect consequence of mitotic control modulation by nitrogen stress or TOR signalling. Nutritional control of gene expression was reflected in multiple targets in RNA metabolism, while significant modulation of actin cytoskeletal components points to adaptations in morphogenesis and cell integrity networks. Reduced phosphorylation of the MAPKK Byr1, at a site whose human equivalent controls docking between MEK and ERK, prevented sexual differentiation when resources were sparse but not eliminated.


Asunto(s)
Fosfoproteínas/metabolismo , Proteoma , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Biomarcadores , Ciclo Celular/genética , Biología Computacional , Metabolismo Energético , Ontología de Genes , Interacciones Microbiota-Huesped , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Nitrógeno/metabolismo , Fosfoproteínas/genética , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Transducción de Señal , Estrés Fisiológico , Serina-Treonina Quinasas TOR/genética
8.
Nat Metab ; 2(1): 41-49, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31993556

RESUMEN

Central to cellular metabolism and cell proliferation are highly conserved signalling pathways controlled by mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK)1,2, dysregulation of which are implicated in pathogenesis of major human diseases such as cancer and type 2 diabetes. AMPK pathways leading to reduced cell proliferation are well established and, in part, act through inhibition of TOR complex-1 (TORC1) activity. Here we demonstrate reciprocal regulation, specifically that TORC1 directly down-regulates AMPK signalling by phosphorylating the evolutionarily conserved residue Ser367 in the fission yeast AMPK catalytic subunit Ssp2, and AMPK α1Ser347/α2Ser345 in the mammalian homologs, which is associated with reduced phosphorylation of activation loop Thr172. Genetic or pharmacological inhibition of TORC1 signalling led to AMPK activation in the absence of increased AMP:ATP ratios; under nutrient stress conditions this was associated with growth limitation in both yeast and human cell cultures. Our findings reveal fundamental, bi-directional regulation between two major metabolic signalling networks and uncover new opportunity for cancer treatment strategies aimed at suppressing cell proliferation in the nutrient-poor tumor microenvironment.


Asunto(s)
Adenilato Quinasa/antagonistas & inhibidores , Proliferación Celular/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Nutrientes/metabolismo , Estrés Fisiológico , Adenilato Quinasa/química , Adenilato Quinasa/metabolismo , Dominio Catalítico , Diabetes Mellitus Tipo 2/metabolismo , Regulación hacia Abajo , Activación Enzimática , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/efectos de los fármacos , Neoplasias/metabolismo , Fosforilación , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Transducción de Señal/fisiología
9.
Mol Metab ; 23: 98-126, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837197

RESUMEN

BACKGROUND: Cancer cells possess a common metabolic phenotype, rewiring their metabolic pathways from mitochondrial oxidative phosphorylation to aerobic glycolysis and anabolic circuits, to support the energetic and biosynthetic requirements of continuous proliferation and migration. While, over the past decade, molecular and cellular studies have clearly highlighted the association of oncogenes and tumor suppressors with cancer-associated glycolysis, more recent attention has focused on the role of microRNAs (miRNAs) in mediating this metabolic shift. Accumulating studies have connected aberrant expression of miRNAs with direct and indirect regulation of aerobic glycolysis and associated pathways. SCOPE OF REVIEW: This review discusses the underlying mechanisms of metabolic reprogramming in cancer cells and provides arguments that the earlier paradigm of cancer glycolysis needs to be updated to a broader concept, which involves interconnecting biological pathways that include miRNA-mediated regulation of metabolism. For these reasons and in light of recent knowledge, we illustrate the relationships between metabolic pathways in cancer cells. We further summarize our current understanding of the interplay between miRNAs and these metabolic pathways. This review aims to highlight important metabolism-associated molecular components in the hunt for selective preventive and therapeutic treatments. MAJOR CONCLUSIONS: Metabolism in cancer cells is influenced by driver mutations but is also regulated by posttranscriptional gene silencing. Understanding the nuanced regulation of gene expression in these cells and distinguishing rapid cellular responses from chronic adaptive mechanisms provides a basis for rational drug design and novel therapeutic strategies.


Asunto(s)
Respiración de la Célula , Glucólisis , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Humanos , Mitocondrias/metabolismo , Oncogenes/genética , Fosforilación Oxidativa , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo
10.
J Cell Sci ; 132(7)2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30814334

RESUMEN

AMP-activated kinase (AMPK) and target of rapamycin (TOR) signalling coordinate cell growth, proliferation, metabolism and cell survival with the nutrient environment of cells. The poor vasculature and nutritional stress experienced by cells in solid tumours raises the question: how do they assimilate sufficient nutrients to survive? Here, we show that human and fission yeast cells import ATP and AMP from their external environment to regulate AMPK and TOR signalling. Exposure of fission yeast (Schizosaccharomyces pombe) and human cells to external AMP impeded cell growth; however, in yeast this restraining impact required AMPK. In contrast, external ATP rescued the growth defect of yeast mutants with reduced TORC1 signalling; furthermore, exogenous ATP transiently enhanced TORC1 signalling in both yeast and human cell lines. Addition of the PANX1 channel inhibitor probenecid blocked ATP import into human cell lines suggesting that this channel may be responsible for both ATP release and uptake in mammals. In light of these findings, it is possible that the higher extracellular ATP concentration reported in solid tumours is both scavenged and recognized as an additional energy source beneficial for cell growth.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/genética , Proliferación Celular , Conexinas/metabolismo , Regulación Fúngica de la Expresión Génica , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Estrés Fisiológico
11.
Sci Rep ; 9(1): 1461, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30728400

RESUMEN

Ammonia can be utilised as an alternative nitrogen source to glutamine to support cell proliferation. However, the underlying molecular mechanisms and whether all cells have this ability is not fully understood. We find that eleven cancer and non-cancerous cell lines have opposite abilities to tolerate and utilise ammonia to support proliferation in a glutamine-depleted environment. HEK293, Huh7, T47D and MCF7 cells can use ammonia, when starved of glutamine, to support proliferation to varying degrees. Glutamine depletion reduced mTORC1 activity, while additional ammonia supplementation diminished this mTORC1 inhibition. Depletion of glutamine promoted a rapid and transient activation of AMPK, whereas, additional ammonia supplementation blocked this starvation-induced AMPK activation. As expected, drug-induced AMPK activation reduced cell proliferation in glutamine-depleted cells supplemented with ammonia. Surprisingly, mTORC1 activity was largely unchanged despite the enhanced AMPK activity, suggesting that AMPK does not inhibit mTORC1 signalling under these conditions. Finally, glutamate dehydrogenase (GDH) inhibition, a key enzyme regulating ammonia assimilation, leads to AMPK activation, mTORC1 inhibition and reduced proliferation. Ammonia provides an alternative nitrogen source that aids certain cancer cells ability to thrive in nutrient-deprived environment. The ability of cells to utilise ammonia as a nitrogen source is intricately linked to AMPK, mTORC1 and GDH.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Amoníaco/metabolismo , Glutamato Deshidrogenasa/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular , Medios de Cultivo/química , Glutamina/deficiencia , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Transducción de Señal
12.
Open Biol ; 8(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29720420

RESUMEN

Nutrient fluctuations in the cellular environment promote changes in cell metabolism and growth to adapt cell proliferation accordingly. The target of rapamycin (TOR) signalling network plays a key role in the coordination of growth and cell proliferation with the nutrient environment and, importantly, nutrient limitation reduces TOR complex 1 (TORC1) signalling. We have performed global quantitative fitness profiling of the collection of Schizosaccharomyces pombe strains from which non-essential genes have been deleted. We identified genes that regulate fitness when cells are grown in a nutrient-rich environment compared with minimal environments, with varying nitrogen sources including ammonium, glutamate and proline. In addition, we have performed the first global screen for genes that regulate fitness when both TORC1 and TORC2 signalling is reduced by Torin1. Analysis of genes whose deletions altered fitness when nutrients were limited, or when TOR signalling was compromised, identified a large number of genes that regulate transmembrane transport, transcription and chromatin organization/regulation and vesicle-mediated transport. The ability to tolerate reduced TOR signalling placed demands upon a large number of biological processes including autophagy, mRNA metabolic processing and nucleocytoplasmic transport. Importantly, novel biological processes and all processes known to be regulated by TOR were identified in our screens. In addition, deletion of 62 genes conserved in humans gave rise to strong sensitivity or resistance to Torin1, and 29 of these 62 genes have novel links to TOR signalling. The identification of chromatin and transcriptional regulation, nutritional uptake and transport pathways in this powerful genetic model now paves the way for a molecular understanding of how cells adapt to the chronic and acute fluctuations in nutrient supply that all eukaryotes experience at some stage, and which is a key feature of cancer cells within solid tumours.


Asunto(s)
Aptitud Genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Nitrógeno/metabolismo , Schizosaccharomyces/crecimiento & desarrollo , Secuencia Conservada , Eliminación de Gen , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Naftiridinas/farmacología , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Estrés Fisiológico
13.
EMBO Rep ; 18(12): 2197-2218, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29079657

RESUMEN

Gene expression regulation is essential for cells to adapt to changes in their environment. Co-activator complexes have well-established roles in transcriptional regulation, but less is known about how they sense and respond to signaling cues. We have previously shown that, in fission yeast, one such co-activator, the SAGA complex, controls gene expression and the switch from proliferation to differentiation in response to nutrient availability. Here, using a combination of genetic, biochemical, and proteomic approaches, we show that SAGA responds to nutrients through the differential phosphorylation of its Taf12 component, downstream of both the TORC1 and TORC2 pathways. Taf12 phosphorylation increases early upon starvation and is controlled by the opposing activities of the PP2A phosphatase, which is activated by TORC1, and the TORC2-activated Gad8AKT kinase. Mutational analyses suggest that Taf12 phosphorylation prevents cells from committing to differentiation until starvation reaches a critical level. Overall, our work reveals that SAGA is a direct target of nutrient-sensing pathways and has uncovered a mechanism by which TORC1 and TORC2 converge to control gene expression and cell fate decisions.


Asunto(s)
Regulación Fúngica de la Expresión Génica , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Citoplasma/metabolismo , Mutación , Fosforilación/genética , Proteómica/métodos , Proteínas de Schizosaccharomyces pombe/metabolismo , Transducción de Señal/genética , Transactivadores/genética , Transcripción Genética
14.
Sci Rep ; 7(1): 12101, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28935859

RESUMEN

The glucocorticoid receptor (GR) is essential for the stress response in mammals. We investigated potential non-transcriptional roles of GR in cellular stress response using fission yeast as a model.We surprisingly discovered marked heat stress resistance in yeast ectopically expressing human GR, which required expression of both the N-terminal transactivation domain, and the C-terminal ligand binding domain, but not the DNA-binding domain of the GR. This effect was not affected by GR ligand exposure, and occurred without significant GR nuclear accumulation. Mechanistically, the GR survival effect required Hsp104, and, indeed, GR expression increased Hsp104 expression. Proteomic analysis revealed GR binding to translasome components, including eIF3, a known partner for Sty1, a pattern of protein interaction which we confirmed using yeast two-hybrid studies.Taken together, we find evidence for a novel pathway conferring stress resistance in yeast that can be activated by the human GR, acting by protein-protein mechanisms in the cytoplasm. This suggests that in organisms where GR is natively expressed, GR likely contributes to stress responses through non-transcriptional mechanisms in addition to its well-established transcriptional responses.


Asunto(s)
Receptores de Glucocorticoides/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Humanos , Unión Proteica , Dominios Proteicos , Mapas de Interacción de Proteínas , Proteómica/métodos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Schizosaccharomyces/citología , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Estrés Fisiológico , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos
15.
PLoS One ; 12(3): e0172740, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28273166

RESUMEN

Tight coupling of cell growth and cell cycle progression enable cells to adjust their rate of division, and therefore size, to the demands of proliferation in varying nutritional environments. Nutrient stress promotes inhibition of Target Of Rapamycin Complex 1 (TORC1) activity. In fission yeast, reduced TORC1 activity advances mitotic onset and switches growth to a sustained proliferation at reduced cell size. A screen for mutants, that failed to advance mitosis upon nitrogen stress, identified a mutant in the PIKFYVE 1-phosphatidylinositol-3-phosphate 5-kinase fission yeast homolog Ste12. Ste12PIKFYVE deficient mutants were unable to advance the cell cycle to reduce cell size after a nitrogen downshift to poor nitrogen (proline) growth conditions. While it is well established that PI(3,5)P2 signalling is required for autophagy and that Ste12PIKFYVE mutants have enlarged vacuoles (yeast lysosomes), neither a block to autophagy or mutants that independently have enlarged vacuoles had any impact upon nitrogen control of mitotic commitment. The addition of rapamycin to Ste12PIKFYVE deficient mutants reduced cell size at division to suggest that Ste12PIKFYVE possibly functions upstream of TORC1. ste12 mutants display increased Torin1 (TOR inhibitor) sensitivity. However, no major impact on TORC1 or TORC2 activity was observed in the ste12 deficient mutants. In summary, Ste12PIKFYVE is required for nitrogen-stress mediated advancement of mitosis to reduce cell size at division.


Asunto(s)
Mitosis , Nitrógeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Schizosaccharomyces/fisiología , Autofagia , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Unión Proteica , Transporte de Proteínas , Schizosaccharomyces/citología , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Transducción de Señal , Estrés Fisiológico/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Vacuolas/metabolismo
17.
PLoS Genet ; 12(5): e1006041, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27191590

RESUMEN

Target of Rapamycin (TOR) signalling allows eukaryotic cells to adjust cell growth in response to changes in their nutritional and environmental context. The two distinct TOR complexes (TORC1/2) localise to the cell's internal membrane compartments; the endoplasmic reticulum (ER), Golgi apparatus and lysosomes/vacuoles. Here, we show that Ppk32, a SCYL family pseudo-kinase, is a novel regulator of TOR signalling. The absence of ppk32 expression confers resistance to TOR inhibition. Ppk32 inhibition of TORC1 is critical for cell survival following Brefeldin A (BFA) induced stress. Treatment of wild type cells with either the TORC1 specific inhibitor rapamycin or the general TOR inhibitor Torin1 confirmed that a reduction in TORC1 activity promoted recovery from BFA induced stress. Phosphorylation of Ppk32 on two residues that are conserved within the SCYL pseudo-kinase family are required for this TOR inhibition. Phosphorylation on these sites controls Ppk32 protein levels and sensitivity to BFA. BFA induced ER stress does not account for the response to BFA that we report here, however BFA is also known to induce Golgi stress and impair traffic to lysosomes. In summary, Ppk32 reduce TOR signalling in response to BFA induced stress to support cell survival.


Asunto(s)
Complejos Multiproteicos/genética , Transporte de Proteínas/genética , Estrés Fisiológico/genética , Serina-Treonina Quinasas TOR/genética , Brefeldino A/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/genética , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/genética , Lisosomas/efectos de los fármacos , Lisosomas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Schizosaccharomyces/genética , Schizosaccharomyces/crecimiento & desarrollo , Sirolimus/metabolismo , Estrés Fisiológico/efectos de los fármacos , Vacuolas/efectos de los fármacos , Vacuolas/genética
18.
J Cell Sci ; 129(13): 2613-24, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27206859

RESUMEN

The timing of cell division is controlled by the coupled regulation of growth and division. The target of rapamycin (TOR) signalling network synchronises these processes with the environmental setting. Here, we describe a novel interaction of the fission yeast TOR complex 2 (TORC2) with the cytokinetic actomyosin ring (CAR), and a novel role for TORC2 in regulating the timing and fidelity of cytokinesis. Disruption of TORC2 or its localisation results in defects in CAR morphology and constriction. We provide evidence that the myosin II protein Myp2 and the myosin V protein Myo51 play roles in recruiting TORC2 to the CAR. We show that Myp2 and TORC2 are co-dependent upon each other for their normal localisation to the cytokinetic machinery. We go on to show that TORC2-dependent phosphorylation of actin-capping protein 1 (Acp1, a known regulator of cytokinesis) controls CAR stability, modulates Acp1-Acp2 (the equivalent of the mammalian CAPZA-CAPZB) heterodimer formation and is essential for survival upon stress. Thus, TORC2 localisation to the CAR, and TORC2-dependent Acp1 phosphorylation contributes to timely control and the fidelity of cytokinesis and cell division.


Asunto(s)
Proteínas de Capping de la Actina/genética , Citocinesis/genética , Complejos Multiproteicos/genética , Cadenas Pesadas de Miosina/genética , Miosinas/genética , Proteínas de Schizosaccharomyces pombe/genética , Serina-Treonina Quinasas TOR/genética , Proteínas de Capping de la Actina/metabolismo , Actinas/genética , Actomiosina/genética , Actomiosina/metabolismo , División Celular/genética , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosinas/metabolismo , Fosforilación , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Cold Spring Harb Protoc ; 2016(4): pdb.err093682, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27037066
20.
Open Biol ; 6(3)2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26935949

RESUMEN

Cell proliferation, metabolism, migration and survival are coordinated through the tight control of two target of rapamycin (TOR) kinase complexes: TORC1 and TORC2. Here, we show that a novel phosphorylation of fission yeast Gad8 (AGC kinase) on the evolutionarily conserved threonine 6 (Thr6) prevents the physical association between Gad8 and TORC2. Accordingly, this block to protein interactions by Gad8 Thr6 phosphorylation decreases TORC2-controlled activation of Gad8. Likewise, phosphorylation of Gad8 Thr6, possibly by PKC, prevents the association of Gad8 with TORC2 thereby increasing TORC2 activity, because it reduces Gad8-mediated feedback inhibition of TORC2. Consistently, the introduction of a Gad8 T6D mutant, that mimics phosphorylation, increased TORC2 activity. Increased PKC(Pck2) expression prevented Gad8-TORC2 binding and so reduced the TORC2-mediated phosphorylation of Gad8 serine 546 that activates Gad8. Interestingly, independent of the Ser546 phosphorylation status, Gad8 Thr6 phosphorylation is important for remodelling the actin cytoskeleton and survival upon potassium ion and heat stresses. In contrast, Ser546 phosphorylation is required for the control of G1 arrest, mating, cell length at division and vascular size. Finally, these findings reveal a novel mode of TORC2 activation that is essential for cell survival following stress.


Asunto(s)
Complejos Multiproteicos/metabolismo , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Actinas/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Fosforilación , Potasio/metabolismo , Unión Proteica , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/química , Schizosaccharomyces/química , Schizosaccharomyces/citología , Proteínas de Schizosaccharomyces pombe/química , Estrés Fisiológico , Treonina/química , Treonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...