Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
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
2.
Am J Med Genet A ; 167A(2): 296-312, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25604658

RESUMEN

Aicardi-Goutières syndrome is an inflammatory disease occurring due to mutations in any of TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR or IFIH1. We report on 374 patients from 299 families with mutations in these seven genes. Most patients conformed to one of two fairly stereotyped clinical profiles; either exhibiting an in utero disease-onset (74 patients; 22.8% of all patients where data were available), or a post-natal presentation, usually within the first year of life (223 patients; 68.6%), characterized by a sub-acute encephalopathy and a loss of previously acquired skills. Other clinically distinct phenotypes were also observed; particularly, bilateral striatal necrosis (13 patients; 3.6%) and non-syndromic spastic paraparesis (12 patients; 3.4%). We recorded 69 deaths (19.3% of patients with follow-up data). Of 285 patients for whom data were available, 210 (73.7%) were profoundly disabled, with no useful motor, speech and intellectual function. Chilblains, glaucoma, hypothyroidism, cardiomyopathy, intracerebral vasculitis, peripheral neuropathy, bowel inflammation and systemic lupus erythematosus were seen frequently enough to be confirmed as real associations with the Aicardi-Goutieres syndrome phenotype. We observed a robust relationship between mutations in all seven genes with increased type I interferon activity in cerebrospinal fluid and serum, and the increased expression of interferon-stimulated gene transcripts in peripheral blood. We recorded a positive correlation between the level of cerebrospinal fluid interferon activity assayed within one year of disease presentation and the degree of subsequent disability. Interferon-stimulated gene transcripts remained high in most patients, indicating an ongoing disease process. On the basis of substantial morbidity and mortality, our data highlight the urgent need to define coherent treatment strategies for the phenotypes associated with mutations in the Aicardi-Goutières syndrome-related genes. Our findings also make it clear that a window of therapeutic opportunity exists relevant to the majority of affected patients and indicate that the assessment of type I interferon activity might serve as a useful biomarker in future clinical trials.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/diagnóstico , Enfermedades Autoinmunes del Sistema Nervioso/genética , ARN Helicasas DEAD-box/genética , Exodesoxirribonucleasas/genética , Proteínas de Unión al GTP Monoméricas/genética , Mutación , Malformaciones del Sistema Nervioso/diagnóstico , Malformaciones del Sistema Nervioso/genética , Fenotipo , Fosfoproteínas/genética , Ribonucleasa H/genética , Estudios de Asociación Genética , Genotipo , Humanos , Helicasa Inducida por Interferón IFIH1 , Interferones/sangre , Interferones/líquido cefalorraquídeo , Pterinas/líquido cefalorraquídeo , Proteína 1 que Contiene Dominios SAM y HD
3.
PLoS Biol ; 9(5): e1001073, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21655302

RESUMEN

Amino-terminal acetylation is probably the most common protein modification in eukaryotes with as many as 50%-80% of proteins reportedly altered in this way. Here we report a systematic analysis of the predicted N-terminal processing of cytosolic proteins versus those destined to be sorted to the secretory pathway. While cytosolic proteins were profoundly biased in favour of processing, we found an equal and opposite bias against such modification for secretory proteins. Mutations in secretory signal sequences that led to their acetylation resulted in mis-sorting to the cytosol in a manner that was dependent upon the N-terminal processing machinery. Hence N-terminal acetylation represents an early determining step in the cellular sorting of nascent polypeptides that appears to be conserved across a wide range of species.


Asunto(s)
Retículo Endoplásmico/metabolismo , Señales de Clasificación de Proteína/genética , Transporte de Proteínas , Acetilación , Biología Computacional/métodos , Citosol/metabolismo , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
4.
Hum Mutat ; 34(8): 1066-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23592335

RESUMEN

Aicardi-Goutières syndrome is an inflammatory disorder resulting from mutations in TREX1, RNASEH2A/2B/2C, SAMHD1, or ADAR1. Here, we provide molecular, biochemical, and cellular evidence for the pathogenicity of two synonymous variants in RNASEH2A. Firstly, the c.69G>A (p.Val23Val) mutation causes the formation of a splice donor site within exon 1, resulting in an out of frame deletion at the end of exon 1, leading to reduced RNase H2 protein levels. The second mutation, c.75C>T (p.Arg25Arg), also introduces a splice donor site within exon 1, and the internal deletion of 18 amino acids. The truncated protein still forms a heterotrimeric RNase H2 complex, but lacks catalytic activity. However, as a likely result of leaky splicing, a small amount of full-length active protein is apparently produced in an individual homozygous for this mutation. Recognition of the disease causing status of these variants allows for diagnostic testing in relevant families.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/genética , Mutación Puntual , Sitios de Empalme de ARN , Ribonucleasa H/genética , Enfermedades Autoinmunes del Sistema Nervioso/diagnóstico , Enfermedades Autoinmunes del Sistema Nervioso/enzimología , Femenino , Variación Genética , Humanos , Lactante , Recién Nacido , Masculino , Mutación Missense , Malformaciones del Sistema Nervioso/diagnóstico , Malformaciones del Sistema Nervioso/enzimología , Ribonucleasa H/metabolismo
5.
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
6.
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
7.
Curr Biol ; 25(4): 445-54, 2015 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-25639242

RESUMEN

BACKGROUND: Cell growth and cell-cycle progression are tightly coordinated to enable cells to adjust their size (timing of division) to the demands of proliferation in varying nutritional environments. In fission yeast, nitrogen stress results in sustained proliferation at a reduced size. RESULTS: Here, we show that cells can sense nitrogen stress to reduce target of rapamycin complex-1 (TORC1) activity. Nitrogen-stress-induced TORC1 inhibition differs from amino-acid-dependent control of TORC1 and requires the Ssp2 (AMPKα) kinase, the Tsc1/2 complex, and Rhb1 GTPase. Importantly, the ß and γ regulatory subunits of AMPK are not required to control cell division in response to nitrogen stress, providing evidence for a nitrogen-sensing mechanism that is independent of changes in intracellular ATP/AMP levels. The CaMKK homolog Ssp1 is constitutively required for phosphorylation of the AMPKα(Ssp2) T loop. However, we find that a second homolog CaMKK(Ppk34) is specifically required to stimulate AMPKα(Ssp2) activation in response to nitrogen stress. Finally, ammonia also controls mTORC1 activity in human cells; mTORC1 is activated upon the addition of ammonium to glutamine-starved Hep3B cancer cells. CONCLUSIONS: The alternative nitrogen source ammonia can simulate TORC1 activity to support growth and division under challenging nutrient settings, a situation often seen in cancer.


Asunto(s)
Complejos Multiproteicos/genética , Nitrógeno/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , División Celular , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Schizosaccharomyces/enzimología , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
8.
Lancet Neurol ; 12(12): 1159-69, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24183309

RESUMEN

BACKGROUND: Aicardi-Goutières syndrome (AGS) is an inflammatory disorder caused by mutations in any of six genes (TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR). The disease is severe and effective treatments are urgently needed. We investigated the status of interferon-related biomarkers in patients with AGS with a view to future use in diagnosis and clinical trials. METHODS: In this case-control study, samples were collected prospectively from patients with mutation-proven AGS. The expression of six interferon-stimulated genes (ISGs) was measured by quantitative PCR, and the median fold change, when compared with the median of healthy controls, was used to create an interferon score for each patient. Scores higher than the mean of controls plus two SD (>2·466) were designated as positive. Additionally, we collated historical data for interferon activity, measured with a viral cytopathic assay, in CSF and serum from mutation-positive patients with AGS. We also undertook neutralisation assays of interferon activity in serum, and looked for the presence of autoantibodies against a panel of interferon proteins. FINDINGS: 74 (90%) of 82 patients had a positive interferon score (median 12·90, IQR 6·14-20·41) compared with two (7%) of 29 controls (median 0·93, IQR 0·57-1·30). Of the eight patients with a negative interferon score, seven had mutations in RNASEH2B (seven [27%] of all 26 patients with mutations in this gene). Repeat sampling in 16 patients was consistent for the presence or absence of an interferon signature on 39 of 41 occasions. Interferon activity (tested in 147 patients) was negatively correlated with age (CSF, r=-0·604; serum, r=-0·289), and was higher in CSF than in serum in 104 of 136 paired samples. Neutralisation assays suggested that measurable antiviral activity was related to interferon α production. We did not record significantly increased concentrations of autoantibodies to interferon subtypes in patients with AGS, or an association between the presence of autoantibodies and interferon score or serum interferon activity. INTERPRETATION: AGS is consistently associated with an interferon signature, which is apparently sustained over time and can thus be used to differentiate patients with AGS from controls. If future studies show that interferon status is a reactive biomarker, the measurement of an interferon score might prove useful in the assessment of treatment efficacy in clinical trials. FUNDING: European Union's Seventh Framework Programme; European Research Council.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Exodesoxirribonucleasas/genética , Regulación de la Expresión Génica , Interferón Tipo I/fisiología , Proteínas de Unión al GTP Monoméricas/genética , Malformaciones del Sistema Nervioso/metabolismo , Fosfoproteínas/genética , Ribonucleasa H/genética , Adolescente , Adulto , Autoanticuerpos/sangre , Enfermedades Autoinmunes del Sistema Nervioso/genética , Biomarcadores , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Heterogeneidad Genética , Genotipo , Humanos , Lactante , Interferón Tipo I/sangre , Interferón Tipo I/líquido cefalorraquídeo , Interferón Tipo I/inmunología , Masculino , Mutación , Malformaciones del Sistema Nervioso/genética , Pruebas de Neutralización , Estudios Prospectivos , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN , Proteína 1 que Contiene Dominios SAM y HD , Regulación hacia Arriba , Adulto Joven
9.
Nat Genet ; 44(11): 1243-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001123

RESUMEN

Adenosine deaminases acting on RNA (ADARs) catalyze the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) and thereby potentially alter the information content and structure of cellular RNAs. Notably, although the overwhelming majority of such editing events occur in transcripts derived from Alu repeat elements, the biological function of non-coding RNA editing remains uncertain. Here, we show that mutations in ADAR1 (also known as ADAR) cause the autoimmune disorder Aicardi-Goutières syndrome (AGS). As in Adar1-null mice, the human disease state is associated with upregulation of interferon-stimulated genes, indicating a possible role for ADAR1 as a suppressor of type I interferon signaling. Considering recent insights derived from the study of other AGS-related proteins, we speculate that ADAR1 may limit the cytoplasmic accumulation of the dsRNA generated from genomic repetitive elements.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Interferón Tipo I , Malformaciones del Sistema Nervioso/genética , ARN Bicatenario/metabolismo , Elementos Alu/genética , Animales , Exoma , Expresión Génica , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Mutación , Conformación Proteica , ARN Bicatenario/genética , Proteínas de Unión al ARN , Análisis de Secuencia de ADN , Transducción de Señal , Relación Estructura-Actividad
10.
J Biol Chem ; 283(49): 33883-8, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18819915

RESUMEN

Misfolded proteins in the endoplasmic reticulum (ER) are exported to the cytosol for degradation by the proteasome in a process known as ER-associated degradation (ERAD). CPY* is a well characterized ERAD substrate whose degradation is dependent upon the Hrd1 complex. However, although the functions of some of the components of this complex are known, the nature of the protein dislocation channel remains obscure. Sec61p has been suggested as an obvious candidate because of its role as a protein-conducting channel through which polypeptides are initially translocated into the ER. However, it has not yet been possible to functionally dissect any role for Sec61p in dislocation from its essential function in translocation. By changing the translocation properties of a series of novel ERAD substrates, we are able to separate these two events and find that functional Sec61p is essential for the ERAD-L pathway.


Asunto(s)
Carboxipeptidasas/fisiología , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/metabolismo , Carboxipeptidasas/química , Membrana Celular/metabolismo , Genotipo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana , Modelos Genéticos , Mutación , Oligonucleótidos/química , Plásmidos/metabolismo , Unión Proteica , Transporte de Proteínas , Canales de Translocación SEC , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Factores de Tiempo , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA