Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Exp Physiol ; 105(12): 2178-2189, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32965751

RESUMEN

NEW FINDINGS: What is the central question of the study? Is Vps34 a nutrient-sensitive activator of mTORC1 in human skeletal muscle? What is the main finding and its importance? We show that altering nutrient availability, via protein-carbohydrate feeding, does not increase Vps34 kinase activity in human skeletal muscle. Instead, feeding increased Vps34-mTORC1 co-localization in parallel to increased mTORC1 activity. These findings may have important implications in the understanding nutrient-induced mTORC1 activation in skeletal muscle via interaction with Vps34. ABSTRACT: The Class III PI3Kinase, Vps34, has recently been proposed as a nutrient sensor, essential for activation of the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1). We therefore investigated the effects of increasing nutrient availability through protein-carbohydrate (PRO-CHO) feeding on Vps34 kinase activity and cellular localization in human skeletal muscle. Eight young, healthy males (21 ± 0.5 yrs, 77.7 ± 9.9 kg, 25.9 ± 2.7 kg/m2 , mean ± SD) ingested a PRO-CHO beverage containing 20/44/1 g PRO/CHO/FAT respectively, with skeletal muscle biopsies obtained at baseline and 1 h and 3 h post-feeding. PRO-CHO feeding did not alter Vps34 kinase activity, but did stimulate Vps34 translocation toward the cell periphery (PRE (mean ± SD) - 0.273 ± 0.040, 1 h - 0.348 ± 0.061, Pearson's Coefficient (r)) where it co-localized with mTOR (PRE - 0.312 ± 0.040, 1 h - 0.348 ± 0.069, Pearson's Coefficient (r)). These alterations occurred in parallel to an increase in S6K1 kinase activity (941 ± 466% of PRE at 1 h post-feeding). Subsequent in vitro experiments in C2C12 and human primary myotubes displayed no effect of the Vps34-specific inhibitor SAR405 on mTORC1 signalling responses to elevated nutrient availability. Therefore, in summary, PRO-CHO ingestion does not increase Vps34 activity in human skeletal muscle, whilst pharmacological inhibition of Vps34 does not prevent nutrient stimulation of mTORC1 in vitro. However, PRO-CHO ingestion promotes Vps34 translocation to the cell periphery, enabling Vps34 to associate with mTOR. Therefore, our data suggests that interaction between Vps34 and mTOR, rather than changes in Vps34 activity per se may be involved in PRO-CHO activation of mTORC1 in human skeletal muscle.


Asunto(s)
Carbohidratos/administración & dosificación , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Ingestión de Alimentos/fisiología , Músculo Esquelético/metabolismo , Adulto , Animales , Línea Celular , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fibras Musculares Esqueléticas/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Adulto Joven
2.
Nutr Cancer ; 68(5): 818-26, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27176674

RESUMEN

The potential anticancer properties of garlic (Allium sativum) may depend on the method of preparation and its storage. Storage of garlic has not been thoroughly investigated to determine whether anticancer properties are retained. Garlic was prepared and processed to mimic normal options for storage and preparation for consumption. Cytotoxicity was determined by crystal violet assay and mechanisms of cytotoxicity were established by microscopy, SDS-PAGE, and Western immunoblotting. Significant (P < 0.0001) cytotoxicity was observed in all preparations, except with boiled (cooked) garlic. Depending on the method of storage, garlic extract induced either type I or type II programmed cell death, detectable by caspase 9 cleavage, or Poly (adenosine diphosphate-ribose) polymerase (PARP) cleavage and LC3-II accumulation, respectively. The conflicting literature on the anticancer properties of garlic may be explained by differences in processing and storage. This study has highlighted that the potency of the antiproliferative properties of cooked garlic, compared to the uncooked form, is diminished in HeLa cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Ajo/química , Extractos Vegetales/química , Extractos Vegetales/farmacología , Caspasa 9/genética , Caspasa 9/metabolismo , Proliferación Celular/efectos de los fármacos , Culinaria , Almacenamiento de Alimentos , Células HeLa , Calor , Humanos
3.
Bioorg Med Chem Lett ; 25(19): 4287-92, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26318998

RESUMEN

Mutations in the Ras-pathway occur in 40-45% of colorectal cancer patients and these are refractory to treatment with anti-EGFR-targeted therapies. With this in mind, we have studied novel guanidinium-based compounds with demonstrated ability to inhibit protein kinases. We have performed docking studies with several proteins involved in the Ras-pathway and evaluated 3,4'-bis-guanidinium derivatives as inhibitors of B-Raf. Compound 3, the most potent in this series, demonstrated strong cytotoxicity in (WT)B-Raf colorectal cancer cells and also cells with (V600E)B-Raf mutations. Cell death was induced by apoptosis, detected by cleavage of PARP. Compound 3 also potently inhibited ERK1/2 signalling, inhibited EGFR activation, as well as Src, STAT3 and AKT phosphorylation. Mechanistically, compound 3 did not inhibit ATP binding to B-Raf, but direct assay of B-Raf activity was inhibited in vitro. Summarizing, we have identified a novel B-Raf type-III inhibitor that exhibits potent cellular cytotoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Guanidina/farmacología , Proteínas ras/metabolismo , Regulación Alostérica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Guanidina/síntesis química , Guanidina/química , Humanos , Estructura Molecular , Relación Estructura-Actividad
4.
Biochem J ; 449(2): 389-400, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23078367

RESUMEN

Hypoxia in the microenvironment of many solid tumours is an important determinant of malignant progression. The ISR (integrated stress response) protects cells from the ER (endoplasmic reticulum) stress caused by severe hypoxia. Likewise, autophagy is a mechanism by which cancer cells can evade hypoxic cell death. In the present paper we report that the autophagy-initiating kinase ULK1 (UNC51-like kinase 1) is a direct transcriptional target of ATF4 (activating transcription factor 4), which drives the expression of ULK1 mRNA and protein in severe hypoxia and ER stress. We demonstrate that ULK1 is required for autophagy in severe hypoxia and that ablation of ULK1 causes caspase-3/7-independent cell death. Furthermore, we report that ULK1 expression is associated with a poor prognosis in breast cancer. Collectively, the findings of the present study identify transcriptional up-regulation of ULK1 as a novel arm of the ISR, and suggest ULK1 as a potentially effective target for cancer therapy.


Asunto(s)
Factor de Transcripción Activador 4/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/genética , Activación Transcripcional , Regulación hacia Arriba , Factor de Transcripción Activador 4/metabolismo , Animales , Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/genética , Estrés del Retículo Endoplásmico/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células MCF-7 , Ratones , Análisis Multivariante , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Pronóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia
5.
Adv Exp Med Biol ; 773: 323-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24563355

RESUMEN

Ran is a small ras-related GTPase that controls the nucleocytoplasmic exchange of macromolecules across the nuclear envelope. It binds to chromatin early during nuclear formation and has important roles during the eukaryotic cell cycle, where it regulates mitotic spindle assembly, nuclear envelope formation and cell cycle checkpoint control. Like other GTPases, Ran relies on the cycling between GTP-bound and GDP-bound conformations to interact with effector proteins and regulate these processes. In nucleocytoplasmic transport, Ran shuttles across the nuclear envelope through nuclear pores. It is concentrated in the nucleus by an active import mechanism where it generates a high concentration of RanGTP by nucleotide exchange. It controls the assembly and disassembly of a range of complexes that are formed between Ran-binding proteins and cellular cargo to maintain rapid nuclear transport. Ran also has been identified as an essential protein in nuclear envelope formation in eukaryotes. This mechanism is dependent on importin-ß, which regulates the assembly of further complexes important in this process, such as Nup107-Nup160. A strong body of evidence is emerging implicating Ran as a key protein in the metastatic progression of cancer. Ran is overexpressed in a range of tumors, such as breast and renal, and these perturbed levels are associated with local invasion, metastasis and reduced patient survival. Furthermore, tumors with oncogenic KRAS or PIK3CA mutations are addicted to Ran expression, which yields exciting future therapeutic opportunities.


Asunto(s)
Metástasis de la Neoplasia , Neoplasias/patología , Membrana Nuclear/fisiología , Proteína de Unión al GTP ran/metabolismo , Transporte Activo de Núcleo Celular , Ciclo Celular , Humanos , Conformación Proteica , Huso Acromático , Proteína de Unión al GTP ran/química , Proteína de Unión al GTP ran/fisiología
6.
Biochem Biophys Res Commun ; 411(2): 227-34, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21708134

RESUMEN

The tumour metastasis suppressor, N-myc Downstream Regulated Gene (NDRG) 1, is a by the protein kinases SGK1 and GSK3ß, but the relevance of its phosphorylation remains unclear. Analysis of HCT116 cells, either proficient or deficient for p53 revealed NDRG1 protein expression and phosphorylation by SGK1 was increased basally in p53-deficient cells. Treatment with the cell cycle inhibitors, aphidicolin or nocodazole also revealed increased NDRG1 phosphorylation in p53-deficient cells. Finally, phosphorylated NDRG1 was found to co-localise with γ-tubulin on centromeres and also to the cleavage furrow during cytokinesis. Taken together, this work demonstrates that NDRG1 phosphorylation, by the protein kinase SGK1, is temporally and spatially controlled during the cell cycle, suggesting a role for NDRG1 in successful mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centrómero/metabolismo , Regulación hacia Abajo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mitosis , Fosforilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
J Physiol ; 587(1): 253-60, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19015198

RESUMEN

Following resistance exercise in the fasted state, both protein synthesis and degradation in skeletal muscle are increased. The addition of essential amino acids potentiates the synthetic response suggesting that an amino acid sensor, which is involved in both synthesis and degradation, may be activated by resistance exercise. One such candidate protein is the class 3 phosphatidylinositol 3OH-kinase (PI3K) Vps34. To determine whether mammalian Vps34 (mVps34) is modulated by high-resistance contractions, mVps34 and S6K1 (an index of mTORC1) activity were measured in the distal hindlimb muscles of rats 0.5, 3, 6 and 18 h after acute unilateral high-resistance contractions with the contralateral muscles serving as a control. In the lengthening tibialis anterior (TA) muscle, S6K1 (0.5 h = 366.3 +/- 112.08%, 3 h = 124.7 +/- 15.96% and 6 h = 129.2 +/- 0%) and mVps34 (3 h = 68.8 +/- 15.1% and 6 h = 36.0 +/- 8.79%) activity both increased, whereas in the shortening soleus and plantaris (PLN) muscles the increase was significantly lower (PLN S6K1 0.5 h = 33.1 +/- 2.29% and 3 h = 47.0 +/- 6.65%; mVps34 3 h = 24.5 +/- 7.92%). HPLC analysis of the TA demonstrated a 25% increase in intramuscular leucine concentration in rats 1.5 h after exercise. A similar level of leucine added to C2C12 cells in vitro increased mVps34 activity 3.2-fold. These data suggest that, following high-resistance contractions, mVps34 activity is stimulated by an influx of essential amino acids such as leucine and this may prolong mTORC1 signalling and contribute to muscle hypertrophy.


Asunto(s)
Contracción Muscular/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Línea Celular , Activación Enzimática , Femenino , Hipertrofia , Leucina/metabolismo , Ratones , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Esfuerzo Físico/fisiología , Ratas , Ratas Wistar , Entrenamiento de Fuerza , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal , Estrés Mecánico , Factores de Transcripción/metabolismo
8.
Sci Rep ; 9(1): 12891, 2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31501494

RESUMEN

Room temperature Cold Atmospheric Plasma (CAP) has shown promising efficacy for the treatment of cancer but the exact mechanisms of action remain unclear. Both apoptosis and necrosis have been implicated as the mode of cell death in various cancer cells. We have previously demonstrated a caspase-independent mechanism of cell death in p53-mutated glioblastoma multiforme (GBM) cells exposed to plasma. The purpose of this study was to elucidate the molecular mechanisms involved in caspase-independent cell death induced by plasma treatment. We demonstrate that plasma induces rapid cell death in GBM cells, independent of caspases. Accumulation of vesicles was observed in plasma treated cells that stained positive with acridine orange. Western immunoblotting confirmed that autophagy is not activated following plasma treatment. Acridine orange intensity correlates closely with the lysosomal marker Lyso TrackerTM Deep Red. Further investigation using isosurface visualisation of confocal imaging confirmed that lysosomal accumulation occurs in plasma treated cells. The accumulation of lysosomes was associated with concomitant cell death following plasma treatment. In conclusion, we observed rapid accumulation of acidic vesicles and cell death following CAP treatment in GBM cells. We found no evidence that either apoptosis or autophagy, however, determined that a rapid accumulation of late stage endosomes/lysosomes precedes membrane permeabilisation, mitochondrial membrane depolarisation and caspase independent cell death.


Asunto(s)
Glioblastoma/patología , Lisosomas/metabolismo , Gases em Plasma/farmacología , Autofagia/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Lisosomas/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Transducción de Señal/efectos de los fármacos
9.
J Cereb Blood Flow Metab ; 39(6): 1111-1121, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29260627

RESUMEN

Nitrones (e.g. α-phenyl-N-tert-butyl nitrone; PBN) are cerebroprotective in experimental stroke. Free radical trapping is their proposed mechanism. As PBN has low radical trapping potency, we tested Sgk1 induction as another possible mechanism. PBN was injected (100 mg/kg, i.p.) into adult male rats and mice. Sgk1 was quantified in cerebral tissue by microarray, quantitative RT-PCR and western analyses. Sgk1+/+ and Sgk1-/- mice were randomized to receive PBN or saline immediately following transient (60 min) occlusion of the middle cerebral artery. Neurological deficit was measured at 24 h and 48 h and infarct volume at 48 h post-occlusion. Following systemic PBN administration, rapid induction of Sgk1 was detected by microarray (at 4 h) and confirmed by RT-PCR and phosphorylation of the Sgk1-specific substrate NDRG1 (at 6 h). PBN-treated Sgk1+/+ mice had lower neurological deficit ( p < 0.01) and infarct volume ( p < 0.01) than saline-treated Sgk1+/+ mice. PBN-treated Sgk1-/- mice did not differ from saline-treated Sgk1-/- mice. Saline-treated Sgk1-/- and Sgk1+/+ mice did not differ. Brain Sgk3:Sgk1 mRNA ratio was 1.0:10.6 in Sgk1+/+ mice. Sgk3 was not augmented in Sgk1-/- mice. We conclude that acute systemic treatment with PBN induces Sgk1 in brain tissue. Sgk1 may play a part in PBN-dependent actions in acute brain ischemia.


Asunto(s)
Óxidos N-Cíclicos/uso terapéutico , Proteínas Inmediatas-Precoces/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Animales , Encéfalo/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Óxidos N-Cíclicos/farmacología , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Ratones , Ratones Noqueados , Óxidos de Nitrógeno/farmacología , Óxidos de Nitrógeno/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/farmacología , Ratas , Accidente Cerebrovascular/tratamiento farmacológico , Activación Transcripcional/efectos de los fármacos
10.
Cancers (Basel) ; 10(6)2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-29848950

RESUMEN

This special issue on mammalian target of rapamycin (mTOR) explores the importance of mTOR in cell growth control and cancer. Cancer cells often exploit mTOR as a mechanism to enhance their capacity to grow. While protein synthesis is by far the best-characterized mTOR-driven process, this special issue also describes a wider array of mTOR-driven biological processes that cancer cells benefit from, including autophagy, cell cycle control, metabolic transformation, angiogenic signaling, and anabolic processes such as nucleotide biosynthesis and ribosomal biogenesis. Other areas of mTOR signaling covered in these reviews delve into cell migration, inflammation, and regulation of transcription factors linked to cancer progression.

11.
Cancers (Basel) ; 10(1)2018 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-29301334

RESUMEN

Throughout the years, research into signalling pathways involved in cancer progression has led to many discoveries of which mechanistic target of rapamycin (mTOR) is a key player. mTOR is a master regulator of cell growth control. mTOR is historically known to promote cell growth by enhancing the efficiency of protein translation. Research in the last decade has revealed that mTOR's role in promoting cell growth is much more multifaceted. While mTOR is necessary for normal human physiology, cancer cells take advantage of mTOR signalling to drive their neoplastic growth and progression. Oncogenic signal transduction through mTOR is a common occurrence in cancer, leading to metabolic transformation, enhanced proliferative drive and increased metastatic potential through neovascularisation. This review focuses on the downstream mTOR-regulated processes that are implicated in the "hallmarks" of cancer with focus on mTOR's involvement in proliferative signalling, metabolic reprogramming, angiogenesis and metastasis.

12.
Essays Biochem ; 61(6): 561-563, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233868

RESUMEN

Essays in Biochemistry volume 61 (issue 6), entitled Signalling Mechanisms in Autophagy, covers a range of topics in autophagy signalling, touching on emerging new details on the mechanisms of autophagy regulation, novel aspects of selective autophagy and how autophagy functions in organelle homeostasis. It also looks at how autophagy research is leading to better understanding of human disease and plant biology that can be exploited for the benefit of society.


Asunto(s)
Autofagia/fisiología , Animales , Autofagia/genética , Homeostasis/genética , Homeostasis/fisiología , Humanos , Orgánulos/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
FEBS Lett ; 579(5): 991-4, 2005 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-15710380

RESUMEN

NDRG1 is phosphorylated by SGK1 (but not PKB) in vivo at three residues each contained within three nonapeptide repeats. Here, we demonstrate that this nonapeptide, like the NDRG1 protein, is phosphorylated by SGK1, but not by PKBalpha or RSK1 in vitro. The inability of PKBalpha and RSK1 to phosphorylate the nonapeptide was traced to residues n+1, n+2 and n-4 (where n is the phosphorylation site). Changing them from Ser, Glu and Ser to Phe, Ala and Pro, respectively, transformed the nonapeptide into an excellent substrate for PKBalpha and RSK1. Our results identify a specific substrate for SGK1 and may facilitate detection of additional physiological substrates for this enzyme.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas Inmediatas-Precoces , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Péptidos/química , Péptidos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-akt , Spodoptera , Especificidad por Sustrato
14.
Methods Enzymol ; 403: 789-99, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16473639

RESUMEN

The hVps34 phosphatidylinositol (PI) 3-kinase plays an important role in the regulation of vesicular trafficking in the endosomal system. hVps34 associates with a myristylated protein kinase, hVps15. The two proteins are targeting to early endosomal membranes by interactions between hVps15 and activated (GTP-bound) Rab5. This leads to the production of the hVps34 product, PI(3)P, in the endosomal membrane, and subsequent recruitment of FYVE and PX domain-containing effector proteins. This chapter describes the analysis of hVps34/hVps15 interactions with Rab5 in tissue culture cells and in vitro.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Técnicas In Vitro , Unión Proteica , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Transcripción Genética , Proteína de Clasificación Vacuolar VPS15 , Proteínas de Unión al GTP rab5/metabolismo
15.
Biochem J ; 384(Pt 3): 489-94, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15461588

RESUMEN

We detected a protein in rabbit skeletal muscle extracts that was phosphorylated rapidly by PKBa (protein kinase Ba), but not by SGK1 (serum- and glucocorticoid-induced kinase 1), and identified it as the cytoskeletal protein FLNc (filamin C). PKBa phosphorylated FLNc at Ser2213 in vitro, which lies in an insert not present in the FLNa and FLNb isoforms. Ser2213 became phosphorylated when C2C12 myoblasts were stimulated with insulin or epidermal growth factor, and phosphorylation was prevented by low concentrations of wortmannin, at which it is a relatively specific inhibitor of phosphoinositide 3-kinase. PD 184352 [an inhibitor of the classical MAPK (mitogen-activated protein kinase) cascade] and/or rapamycin [an inhibitor of mTOR (mammalian target of rapamycin)] had no effect. Insulin also induced the phosphorylation of FLNc at Ser2213 in cardiac muscle in vivo, but not in cardiac muscle that does not express PDK1 (3-phosphoinositide-dependent kinase 1), the upstream activator of PKB. These results identify the muscle-specific isoform FLNc as a new physiological substrate for PKB.


Asunto(s)
Proteínas Contráctiles/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Secuencia de Aminoácidos , Androstadienos/farmacología , Animales , Especificidad de Anticuerpos , Benzamidas/farmacología , Extractos Celulares/química , Línea Celular , Proteínas Contráctiles/química , Proteínas Contráctiles/inmunología , Factor de Crecimiento Epidérmico/farmacología , Filaminas , Humanos , Proteínas Inmediatas-Precoces , Insulina/farmacología , Isoenzimas/metabolismo , Ratones , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/inmunología , Datos de Secuencia Molecular , Peso Molecular , Músculo Esquelético/química , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt , Conejos , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Especificidad por Sustrato , Wortmanina
16.
Biochem J ; 384(Pt 3): 477-88, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15461589

RESUMEN

We detected a protein in rabbit skeletal muscle extracts that was phosphorylated rapidly by SGK1 (serum- and glucocorticoid-induced kinase 1), but not by protein kinase Ba, and identified it as NDRG2 (N-myc downstream-regulated gene 2). SGK1 phosphorylated NDRG2 at Thr330, Ser332 and Thr348 in vitro. All three residues were phosphorylated in skeletal muscle from wild-type mice, but not from mice that do not express SGK1. SGK1 also phosphorylated the related NDRG1 isoform at Thr328, Ser330 and Thr346 (equivalent to Thr330, Ser332 and Thr348 of NDRG2), as well as Thr356 and Thr366. Residues Thr346, Thr356 and Thr366 are located within identical decapeptide sequences GTRSRSHTSE, repeated three times in NDRG1. These threonines were phosphorylated in NDRG1 in the liver, lung, spleen and skeletal muscle of wild-type mice, but not in SGK1-/- mice. Knock-down of SGK1 in HeLa cells using small interfering RNA also suppressed phosphorylation of the threonine residues in the repeat region of NDRG1. The phosphorylation of NDRG1 by SGK1 transformed it into an excellent substrate for GSK3 (glycogen synthase kinase 3), which could then phosphorylate Ser342, Ser352 and Ser362 in the repeat region. Incubation of HeLa cells with the specific GSK3 inhibitor CT 99021 increased the electrophoretic mobility of NDRG1 in HeLa cells, demonstrating that this protein is phosphorylated by GSK3 in cells. Our results identify NDRG1 and NDRG2 as physiological substrates for SGK1, and demonstrate that phosphorylation of NDRG1 by SGK1 primes it for phosphorylation by GSK3.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos , Proteínas de Ciclo Celular/clasificación , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Línea Celular , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células HeLa , Humanos , Proteínas Inmediatas-Precoces , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Peso Molecular , Familia de Multigenes , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas/clasificación , Proteínas/genética , Proteínas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Conejos , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Especificidad por Sustrato , Proteínas Supresoras de Tumor
17.
Autophagy ; 10(10): 1787-800, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25136802

RESUMEN

Autophagy, a "self-eating" cellular process, has dual roles in promoting and suppressing tumor growth, depending on cellular context. PTP4A3/PRL-3, a plasma membrane and endosomal phosphatase, promotes multiple oncogenic processes including cell proliferation, invasion, and cancer metastasis. In this study, we demonstrate that PTP4A3 accumulates in autophagosomes upon inhibition of autophagic degradation. Expression of PTP4A3 enhances PIK3C3-BECN1-dependent autophagosome formation and accelerates LC3-I to LC3-II conversion in an ATG5-dependent manner. PTP4A3 overexpression also enhances the degradation of SQSTM1, a key autophagy substrate. These functions of PTP4A3 are dependent on its catalytic activity and prenylation-dependent membrane association. These results suggest that PTP4A3 functions to promote canonical autophagy flux. Unexpectedly, following autophagy activation, PTP4A3 serves as a novel autophagic substrate, thereby establishing a negative feedback-loop that may be required to fine-tune autophagy activity. Functionally, PTP4A3 utilizes the autophagy pathway to promote cell growth, concomitant with the activation of AKT. Clinically, from the largest ovarian cancer data set (GSE 9899, n = 285) available in GEO, high levels of expression of both PTP4A3 and autophagy genes significantly predict poor prognosis of ovarian cancer patients. These studies reveal a critical role of autophagy in PTP4A3-driven cancer progression, suggesting that autophagy could be a potential Achilles heel to block PTP4A3-mediated tumor progression in stratified patients with high expression of both PTP4A3 and autophagy genes.


Asunto(s)
Autofagia , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Biocatálisis/efectos de los fármacos , Células CHO , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cloroquina/farmacología , Cricetinae , Cricetulus , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Neoplasias Ováricas/genética , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Prenilación/efectos de los fármacos , Proteína Sequestosoma-1 , Especificidad por Sustrato/efectos de los fármacos , Análisis de Supervivencia
18.
Autophagy ; 10(10): 1749-60, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25126726

RESUMEN

Birt-Hogg-Dubé (BHD) syndrome is a rare autosomal dominant condition caused by mutations in the FLCN gene and characterized by benign hair follicle tumors, pneumothorax, and renal cancer. Folliculin (FLCN), the protein product of the FLCN gene, is a poorly characterized tumor suppressor protein, currently linked to multiple cellular pathways. Autophagy maintains cellular homeostasis by removing damaged organelles and macromolecules. Although the autophagy kinase ULK1 drives autophagy, the underlying mechanisms are still being unraveled and few ULK1 substrates have been identified to date. Here, we identify that loss of FLCN moderately impairs basal autophagic flux, while re-expression of FLCN rescues autophagy. We reveal that the FLCN complex is regulated by ULK1 and elucidate 3 novel phosphorylation sites (Ser406, Ser537, and Ser542) within FLCN, which are induced by ULK1 overexpression. In addition, our findings demonstrate that FLCN interacts with a second integral component of the autophagy machinery, GABA(A) receptor-associated protein (GABARAP). The FLCN-GABARAP association is modulated by the presence of either folliculin-interacting protein (FNIP)-1 or FNIP2 and further regulated by ULK1. As observed by elevation of GABARAP, sequestome 1 (SQSTM1) and microtubule-associated protein 1 light chain 3 (MAP1LC3B) in chromophobe and clear cell tumors from a BHD patient, we found that autophagy is impaired in BHD-associated renal tumors. Consequently, this work reveals a novel facet of autophagy regulation by ULK1 and substantially contributes to our understanding of FLCN function by linking it directly to autophagy through GABARAP and ULK1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Homólogo de la Proteína 1 Relacionada con la Autofagia , Síndrome de Birt-Hogg-Dubé/metabolismo , Síndrome de Birt-Hogg-Dubé/patología , Proteínas Portadoras/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/deficiencia , Proteína Sequestosoma-1 , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia
19.
Oncotarget ; 5(6): 1609-20, 2014 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-24742492

RESUMEN

TBX2 is an oncogenic transcription factor known to drive breast cancer proliferation. We have identified the cysteine protease inhibitor Cystatin 6 (CST6) as a consistently repressed TBX2 target gene, co-repressed through a mechanism involving Early Growth Response 1 (EGR1). Exogenous expression of CST6 in TBX2-expressing breast cancer cells resulted in significant apoptosis whilst non-tumorigenic breast cells remained unaffected. CST6 is an important tumor suppressor in multiple tissues, acting as a dual protease inhibitor of both papain-like cathepsins and asparaginyl endopeptidases (AEPs) such as Legumain (LGMN). Mutation of the CST6 LGMN-inhibitory domain completely abrogated its ability to induce apoptosis in TBX2-expressing breast cancer cells, whilst mutation of the cathepsin-inhibitory domain or treatment with a pan-cathepsin inhibitor had no effect, suggesting that LGMN is the key oncogenic driver enzyme. LGMN activity assays confirmed the observed growth inhibitory effects were consistent with CST6 inhibition of LGMN. Knockdown of LGMN and the only other known AEP enzyme (GPI8) by siRNA confirmed that LGMN was the enzyme responsible for maintaining breast cancer proliferation. CST6 did not require secretion or glycosylation to elicit its cell killing effects, suggesting an intracellular mode of action. Finally, we show that TBX2 and CST6 displayed reciprocal expression in a cohort of primary breast cancers with increased TBX2 expression associating with increased metastases. We have also noted that tumors with altered TBX2/CST6 expression show poor overall survival. This novel TBX2-CST6-LGMN signaling pathway, therefore, represents an exciting opportunity for the development of novel therapies to target TBX2 driven breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Cistatina M/genética , Cisteína Endopeptidasas/metabolismo , Proteínas de Dominio T Box/metabolismo , Apoptosis , Western Blotting , Neoplasias de la Mama/genética , Inmunoprecipitación de Cromatina , Cistatina M/metabolismo , Cisteína Endopeptidasas/genética , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteínas de Dominio T Box/antagonistas & inhibidores , Proteínas de Dominio T Box/genética , Células Tumorales Cultivadas
20.
Physiol Rep ; 1(5): e00076, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24303161

RESUMEN

Nutrient provision after sprint exercise enhances mammalian target of rapamycin (mTOR) signaling. One suggested that nutrient sensor is the class III phosphatidylinositol 3-kinase, vacuolar protein sorting 34 (Vps34), not previously studied in human skeletal muscle. It is hypothesized that oral ingestion of essential amino acids (EAA) and carbohydrates (Carb) increases Vps34 activity and mTOR signaling in human skeletal (hVps34) muscle after sprint exercise. Nine subjects were performed 3 × 30-sec all-out sprints with or without ingestion of EAA + Carb or placebo drinks in a randomized order with a month interval. Muscle biopsies were performed at rest and 140 min after last sprint and analyzed for p-mTOR, p-p70S6k, p-eEF2 and for hVps34 activity and hVps34 protein content. Venous blood samples were collected and analyzed for amino acids, glucose, lactate, and insulin. During the sprint exercise session, EAA, glucose, and insulin in blood increased significantly more in EAA + Carb than in placebo. P-mTOR and p-p70S6k were significantly increased above rest in EAA + Carb (P = 0.03, P = 0.007) 140 min after last sprint, but not in placebo. Activity and protein expression of hVps34 were not significantly changed from rest in EAA + Carb 140 min after the last sprint. However, hVps34 activity and protein expression tended to increase in placebo (both P = 0.08). In conclusion, on the contrary to the hypothesis, no increase in activation of hVps34 was found following sprint exercise in EAA + Carb condition. In spite of this, the results support an activation of mTOR during this condition. However, this does not exclude the permissive role of hVps34 in mediating the amino acid-induced activation of mTOR and muscle protein synthesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA