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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 14(5): 500-8, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23525088

RESUMO

T lymphocytes must regulate nutrient uptake to meet the metabolic demands of an immune response. Here we show that the intracellular supply of large neutral amino acids (LNAAs) in T cells was regulated by pathogens and the T cell antigen receptor (TCR). T cells responded to antigen by upregulating expression of many amino-acid transporters, but a single System L ('leucine-preferring system') transporter, Slc7a5, mediated uptake of LNAAs in activated T cells. Slc7a5-null T cells were unable to metabolically reprogram in response to antigen and did not undergo clonal expansion or effector differentiation. The metabolic catastrophe caused by loss of Slc7a5 reflected the requirement for sustained uptake of the LNAA leucine for activation of the serine-threonine kinase complex mTORC1 and for expression of the transcription factor c-Myc. Control of expression of the System L transporter by pathogens is thus a critical metabolic checkpoint for T cells.


Assuntos
Aminoácidos Neutros/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Citotoxicidade Imunológica , Interferon gama/metabolismo , Interleucina-2/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Transporte Proteico , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Regulação para Cima
2.
EMBO Rep ; 21(1): e48469, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31789450

RESUMO

Amino acids are essential for cellular metabolism, and it is important to understand how nutrient supply is coordinated with changing energy requirements during embryogenesis. Here, we show that the amino acid transporter Slc7a5/Lat1 is highly expressed in tissues undergoing morphogenesis and that Slc7a5-null mouse embryos have profound neural and limb bud outgrowth defects. Slc7a5-null neural tissue exhibited aberrant mTORC1 activity and cell proliferation; transcriptomics, protein phosphorylation and apoptosis analyses further indicated induction of the integrated stress response as a potential cause of observed defects. The pattern of stress response gene expression induced in Slc7a5-null embryos was also detected at low level in wild-type embryos and identified stress vulnerability specifically in tissues undergoing morphogenesis. The Slc7a5-null phenotype is reminiscent of Wnt pathway mutants, and we show that Wnt/ß-catenin loss inhibits Slc7a5 expression and induces this stress response. Wnt signalling therefore normally supports the metabolic demands of morphogenesis and constrains cellular stress. Moreover, operation in the embryo of the integrated stress response, which is triggered by pathogen-mediated as well as metabolic stress, may provide a mechanistic explanation for a range of developmental defects.


Assuntos
Transportador 1 de Aminoácidos Neutros Grandes , Via de Sinalização Wnt , Animais , Proliferação de Células/genética , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Morfogênese
3.
J Biol Chem ; 290(13): 8173-84, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25653282

RESUMO

Expression and activity of the System A/SNAT2 (SLC38A2) amino acid transporter is up-regulated by amino acid starvation and hypertonicity by a mechanism dependent on both ATF4-mediated transcription of the SLC38A2 gene and enhanced stabilization of SNAT2 itself, which forms part of an integrated cellular stress response to nutrient deprivation and osmotic stress. Here we demonstrate that this adaptive increase in System A function is restrained in cells subjected to prior incubation with linoleic acid (LOA, an unsaturated C18:2 fatty acid) for 24 h. While fatty acid treatment had no detectable effect upon stress-induced SNAT2 or ATF4 gene transcription, the associated increase in SNAT2 protein/membrane transport activity were strongly suppressed in L6 myotubes or HeLa cells preincubated with LOA. Cellular ubiquitination of many proteins was increased by LOA and although the fatty acid-induced loss of SNAT2 could be attenuated by proteasomal inhibition, the functional increase in System A transport activity associated with amino acid starvation/hypertonicity that depends upon processing/maturation and delivery of SNAT2 to the cell surface could not be rescued. LOA up-regulated cellular expression of Nedd4.2, an E3-ligase implicated in SNAT2 ubiquitination, but shRNA-directed Nedd4.2 gene silencing could not curb fatty acid-induced loss of SNAT2 adaptation. However, expression of SNAT2 in which seven putative lysyl-ubiquitination sites in the cytoplasmic N-terminal domain were mutated to alanine protected SNAT2 against LOA-induced proteasomal degradation. Collectively, our findings indicate that increased availability of unsaturated fatty acids can compromise the stress-induced induction/adaptation in SNAT2 expression and function by promoting its degradation via the ubiquitin-proteasome system.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Ácido Linoleico/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Células HEK293 , Células HeLa , Humanos , Fibras Musculares Esqueléticas/metabolismo , Ubiquitina-Proteína Ligases Nedd4 , Pressão Osmótica , Ratos , Transcrição Gênica , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima
4.
Biochem J ; 470(2): 207-21, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26348909

RESUMO

The mammalian or mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) is a ubiquitously expressed multimeric protein kinase complex that integrates nutrient and growth factor signals for the co-ordinated regulation of cellular metabolism and cell growth. Herein, we demonstrate that suppressing the cellular activity of glycogen synthase kinase-3 (GSK3), by use of pharmacological inhibitors or shRNA-mediated gene silencing, results in substantial reduction in amino acid (AA)-regulated mTORC1-directed signalling, as assessed by phosphorylation of multiple downstream mTORC1 targets. We show that GSK3 regulates mTORC1 activity through its ability to phosphorylate the mTOR-associated scaffold protein raptor (regulatory-associated protein of mTOR) on Ser(859). We further demonstrate that either GSK3 inhibition or expression of a S859A mutated raptor leads to reduced interaction between mTOR and raptor and under these circumstances, irrespective of AA availability, there is a consequential loss in phosphorylation of mTOR substrates, such as p70S6K1 (ribosomal S6 kinase 1) and uncoordinated-51-like kinase (ULK1), which results in increased autophagic flux and reduced cellular proliferation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aminoácidos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Autofagia , Linhagem Celular , Proliferação de Células , Inativação Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Humanos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Dados de Sequência Molecular , Mutação , Fosforilação , RNA Interferente Pequeno/genética , Ratos , Proteína Regulatória Associada a mTOR , Serina/genética , Serina/metabolismo , Transdução de Sinais
5.
Nucleic Acids Res ; 42(8): 5083-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24526220

RESUMO

Ribosome biogenesis is a key process for maintaining protein synthetic capacity in dividing or growing cells, and requires coordinated production of ribosomal proteins and ribosomal RNA (rRNA), including the processing of the latter. Signalling through mammalian target of rapamycin complex 1 (mTORC1) activates all these processes. Here, we show that, in human cells, impaired rRNA processing, caused by expressing an interfering mutant of BOP1 or by knocking down components of the PeBoW complex elicits activation of mTORC1 signalling. This leads to enhanced phosphorylation of its substrates S6K1 and 4E-BP1, and stimulation of proteins involved in translation initiation and elongation. In particular, we observe both inactivation and downregulation of the eukaryotic elongation factor 2 kinase, which normally inhibits translation elongation. The latter effect involves decreased expression of the eEF2K mRNA. The mRNAs for ribosomal proteins, whose translation is positively regulated by mTORC1 signalling, also remain associated with ribosomes. Therefore, our data demonstrate that disrupting rRNA production activates mTORC1 signalling to enhance the efficiency of the translational machinery, likely to help compensate for impaired ribosome production.


Assuntos
Complexos Multiproteicos/metabolismo , Biossíntese de Proteínas , RNA Ribossômico/biossíntese , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Fator 2 de Elongação de Peptídeos/metabolismo , Fatores de Alongamento de Peptídeos/metabolismo , Fosforilação , Polirribossomos/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteínas de Ligação a RNA , Proteínas Quinases S6 Ribossômicas/metabolismo , Deleção de Sequência , Transdução de Sinais
6.
Curr Opin Clin Nutr Metab Care ; 16(1): 57-65, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23196813

RESUMO

PURPOSE OF REVIEW: We consider recent advances in epithelial amino acid transport physiology and our understanding of the functioning of amino acid transporters as sensors, as well as carriers, of tissue nutrient supplies. RECENT FINDINGS: Gut hormones (e.g. leptin) may regulate intestinal amino acid transporter activity by a variety of mechanisms, although the overall functional significance of such regulation is not yet fully understood. Important functional interactions between amino acid transporters and nutrient-signalling pathways which regulate metabolism [e.g. the mammalian target of rapamycin (mTOR)C1 pathway which promotes cell growth] have been revealed in recent studies. Amino acid transporters on endosomal (e.g. lysosomal) membranes may be of unexpected significance as intracellular nutrient sensors. It is also now evident that certain amino acid transporters may have dual receptor-transporter functions and act as 'transceptors' to sense amino acid availability upstream of signal pathways. SUMMARY: Increased knowledge on the timescale of the amino acid sensor-signal-effector process(es) should help in the optimization of protein-feeding regimes to gain maximum anabolic effect. New opportunities for nutritional therapy include targeting of amino acid transceptors to promote protein-anabolic signals and mechanisms up-regulating amino acid transporter expression to improve absorptive capacity for nutrients.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Alimentos , Absorção , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+L de Transporte de Aminoácidos , Aminoácidos/farmacocinética , Animais , Hormônios Gastrointestinais/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Proteínas/genética , Proteínas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR , Regulação para Cima
7.
J Physiol ; 587(1): 253-60, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19015198

RESUMO

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.


Assuntos
Contração Muscular/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Feminino , Hipertrofia , Leucina/metabolismo , Camundongos , Músculo Esquelético/patologia , Músculo Esquelético/fisiologia , Esforço Físico/fisiologia , Ratos , Ratos Wistar , Treinamento Resistido , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Estresse Mecânico , Fatores de Transcrição/metabolismo
8.
Am J Physiol Endocrinol Metab ; 297(3): E822-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19622785

RESUMO

The System L transporter facilitates cellular import of large neutral amino acids (AAs) such as Leu, a potent activator of the intracellular target of rapamycin (TOR) pathway, which signals for cell growth. System L is an AA exchanger, proposed to accumulate certain AAs by coupling to dissipation of concentration gradient(s) of exchange substrates generated by secondary active AA transporters such as System A (SNAT2). We addressed the hypothesis that this type of coupling (termed tertiary active transport) acts as an indirect mechanism to extend the range of AA stimulating TOR to those transported by both Systems A and L (e.g., Gln) through downstream enhancement of Leu accumulation. System A overexpression enabled Xenopus oocytes to accumulate substrate AAs (notably Ser, Gln, Ala, Pro, Met; totaling 2.6 nmol/oocyte) from medium containing a physiological AA mixture at plasma concentrations. Net accumulation of System L (4F2hc-xLAT1) substrates from this medium by System L-overexpressing oocytes was increased by 90% (from 0.7 to 1.35 nmol/oocyte; mainly Leu, Ile) when Systems A and L were coexpressed, coincident with a decline in accumulation of specific System A substrates (Gln, Ser, Met), as expected if the latter were also System L substrates and functional coupling of the transport Systems occurred. AA flux coupling was confirmed as trans-stimulation of Leu influx in System L-expressing oocytes by Gln injection (0.5 nmol/oocyte). The observed changes in Leu accumulation are sufficient to activate the TOR pathway in oocytes, although intracellular AA metabolism limits the potential for AA accumulation by tertiary active transport in this system.


Assuntos
Sistema A de Transporte de Aminoácidos/genética , Sistema L de Transporte de Aminoácidos/genética , Aminoácidos/metabolismo , Expressão Gênica , Oócitos/metabolismo , Xenopus/genética , Sistema A de Transporte de Aminoácidos/metabolismo , Sistema A de Transporte de Aminoácidos/fisiologia , Sistema L de Transporte de Aminoácidos/metabolismo , Sistema L de Transporte de Aminoácidos/fisiologia , Animais , Transporte Biológico/genética , Feminino , Expressão Gênica/fisiologia , Modelos Biológicos , Fatores de Tempo , Transfecção , Xenopus/metabolismo , beta-Alanina/análogos & derivados , beta-Alanina/farmacocinética
9.
Biochem Soc Trans ; 37(Pt 1): 237-41, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19143639

RESUMO

Nutrient signalling by the mTOR (mammalian target of rapamycin) pathway involves upstream sensing of free AA (amino acid) concentrations. Several AA-regulated kinases have recently been identified as putative intracellular AA sensors. Their activity will reflect the balance between AA flows through underlying mechanisms which together determine the size of the intracellular free AA pool. For indispensable AAs, these mechanisms are primarily (i) AA transport across the cell membrane, and (ii) protein synthesis/breakdown. The System L AA transporter is the primary conduit for cellular entry of indispensable neutral AAs (including leucine and phenylalanine) and potentially a key modulator of AA-sensitive mTOR signalling. Coupling of substrate flows through System L and other AA transporters (e.g. System A) may extend the scope for sensing nutrient abundance. Factors influencing AA transporter activity (e.g. hormones) may affect intracellular AA concentrations and hence indirectly mTOR pathway activity. Several AA transporters are themselves regulated by AA availability through 'adaptive regulation', which may help to adjust the gain of AA sensing. The substrate-binding sites of AA transporters are potentially direct sensors of AA availability at both faces of the cell surface, and there is growing evidence that AA transporters of the SNAT (sodium-coupled neutral AA transporter) and PAT (proton-assisted AA transporter) families may operate, at least under some circumstances, as transporter-like sensors (or 'transceptors') upstream of mTOR.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Alimentos , Transdução de Sinais , Aminoácidos/metabolismo , Animais , Humanos , Espaço Intracelular/metabolismo , Modelos Biológicos
10.
Biochim Biophys Acta Mol Cell Res ; 1866(6): 978-991, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30857869

RESUMO

Extracellular amino acid (AA) withdrawal/restriction invokes an integrated stress response (ISR) that induces global suppression of protein synthesis whilst allowing transcription and translation of a select group of genes, whose protein products facilitate cellular adaptation to AA insufficiency. Transcriptional induction of the System A/SNAT2 AA transporter represents a classic adaptation response and crucially depends upon activation of the General Control Nonderepressible-2 kinase/Activating transcription factor 4 (GCN2/ATF4) pathway. However, the ISR may also include additional signalling inputs operating in conjunction or independently of GCN2/ATF4 to upregulate SNAT2. Herein, we show that whilst pharmacological inhibition of MEK-ERK, mTORC1 and p38 MAP kinase signalling has no detectable effect on System A upregulation, inhibitors targeting GSK3 (e.g. SB415286) caused significant repression of the SNAT2 adaptation response. Strikingly, the effects of SB415286 persist in cells in which GSK3α/ß have been stably silenced indicating an off-target effect. We show that SB415286 can also inhibit cyclin-dependent kinases (CDK) and that roscovitine and flavopiridol (two pan CDK inhibitors) are effective repressors of the SNAT2 adaptive response. In particular, our work reveals that CDK7 activity is upregulated in AA-deprived cells in a GCN-2-dependent manner and that a potent and selective CDK7 inhibitor, THZ-1, not only attenuates the increase in ATF4 expression but blocks System A adaptation. Importantly, the inhibitory effects of THZ-1 on System A adaptation are mitigated in cells expressing a doxycycline-inducible drug-resistant form of CDK7. Our data identify CDK7 as a novel component of the ISR regulating System A adaptation in response to AA insufficiency.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Aminoácidos/deficiência , Quinases Ciclina-Dependentes/metabolismo , Estresse Fisiológico , Fator 4 Ativador da Transcrição/metabolismo , Aminofenóis/farmacologia , Animais , Linhagem Celular , Flavonoides/farmacologia , Células HEK293 , Células HeLa , Humanos , Maleimidas/farmacologia , Fenilenodiaminas/farmacologia , Piperidinas/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/farmacologia , Ratos , Roscovitina/farmacologia , Quinase Ativadora de Quinase Dependente de Ciclina
11.
Sci Signal ; 12(589)2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31289211

RESUMO

L-type amino acid transporter 1 (LAT1), which is encoded by solute carrier transporter 7a5 (Slc7a5), plays a crucial role in amino acid sensing and signaling in specific cell types, contributing to the pathogenesis of cancer and neurological disorders. Amino acid substrates of LAT1 have a beneficial effect on bone health directly and indirectly, suggesting a potential role for LAT1 in bone homeostasis. Here, we identified LAT1 in osteoclasts as important for bone homeostasis. Slc7a5 expression was substantially reduced in osteoclasts in a mouse model of ovariectomy-induced osteoporosis. The osteoclast-specific deletion of Slc7a5 in mice led to osteoclast activation and bone loss in vivo, and Slc7a5 deficiency increased osteoclastogenesis in vitro. Loss of Slc7a5 impaired activation of the mechanistic target of rapamycin complex 1 (mTORC1) pathway in osteoclasts, whereas genetic activation of mTORC1 corrected the enhanced osteoclastogenesis and bone loss in Slc7a5-deficient mice. Last, Slc7a5 deficiency increased the expression of nuclear factor of activated T cells, cytoplasmic 1 (Nfatc1) and the nuclear accumulation of NFATc1, a master regulator of osteoclast function, possibly through the canonical nuclear factor κB pathway and the Akt-glycogen synthase kinase 3ß signaling axis, respectively. These findings suggest that the LAT1-mTORC1 axis plays a pivotal role in bone resorption and bone homeostasis by modulating NFATc1 in osteoclasts, thereby providing a molecular connection between amino acid intake and skeletal integrity.


Assuntos
Sistema y+L de Transporte de Aminoácidos/genética , Osso e Ossos/metabolismo , Homeostase/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Osteoclastos/metabolismo , Osteogênese/genética , Sistema y+L de Transporte de Aminoácidos/deficiência , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Osso e Ossos/citologia , Células Cultivadas , Feminino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Ovariectomia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética
12.
Elife ; 82019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30916644

RESUMO

Immune activated T lymphocytes modulate the activity of key metabolic pathways to support the transcriptional reprograming and reshaping of cell proteomes that permits effector T cell differentiation. The present study uses high resolution mass spectrometry and metabolic labelling to explore how murine T cells control the methionine cycle to produce methyl donors for protein and nucleotide methylations. We show that antigen receptor engagement controls flux through the methionine cycle and RNA and histone methylations. We establish that the main rate limiting step for protein synthesis and the methionine cycle is control of methionine transporter expression. Only T cells that respond to antigen to upregulate and sustain methionine transport are supplied with methyl donors that permit the dynamic nucleotide methylations and epigenetic reprogramming that drives T cell differentiation. These data highlight how the regulation of methionine transport licenses use of methionine for multiple fundamental processes that drive T lymphocyte proliferation and differentiation.


Assuntos
Metionina/metabolismo , Receptores de Antígenos/metabolismo , Linfócitos T/metabolismo , Animais , Histonas/metabolismo , Espectrometria de Massas , Análise do Fluxo Metabólico , Metilação , Camundongos Endogâmicos C57BL , Processamento de Proteína Pós-Traducional , RNA/metabolismo , Processamento Pós-Transcricional do RNA
13.
Nat Commun ; 9(1): 1981, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773791

RESUMO

The tryptophan metabolite kynurenine has critical immunomodulatory properties and can function as an aryl hydrocarbon receptor (AHR) ligand. Here we show that the ability of T cells to transport kynurenine is restricted to cells activated by the T-cell antigen receptor or proinflammatory cytokines. Kynurenine is transported across the T-cell membrane by the System L transporter SLC7A5. Accordingly, the ability of kynurenine to activate the AHR is restricted to T cells that express SLC7A5. We use the fluorescence spectral properties of kynurenine to develop a flow cytometry-based assay for rapid, sensitive and quantitative measurement of the kynurenine transport capacity in a single cell. Our findings provide a method to assess the susceptibility of T cells to kynurenine, and a sensitive single cell assay to monitor System L amino acid transport.


Assuntos
Cinurenina/imunologia , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Análise de Célula Única , Linfócitos T/imunologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Cinurenina/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Transportador 1 de Aminoácidos Neutros Grandes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T/metabolismo
14.
Front Pharmacol ; 9: 63, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29467657

RESUMO

The SNAT2 (SLC38A2) System A amino acid transporter mediates Na+-coupled cellular uptake of small neutral α-amino acids (AAs) and is extensively regulated in response to humoral and nutritional cues. Understanding the basis of such regulation is important given that AA uptake via SNAT2 has been linked to activation of mTORC1; a major controller of many important cellular processes including, for example, mRNA translation, lipid synthesis, and autophagy and whose dysregulation has been implicated in the development of cancer and conditions such as obesity and type 2 diabetes. Extracellular AA withdrawal induces an adaptive upregulation of SNAT2 gene transcription and SNAT2 protein stability but, as yet, the sensing mechanism(s) that initiate this response remain poorly understood although interactions between SNAT2 and its substrates may play a vital role. Herein, we have explored how changes in substrate (AA and Na+) availability impact upon the adaptive regulation of SNAT2 in HeLa cells. We show that while AA deprivation induces SNAT2 gene expression, this induction was not apparent if extracellular Na+ was removed during the AA withdrawal period. Furthermore, we show that the increase in SNAT2 protein stability associated with AA withdrawal is selectively repressed by provision of SNAT2 AA substrates (N-methylaminoisobutyric acid and glutamine), but not non-substrates. This stabilization and substrate-induced repression were critically dependent upon the cytoplasmic N-terminal tail of SNAT2 (containing lysyl residues which are putative targets of the ubiquitin-proteasome system), because "grafting" this tail onto SNAT5, a related SLC38 family member that does not exhibit adaptive regulation, confers substrate-induced changes in stability of the SNAT2-5 chimeric transporter. In contrast, expression of SNAT2 in which the N-terminal lysyl residues were mutated to alanine rendered the transporter stable and insensitive to substrate-induced changes in protein stability. Intriguingly, SNAT2 protein stability was dramatically reduced in the absence of extracellular Na+ irrespective of whether substrate AAs were present or absent. Our findings indicate that the presence of extracellular Na+ (and potentially its binding to SNAT2) may be crucial for not only sensing SNAT2 AA occupancy and consequently for initiating the adaptive response under AA insufficient conditions, but for enabling substrate-induced changes in SNAT2 protein stability.

15.
Best Pract Res Clin Endocrinol Metab ; 21(2): 237-51, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17574006

RESUMO

Thyroid hormones (THs) -- thyroxine (T4) and tri-iodothyronine (T3) -- are iodinated derivatives of the amino acid tyrosine, which regulates growth, development and critical metabolic functions. THs are taken up by target cells and act at the genomic level via nuclear thyroid receptors. Saturable transport mechanisms mediate the greater part of TH movement across the plasma membrane. System L1 permease is a transporter of THs and amino acids in mammalian adipose tissue, placenta and brain. T(3) is also a substrate of a putative System T transporter, which is selective for aromatic amino acids. The activity and functional mechanisms of these transporters can be crucial to cells in determining both their hormone sensitivity and their responses to change in circulating hormone concentrations or availability of competing substrates (e.g. amino acids). TH transporters are potentially important pharmacological targets in the design of novel or improved therapies for thyroid-related disorders.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Hormônios Tireóideos/metabolismo , Doença Aguda , Tecido Adiposo/metabolismo , Sistemas de Transporte de Aminoácidos/fisiologia , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Humanos , Modelos Biológicos , Especificidade de Órgãos , Fenilcetonúrias/metabolismo , Placenta/metabolismo , Hormônios Tireóideos/farmacologia , Triptofano/deficiência , Triptofano/metabolismo
16.
Biochem J ; 397(2): 369-75, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16629640

RESUMO

System A and N amino acid transporters are key effectors of movement of amino acids across the plasma membrane of mammalian cells. These Na+-dependent transporters of the SLC38 gene family are highly sensitive to changes in pH within the physiological range, with transport markedly depressed at pH 7.0. We have investigated the possible role of histidine residues in the transporter proteins in determining this pH-sensitivity. The histidine-modifying agent DEPC (diethyl pyrocarbonate) markedly reduces the pH-sensitivity of SNAT2 and SNAT5 transporters (representative isoforms of System A and N respectively, overexpressed in Xenopus oocytes) in a concentration-dependent manner but does not completely inactivate transport activity. These effects of DEPC were reversed by hydroxylamine and partially blocked in the presence of excess amino acid substrate. DEPC treatment also blocked a reduction in apparent affinity for Na+ (K0.5Na+) of the SNAT2 transporter at low external pH. Mutation of the highly conserved C-terminal histidine residue to alanine in either SNAT2 (H504A) or SNAT5 (H471A) produced a transport phenotype exhibiting reduced, DEPC-resistant pH-sensitivity with no change in K0.5Na+ at low external pH. We suggest that the pH-sensitivity of these structurally related transporters results at least partly from a common allosteric mechanism influencing Na+ binding, which involves an H+-modifier site associated with C-terminal histidine residues.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/fisiologia , Sistemas de Transporte de Aminoácidos/fisiologia , Histidina/química , Sítio Alostérico , Sistema A de Transporte de Aminoácidos , Animais , Membrana Celular/metabolismo , Sequência Conservada , Feminino , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Oócitos/metabolismo , Transporte Proteico , Ratos , Xenopus laevis
17.
FASEB J ; 19(3): 461-3, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15611152

RESUMO

Skeletal muscle is a major insulin target tissue and has a prominent role in the control of body amino acid economy, being the principal store of free and protein-bound amino acids and a dominant locus for amino acid metabolism. Interplay between diverse stimuli (e.g., hormonal/nutritional/mechanical) modulates muscle insulin action to serve physiological need through the action of factors such as intramuscular signaling molecules. Ceramide, a product of sphingolipid metabolism and cytokine signaling, has a potent contra-insulin action with respect to the transport and deposition of glucose in skeletal muscle, although ceramide effects on muscle amino acid turnover have not previously been documented. Here, membrane permeant C2-ceramide is shown to attenuate the basal and insulin-stimulated activity of the Na+-dependent System A amino acid transporter in rat muscle cells (L6 myotubes) by depletion of the plasma membrane abundance of SNAT2 (a System A isoform). Concomitant with transporter down-regulation, ceramide diminished both intramyocellular amino acid abundance and the phosphorylation of translation regulators lying downstream of mTOR. The physiological outcome of ceramide signaling in this instance is a marked reduction in cellular protein synthesis, a result that is likely to represent an important component of the processes leading to muscle wasting in catabolic conditions.


Assuntos
Sistema A de Transporte de Aminoácidos/antagonistas & inibidores , Ceramidas/farmacologia , Células Musculares/metabolismo , Proteínas Musculares/biossíntese , Músculo Esquelético/metabolismo , Sistema A de Transporte de Aminoácidos/efeitos dos fármacos , Sistema A de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos/análise , Sistemas de Transporte de Aminoácidos/antagonistas & inibidores , Sistemas de Transporte de Aminoácidos/fisiologia , Aminoácidos/análise , Aminoácidos/metabolismo , Animais , Linhagem Celular , Membrana Celular/química , Glucose/metabolismo , Células Musculares/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/química , Músculo Esquelético/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR
18.
FASEB J ; 19(3): 422-4, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15596483

RESUMO

The nature of the deficit underlying age-related muscle wasting remains controversial. To test whether it could be due to a poor anabolic response to dietary amino acids, we measured the rates of myofibrillar and sarcoplasmic muscle protein synthesis (MPS) in 44 healthy young and old men, of similar body build, after ingesting different amounts of essential amino acids (EAA). Basal rates of MPS were indistinguishable, but the elderly showed less anabolic sensitivity and responsiveness of MPS to EAA, possibly due to decreased intramuscular expression, and activation (phosphorylation) after EAA, of amino acid sensing/signaling proteins (mammalian target of rapamycin, mTOR; p70 S6 kinase, or p70(S6k); eukaryotic initiation factor [eIF]4BP-1; and eIF2B). The effects were independent of insulin signaling since plasma insulin was clamped at basal values. Associated with the anabolic deficits were marked increases in NFkappaB, the inflammation-associated transcription factor. These results demonstrate first, EAA stimulate MPS independently of increased insulin availability; second, in the elderly, a deficit in MPS in the basal state is unlikely; and third, the decreased sensitivity and responsiveness of MPS to EAA, associated with decrements in the expression and activation of components of anabolic signaling pathways, are probably major contributors to the failure of muscle maintenance in the elderly. Countermeasures to maximize muscle maintenance should target these deficits.


Assuntos
Envelhecimento/fisiologia , Aminoácidos Essenciais/metabolismo , Proteínas Musculares/biossíntese , Músculo Esquelético/fisiologia , Transdução de Sinais , Adulto , Idoso , Aminoácidos Essenciais/administração & dosagem , Isótopos de Carbono , Ingestão de Líquidos , Resistência a Medicamentos , Fatores de Iniciação em Eucariotos/análise , Humanos , Cetoácidos/administração & dosagem , Cetoácidos/metabolismo , Cetoácidos/farmacocinética , Cinética , Leucina/administração & dosagem , Leucina/metabolismo , Leucina/farmacocinética , Masculino , Músculo Esquelético/química , NF-kappa B/análise , Proteínas Quinases/análise , Proteínas Quinases S6 Ribossômicas 70-kDa/análise , Soluções , Serina-Treonina Quinases TOR
19.
Cell Signal ; 28(5): 412-424, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26827808

RESUMO

Iron is an indispensable micronutrient that regulates many aspects of cell function, including growth and proliferation. These processes are critically dependent upon signalling via the mammalian or mechanistic target of rapamycin complex 1 (mTORC1). Herein, we test whether iron depletion induced by cell incubation with the iron chelator, deferoxamine (DFO), mediates its effects on cell growth through mTORC1-directed signalling and protein synthesis. We have used Caco-2 cells, a well-established in vitro model of human intestinal epithelia. Iron depletion increased expression of iron-regulated proteins (TfR, transferrin receptor and DMT1, divalent metal transporter, as predicted, but it also promoted a marked reduction in growth and proliferation of Caco-2 cells. This was strongly associated with suppressed mTORC1 signalling, as judged by reduced phosphorylation of mTOR substrates, S6K1 and 4E-BP1, and diminished protein synthesis. The reduction in mTORC1 signalling was tightly coupled with increased expression and accumulation of REDD1 (regulated in DNA damage and development 1) and reduced phosphorylation of Akt and TSC2. The increase in REDD1 abundance was rapidly reversed upon iron repletion of cells but was also attenuated by inhibitors of gene transcription, protein phosphatase 2A (PP2A) and by REDD1 siRNA--strategies that also antagonised the loss in mTORC1 signalling associated with iron depletion. Our findings implicate REDD1 and PP2A as crucial regulators of mTORC1 activity in iron-depleted cells and indicate that their modulation may help mitigate atrophy of the intestinal mucosa that may occur in response to iron deficiency.


Assuntos
Mucosa Intestinal/metabolismo , Deficiências de Ferro , Complexos Multiproteicos/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo , Aminoácidos/metabolismo , Células CACO-2 , Proliferação de Células/efeitos dos fármacos , Desferroxamina/farmacologia , Regulação para Baixo , Humanos , Mucosa Intestinal/enzimologia , Quelantes de Ferro/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Biossíntese de Proteínas , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/biossíntese
20.
Pharmacol Ther ; 94(3): 235-51, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12113800

RESUMO

The thyroid hormone (TH; 3,3',5,5'-tetra-iodothyronine and 3,3',5'-triiodothyronine) regulates growth, development, and critical metabolic functions. Thyroid diseases are among the most prevalent group of metabolic disorders in the Western world. TH exerts effects through complex biological pathways, which offer a wealth of opportunities to pharmacologically intervene in TH signalling at numerous steps. These include biosynthesis, cell-specific uptake or export (involving L-type amino acid transporter, organic anion transporter, organic cation transporter, or multidrug resistance transporter), as well as nuclear targeting and actions (the latter including TH receptor binding and histone acetylation/deacetylation). Such processes represent potentially important pharmacological targets for the design of novel or improved therapies for TH disorders, obesity, and cardiovascular diseases.


Assuntos
Hormônios Tireóideos/fisiologia , Receptores dos Hormônios Tireóideos/metabolismo , Hormônios Tireóideos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA