Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
HLA ; 103(1): e15252, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37848366

RESUMEN

T cell therapy strategies, from allogeneic stem cell transplantation toward genetically-modified T cells infusion, develop powerful anti-tumor effects but are often accompanied by side effects and their efficacy remains sometimes to be improved. It therefore appears important to provide a flexible and easily reversible gene expression regulation system to control T cells activity. We developed a gene expression regulation technology that exploits the physiological GCN2-ATF4 pathway's ability to induce gene expression in T cells in response to one essential amino acid deficiency. We first demonstrated the functionality of NUTRIREG in human T cells by transient expression of reporter genes. We then validated that NUTRIREG can be used in human T cells to transiently express a therapeutic gene such as IL-10. Overall, our results represent a solid basis for the promising use of NUTRIREG to regulate transgene expression in human T cells in a reversible way, and more generally for numerous preventive or curative therapeutic possibilities in cellular immunotherapy strategies.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Enfermedad Injerto contra Huésped/prevención & control , Trasplante Homólogo , Aminoácidos , Alelos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfocitos T , Transgenes
2.
Int J Mol Sci ; 23(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35806203

RESUMEN

Chronic treatment with acetaminophen (APAP) induces cysteine (Cys) and glutathione (GSH) deficiency which leads to adverse metabolic effects including muscle atrophy. Mammalian cells respond to essential amino acid deprivation through the phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α). Phosphorylated eIF2α leads to the recruitment of activating transcription factor 4 (ATF4) to specific CCAAT/enhancer-binding protein-ATF response element (CARE) located in the promoters of target genes. Our purpose was to study the activation of the eIF2α-ATF4 pathway in response to APAP-induced Cys deficiency, as well as the potential contribution of the eIF2α kinase GCN2 and the effect of dietary supplementation with Cys. Our results showed that chronic treatment with APAP activated both GCN2 and PERK eIF2α kinases and downstream target genes in the liver. Activation of the eIF2α-ATF4 pathway in skeletal muscle was accompanied by muscle atrophy even in the absence of GCN2. The dietary supplementation with cysteine reversed APAP-induced decreases in plasma-free Cys, liver GSH, muscle mass, and muscle GSH. Our new findings demonstrate that dietary Cys supplementation also reversed the APAP-induced activation of GCN2 and PERK and downstream ATF4-target genes in the liver.


Asunto(s)
Factor de Transcripción Activador 4 , Factor 2 Eucariótico de Iniciación , Acetaminofén/efectos adversos , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Cisteína/metabolismo , Suplementos Dietéticos , Factor 2 Eucariótico de Iniciación/metabolismo , Glutatión/metabolismo , Mamíferos/metabolismo , Atrofia Muscular/inducido químicamente , Fosforilación , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
3.
Biol Reprod ; 106(3): 463-476, 2022 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-34875016

RESUMEN

Infertility represents a growing burden worldwide, with one in seven couples presenting difficulties conceiving. Among these, 10-15% of the men have idiopathic infertility that does not correlate with any defect in the classical sperm parameters measured. In the present study, we used a mouse model to investigate the effects of maternal undernutrition on fertility in male progeny. Our results indicate that mothers fed on a low-protein diet during gestation and lactation produce male offspring with normal sperm morphology, concentration, and motility but exhibiting an overall decrease of fertility when they reach adulthood. Particularly, in contrast to control, sperm from these offspring show a remarkable lower capacity to fertilize oocytes when copulation occurs early in the estrus cycle relative to ovulation, due to an altered sperm capacitation. Our data demonstrate for the first time that maternal nutritional stress can have long-term consequences on the reproductive health of male progeny by affecting sperm physiology, especially capacitation, with no observable impact on spermatogenesis and classical quantitative and qualitative sperm parameters. Moreover, our experimental model could be of major interest to study, explain, and ultimately treat certain categories of infertilities.


Asunto(s)
Infertilidad Masculina , Desnutrición , Adulto , Animales , Femenino , Fertilidad , Humanos , Infertilidad Masculina/etiología , Lactancia , Masculino , Desnutrición/complicaciones , Ratones , Embarazo , Capacitación Espermática , Motilidad Espermática , Espermatozoides/fisiología
4.
Int J Mol Sci ; 24(1)2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36614063

RESUMEN

Activating transcription factor 4 (ATF4) is involved in muscle atrophy through the overexpression of some atrogenes. However, it also controls the transcription of genes involved in muscle homeostasis maintenance. Here, we explored the effect of ATF4 activation by the pharmacological molecule halofuginone during hindlimb suspension (HS)-induced muscle atrophy. Firstly, we reported that periodic activation of ATF4-regulated atrogenes (Gadd45a, Cdkn1a, and Eif4ebp1) by halofuginone was not associated with muscle atrophy in healthy mice. Secondly, halofuginone-treated mice even showed reduced atrophy during HS, although the induction of the ATF4 pathway was identical to that in untreated HS mice. We further showed that halofuginone inhibited transforming growth factor-ß (TGF-ß) signalling, while promoting bone morphogenetic protein (BMP) signalling in healthy mice and slightly preserved protein synthesis during HS. Finally, ATF4-regulated atrogenes were also induced in the atrophy-resistant muscles of hibernating brown bears, in which we previously also reported concurrent TGF-ß inhibition and BMP activation. Overall, we show that ATF4-induced atrogenes can be uncoupled from muscle atrophy. In addition, our data also indicate that halofuginone can control the TGF-ß/BMP balance towards muscle mass maintenance. Whether halofuginone-induced BMP signalling can counteract the effect of ATF4-induced atrogenes needs to be further investigated and may open a new avenue to fight muscle atrophy. Finally, our study opens the way for further studies to identify well-tolerated chemical compounds in humans that are able to fine-tune the TGF-ß/BMP balance and could be used to preserve muscle mass during catabolic situations.


Asunto(s)
Factor de Transcripción Activador 4 , Atrofia Muscular , Ursidae , Animales , Ratones , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Hibernación
6.
Sci Rep ; 8(1): 17939, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30560874

RESUMEN

Endoplasmic Reticulum (ER) stress of alveolar epithelial cells (AECs) is recognized as a key event of cell dysfunction in pulmonary fibrosis (PF). However, the mechanisms leading to AECs ER stress and ensuing unfolded protein response (UPR) pathways in idiopathic PF (IPF) remain unclear. We hypothesized that alveolar hypoxic microenvironment would generate ER stress and AECs apoptosis through the hypoxia-inducible factor-1α (HIF-1α). Combining ex vivo, in vivo and in vitro experiments, we investigated the effects of hypoxia on the UPR pathways and ER stress-mediated apoptosis, and consecutively the mechanisms linking hypoxia, HIF-1α, UPR and apoptosis. HIF-1α and the pro-apoptotic ER stress marker C/EBP homologous protein (CHOP) were co-expressed in hyperplastic AECs from bleomycin-treated mice and IPF lungs, not in controls. Hypoxic exposure of rat lungs or primary rat AECs induced HIF-1α, CHOP and apoptosis markers expression. In primary AECs, hypoxia activated UPR pathways. Pharmacological ER stress inhibitors and pharmacological inhibition or silencing of HIF-1α both prevented hypoxia-induced upregulation of CHOP and apoptosis. Interestingly, overexpression of HIF-1α in normoxic AECs increased UPR pathways transcription factors activities, and CHOP expression. These results indicate that hypoxia and HIF-1α can trigger ER stress and CHOP-mediated apoptosis in AECs, suggesting their potential contribution to the development of IPF.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Factor de Transcripción CHOP/metabolismo , Anciano , Células Epiteliales Alveolares/patología , Animales , Apoptosis/genética , Biopsia , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , Persona de Mediana Edad , Ratas , Factor de Transcripción CHOP/genética , Respuesta de Proteína Desplegada
7.
Cell Death Differ ; 24(9): 1518-1529, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28644439

RESUMEN

The unfolded protein response (UPR) is an endoplasmic reticulum (ER) -related stress conserved pathway that aims to protect cells from being overwhelmed. However, when prolonged, UPR activation converts to a death signal, which relies on its PERK-eIF2α branch. Overactivation of the UPR has been implicated in many neurological diseases, including cerebral ischaemia. Here, by using an in vivo thromboembolic model of stroke on transgenic ER stress-reporter mice and neuronal in vitro models of ischaemia, we demonstrate that ischaemic stress leads to the deleterious activation of the PERK branch of the UPR. Moreover, we show that the serine protease tissue-type plasminogen activator (tPA) can bind to cell surface Grp78 (78 kD glucose-regulated protein), leading to a decrease of the PERK pathway activation, thus a decrease of the deleterious factor CHOP, and finally promotes neuroprotection. Altogether, this work highlights a new role and a therapeutic potential of the chaperone protein Grp78 as a membrane receptor of tPA capable to prevent from ER stress overactivation.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Fibrinolíticos/farmacología , Ratones , Neuronas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Tromboembolia/terapia , Activador de Tejido Plasminógeno/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
8.
Oncotarget ; 8(16): 27440-27453, 2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28460466

RESUMEN

The uncontrolled growth of tumor can lead to the formation of area deprived in nutrients. Due to their high genetic instability, tumor cells can adapt and develop resistance to this pro-apoptotic environment. Among the resistance mechanisms, those involved in the resistance to long-term amino acid restriction are not elucidated. A long-term amino acid restriction is particularly deleterious since nine of them cannot be synthetized by the cells. In order to determine how cancer cells face a long-term amino acid deprivation, we developed a cell model selected for its capacity to resist a long-term amino acid limitation. We exerted a selection pressure on mouse embryonic fibroblast to isolate clones able to survive with low amino acid concentration. The study of several clones revealed an alteration of the eiF2α/ATF4 pathway. Compared to the parental cells, the clones exhibited a decreased expression of the transcription factor ATF4 and its target genes. Likewise, the knock-down of ATF4 in parental cells renders them resistant to amino acid deprivation. Moreover, this association between a low level of ATF4 protein and the resistance to amino acid deprivation was also observed in the cancer cell line BxPC-3. This resistance was abolished when ATF4 was overexpressed. Therefore, decreasing ATF4 expression may be one important mechanism for cancer cells to survive under prolonged amino acid deprivation.


Asunto(s)
Factor de Transcripción Activador 4/genética , Aminoácidos/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Modelos Biológicos , Neoplasias/genética , Neoplasias/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal
9.
Curr Opin Clin Nutr Metab Care ; 20(3): 175-180, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28376508

RESUMEN

PURPOSE OF REVIEW: The article highlights the recent development of an ATF4 (activating transcription factor) inducible luciferase (LUC) mouse model to monitor the integrated stress response pathway (ISR) in vivo. RECENT FINDING: The ISR pathway plays a key role in cellular adaptation to stress and is dysregulated in numerous diseases. The core event in this pathway is the phosphorylation of eukaryotic translation initiation factor 2 α, which leads to the recruitment of the transcription factor ATF4 to specific CCAAT/enhancer-binding protein-ATF response elements (CAREs) located in the promoters of target genes. To monitor the modulation of this pathway in the whole animal and at tissue and cellular levels, we generated a CARE-driven LUC mouse model. We validated the relevance of this model to study stress-related pathologies and recently observed the correlation between the ISR pathway induction in muscle and the occurrence of stress-induced skeletal muscle atrophy. SUMMARY: The CARE-LUC mouse model represents an innovative tool for investigating the role of the ISR pathway in physiology and disease and opens new avenues for the development of drugs that could modify this important pathway in stress-related human diseases.


Asunto(s)
Factor de Transcripción Activador 4/fisiología , Luciferasas/fisiología , Modelos Animales , Transducción de Señal/fisiología , Estrés Fisiológico/fisiología , Animales , Ratones , Músculo Esquelético/metabolismo , Atrofia Muscular/fisiopatología , Fosforilación/fisiología , Regiones Promotoras Genéticas/fisiología , Proteínas Serina-Treonina Quinasas
11.
Sci Rep ; 6: 27278, 2016 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-27255611

RESUMEN

The hexosamine biosynthetic pathway (HBP) is a nutrient-sensing metabolic pathway that produces the activated amino sugar UDP-N-acetylglucosamine, a critical substrate for protein glycosylation. Despite its biological significance, little is known about the regulation of HBP flux during nutrient limitation. Here, we report that amino acid or glucose shortage increase GFAT1 production, the first and rate-limiting enzyme of the HBP. GFAT1 is a transcriptional target of the activating transcription factor 4 (ATF4) induced by the GCN2-eIF2α signalling pathway. The increased production of GFAT1 stimulates HBP flux and results in an increase in O-linked ß-N-acetylglucosamine protein modifications. Taken together, these findings demonstrate that ATF4 provides a link between nutritional stress and the HBP for the regulation of the O-GlcNAcylation-dependent cellular signalling.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Aminoácidos/metabolismo , Glucosa/metabolismo , Hexosaminas/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Acetilglucosamina/metabolismo , Animales , Vías Biosintéticas , Línea Celular , Células HeLa , Humanos , Ratones , Transferasas de Grupos Nitrogenados/metabolismo , Ratas , Transducción de Señal
12.
Sci Rep ; 6: 27698, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27297692

RESUMEN

It is well known that the GCN2 and mTORC1 signaling pathways are regulated by amino acids and share common functions, in particular the control of translation. The regulation of GCN2 activity by amino acid availability relies on the capacity of GCN2 to sense the increased levels of uncharged tRNAs upon amino acid scarcity. In contrast, despite recent progress in the understanding of the regulation of mTORC1 by amino acids, key aspects of this process remain unsolved. In particular, while leucine is well known to be a potent regulator of mTORC1, the mechanisms by which this amino acid is sensed and control mTORC1 activity are not well defined. Our data establish that GCN2 is involved in the inhibition of mTORC1 upon leucine or arginine deprivation. However, the activation of GCN2 alone is not sufficient to inhibit mTORC1 activity, indicating that leucine and arginine exert regulation via additional mechanisms. While the mechanism by which GCN2 contributes to the initial step of mTORC1 inhibition involves the phosphorylation of eIF2α, we show that it is independent of the downstream transcription factor ATF4. These data point to a novel role for GCN2 and phosphorylation of eIF2α in the control of mTORC1 by certain amino acids.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Leucina/deficiencia , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Arginina/deficiencia , Embrión de Mamíferos/citología , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Fosforilación , Transducción de Señal
13.
Nat Biotechnol ; 34(7): 746-51, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27272383

RESUMEN

Widespread application of gene therapy will depend on the development of simple methods to regulate the expression of therapeutic genes. Here we harness an endogenous signaling pathway to regulate therapeutic gene expression through diet. The GCN2-eIF2α signaling pathway is specifically activated by deficiencies in any essential amino acid (EAA); EAA deficiency leads to rapid expression of genes regulated by ATF4-binding cis elements. We found that therapeutic genes under the control of optimized amino acid response elements (AAREs) had low basal expression and high induced expression. We applied our system to regulate the expression of TNFSF10 (TRAIL) in the context of glioma therapy and found that intermittent activation of this gene by EEA-deficient meals retained its therapeutic efficacy while abrogating its toxic effects on normal tissue. The GCN2-eIF2α pathway is expressed in many tissues, including the brain, and is highly specific to EAA deficiency. Our system may be particularly well suited for intermittent regulation of therapeutic transgenes over short or long time periods.


Asunto(s)
Aminoácidos Esenciales/administración & dosificación , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Administración Oral , Aminoácidos Esenciales/farmacocinética , Animales , Suplementos Dietéticos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Masculino , Ratones , Transgenes/genética , Resultado del Tratamiento
14.
Autophagy ; 12(5): 770-83, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-26986695

RESUMEN

The intestinal mucosa of Crohn disease (CD) patients is abnormally colonized by adherent-invasive E. coli (AIEC). Upon AIEC infection, autophagy is induced in host cells to restrain bacterial intracellular replication. The underlying mechanism, however, remains unknown. Here, we investigated the role of the EIF2AK4-EIF2A/eIF2α-ATF4 pathway in the autophagic response to AIEC infection. We showed that infection of human intestinal epithelial T84 cells with the AIEC reference strain LF82 activated the EIF2AK4-EIF2A-ATF4 pathway, as evidenced by increased phospho-EIF2AK4, phospho-EIF2A and ATF4 levels. EIF2AK4 depletion inhibited autophagy activation in response to LF82 infection, leading to increased LF82 intracellular replication and elevated pro-inflammatory cytokine production. Mechanistically, EIF2AK4 depletion suppressed the LF82-induced ATF4 binding to promoters of several autophagy genes including MAP1LC3B, BECN1, SQSTM1, ATG3 and ATG7, and this subsequently inhibited transcription of these genes. LF82 infection of wild-type (WT), but not eif2ak4(-/-), mice activated the EIF2AK4-EIF2A-ATF4 pathway, inducing autophagy gene transcription and autophagy response in enterocytes. Consequently, eif2ak4(-/-) mice exhibited increased intestinal colonization by LF82 bacteria and aggravated inflammation compared to WT mice. Activation of the EIF2AK4-EIF2A-ATF4 pathway was observed in ileal biopsies from patients with noninflamed CD, and this was suppressed in inflamed CD, suggesting that a defect in the activation of this pathway could be one of the mechanisms contributing to active disease. In conclusion, we show that activation of the EIF2AK4-EIF2A-ATF4 pathway upon AIEC infection serves as a host defense mechanism to induce functional autophagy to control AIEC intracellular replication.


Asunto(s)
Autofagia/fisiología , Enfermedad de Crohn/microbiología , Infecciones por Escherichia coli/metabolismo , Mucosa Intestinal/microbiología , Transducción de Señal , Factor de Transcripción Activador 4/metabolismo , Adolescente , Adulto , Enfermedad de Crohn/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Intestinos/patología , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Adulto Joven
15.
Biotechniques ; 60(1): 47-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26757812

RESUMEN

It has been reported that breast-feeding more than 6 months strongly decreases the risk of allergy, diabetes, obesity, and hypertension in humans. In order to understand the mechanisms responsible for this benefit, it is important to evaluate precisely the composition of maternal milk, especially in response to environmental cues. Mouse models offer a unique opportunity to study the impact of maternal milk composition on the development and health of offspring. Oxytocin injection of the dam is usually used to stimulate milk ejection; however, exogenous oxytocin might have deleterious effects under some experimental conditions by modifying milk content as well as the physiology and behavior of the dam. Taking advantage of the natural stimulation of the mammary gland that occurs after the reunion of a dam that has been separated from her pups, we developed a new procedure to collect mouse milk without the injection of oxytocin. This method is easy to use, low-cost ,and non-invasive. Moreover, it provides a sufficient amount of milk for use in a wide range of biological analyses.


Asunto(s)
Lactancia Materna , Glándulas Mamarias Animales/fisiología , Eyección Láctea/fisiología , Leche , Animales , Mama/metabolismo , Mama/fisiología , Femenino , Humanos , Ratones , Oxitocina/farmacología
17.
Sci Signal ; 8(374): rs5, 2015 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-25921292

RESUMEN

The eIF2α-ATF4 pathway is involved in cellular adaptation to stress and is dysregulated in numerous diseases. Activation of this pathway leads to phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α) and the recruitment of the transcription factor ATF4 (activating transcription factor 4) to specific CCAAT/enhancer binding protein (C/EBP)-ATF response elements (CAREs) located in the promoters of target genes. To monitor the spatiotemporal modulation of this pathway in living animals, we generated a novel CARE-driven luciferase mouse model (CARE-LUC). These transgenic mice enable the investigation of the eIF2α-ATF4 pathway activity in the whole organism and at the tissue and cellular levels by combining imaging, luciferase assays, and immunochemistry. Using this mouse line, we showed the tissue-specific activation pattern of this pathway in response to amino acid deficiency or endoplasmic reticulum stress and the hepatic induction of this pathway in a stress-related pathology model of liver fibrosis. The CARE-LUC mouse model represents an innovative tool to investigate the eIF2α-ATF4 axis and to develop drugs targeting this important pathway in the remediation of related pathologies.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Imagen Molecular , Transducción de Señal , Estrés Fisiológico , Factor de Transcripción Activador 4/genética , Animales , Factor 2 Eucariótico de Iniciación/genética , Ratones , Ratones Transgénicos
18.
Biol Aujourdhui ; 209(4): 317-23, 2015.
Artículo en Francés | MEDLINE | ID: mdl-27021050

RESUMEN

In mammals, metabolic adaptations are required to overcome nutritional deprivation in amino-acids/proteins as well as episodes of malnutrition. GCN2 protein kinase, which phosphorylates the α subunit of the translation initiation factor eIF2, is a sensor of amino-acid(s) deficiencies. On one hand, this review briefly describes the main features of amino-acid metabolism. On the other hand, it describes the role of GCN2 in regulating numerous physiological functions.


Asunto(s)
Adaptación Biológica/fisiología , Aminoácidos Esenciales/administración & dosificación , Dieta , Proteínas Serina-Treonina Quinasas/fisiología , Aminoácidos , Aminoácidos Esenciales/deficiencia , Animales , Factor 2 Eucariótico de Iniciación/metabolismo , Homeostasis , Humanos , Inmunidad , Desnutrición , Plasticidad Neuronal , Estado Nutricional , Fosforilación , Transducción de Señal/fisiología
19.
PLoS One ; 9(8): e104896, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25118945

RESUMEN

Epidemiological findings indicate that transient environmental influences during perinatal life, especially nutrition, may have deleterious heritable health effects lasting for the entire life. Indeed, the fetal organism develops specific adaptations that permanently change its physiology/metabolism and that persist even in the absence of the stimulus that initiated them. This process is termed "nutritional programming". We previously demonstrated that mothers fed a Low-Protein-Diet (LPD) during gestation and lactation give birth to F1-LPD animals presenting metabolic consequences that are different from those observed when the nutritional stress is applied during gestation only. Compared to control mice, adult F1-LPD animals have a lower body weight and exhibit a higher food intake suggesting that maternal protein under-nutrition during gestation and lactation affects the energy metabolism of F1-LPD offspring. In this study, we investigated the origin of this apparent energy wasting process in F1-LPD and demonstrated that minimal energy expenditure is increased, due to both an increased mitochondrial function in skeletal muscle and an increased mitochondrial density in White Adipose Tissue. Importantly, F1-LPD mice are protected against high-fat-diet-induced obesity. Clearly, different paradigms of exposure to malnutrition may be associated with differences in energy expenditure, food intake, weight and different susceptibilities to various symptoms associated with metabolic syndrome. Taken together these results demonstrate that intra-uterine environment is a major contributor to the future of individuals and disturbance at a critical period of development may compromise their health. Consequently, understanding the molecular mechanisms may give access to useful knowledge regarding the onset of metabolic diseases.


Asunto(s)
Metabolismo Energético/genética , Lactancia/fisiología , Mitocondrias/fisiología , Obesidad/prevención & control , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Deficiencia de Proteína/fisiopatología , Tejido Adiposo Blanco/metabolismo , Factores de Edad , Animales , Western Blotting , Temperatura Corporal , Peso Corporal , Calorimetría Indirecta , Cartilla de ADN/genética , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Femenino , Ratones , Ratones Endogámicos BALB C , Obesidad/metabolismo , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cell Signal ; 26(9): 1918-27, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24793303

RESUMEN

The mammalian target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth and metabolism. It controls many cell functions by integrating nutrient availability and growth factor signals. Amino acids, and in particular leucine, are among the main positive regulators of mTORC1 signaling. The current model for the regulation of mTORC1 by amino acids involves the movement of mTOR to the lysosome mediated by the Rag-GTPases. Here, we have examined the control of mTORC1 signaling and mTOR localization by amino acids and leucine in serum-fed cells, because both serum growth factors (or, e.g., insulin) and amino acids are required for full activation of mTORC1 signaling. We demonstrate that mTORC1 activity does not closely correlate with the lysosomal localization of mTOR. In particular, leucine controls mTORC1 activity without any detectable modification of the lysosomal localization of mTOR, indicating that the signal(s) exerted by leucine is likely distinct from those exerted by other amino acids. In addition, knock-down of the Rag-GTPases attenuated the inhibitory effect of amino acid- or leucine-starvation on the phosphorylation of mTORC1 targets. Furthermore, data from cells where Rag expression has been knocked down revealed that leucine can promote mTORC1 signaling independently of the lysosomal localization of mTOR. Our data complement existing models for the regulation of mTORC1 by amino acids and provide new insights into this important topic.


Asunto(s)
Aminoácidos/metabolismo , Leucina/metabolismo , Lisosomas/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/farmacología , Animales , Línea Celular , Leucina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Proteínas de Unión al GTP Monoméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...