Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cell ; 186(24): 5328-5346.e26, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37883971

ABSTRACT

Lysosomes serve dual antagonistic functions in cells by mediating anabolic growth signaling and the catabolic turnover of macromolecules. How these janus-faced activities are regulated in response to cellular nutrient status is poorly understood. We show here that lysosome morphology and function are reversibly controlled by a nutrient-regulated signaling lipid switch that triggers the conversion between peripheral motile mTOR complex 1 (mTORC1) signaling-active and static mTORC1-inactive degradative lysosomes clustered at the cell center. Starvation-triggered relocalization of phosphatidylinositol 4-phosphate (PI(4)P)-metabolizing enzymes reshapes the lysosomal surface proteome to facilitate lysosomal proteolysis and to repress mTORC1 signaling. Concomitantly, lysosomal phosphatidylinositol 3-phosphate (PI(3)P), which marks motile signaling-active lysosomes in the cell periphery, is erased. Interference with this PI(3)P/PI(4)P lipid switch module impairs the adaptive response of cells to altering nutrient supply. Our data unravel a key function for lysosomal phosphoinositide metabolism in rewiring organellar membrane dynamics in response to cellular nutrient status.


Subject(s)
Lysosomes , Signal Transduction , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nutrients , Cell Physiological Phenomena
2.
PLoS Biol ; 21(4): e3002044, 2023 04.
Article in English | MEDLINE | ID: mdl-37068088

ABSTRACT

Unlike immature neurons and the ones from the peripheral nervous system (PNS), mature neurons from the central nervous system (CNS) cannot regenerate after injury. In the past 15 years, tremendous progress has been made to identify molecules and pathways necessary for neuroprotection and/or axon regeneration after CNS injury. In most regenerative models, phosphorylated ribosomal protein S6 (p-RPS6) is up-regulated in neurons, which is often associated with an activation of the mTOR (mammalian target of rapamycin) pathway. However, the exact contribution of posttranslational modifications of this ribosomal protein in CNS regeneration remains elusive. In this study, we demonstrate that RPS6 phosphorylation is essential for PNS and CNS regeneration in mice. We show that this phosphorylation is induced during the preconditioning effect in dorsal root ganglion (DRG) neurons and that it is controlled by the p90S6 kinase RSK2. Our results reveal that RSK2 controls the preconditioning effect and that the RSK2-RPS6 axis is key for this process, as well as for PNS regeneration. Finally, we demonstrate that RSK2 promotes CNS regeneration in the dorsal column, spinal cord synaptic plasticity, and target innervation leading to functional recovery. Our data establish the critical role of RPS6 phosphorylation controlled by RSK2 in CNS regeneration and give new insights into the mechanisms related to axon growth and circuit formation after traumatic lesion.


Subject(s)
Axons , Nerve Regeneration , Ribosomal Protein S6 Kinases, 90-kDa , Animals , Mice , Axons/metabolism , Central Nervous System , Nerve Regeneration/physiology , Neurons/physiology , Spinal Cord
3.
EMBO J ; 38(1)2019 01 03.
Article in English | MEDLINE | ID: mdl-30420558

ABSTRACT

As a consequence of impaired glucose or fatty acid metabolism, bioenergetic stress in skeletal muscles may trigger myopathy and rhabdomyolysis. Genetic mutations causing loss of function of the LPIN1 gene frequently lead to severe rhabdomyolysis bouts in children, though the metabolic alterations and possible therapeutic interventions remain elusive. Here, we show that lipin1 deficiency in mouse skeletal muscles is sufficient to trigger myopathy. Strikingly, muscle fibers display strong accumulation of both neutral and phospholipids. The metabolic lipid imbalance can be traced to an altered fatty acid synthesis and fatty acid oxidation, accompanied by a defect in acyl chain elongation and desaturation. As an underlying cause, we reveal a severe sarcoplasmic reticulum (SR) stress, leading to the activation of the lipogenic SREBP1c/SREBP2 factors, the accumulation of the Fgf21 cytokine, and alterations of SR-mitochondria morphology. Importantly, pharmacological treatments with the chaperone TUDCA and the fatty acid oxidation activator bezafibrate improve muscle histology and strength of lipin1 mutants. Our data reveal that SR stress and alterations in SR-mitochondria contacts are contributing factors and potential intervention targets of the myopathy associated with lipin1 deficiency.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Muscular Diseases/genetics , Phosphatidate Phosphatase/genetics , Sarcoplasmic Reticulum/metabolism , Taurochenodeoxycholic Acid/pharmacology , Animals , Endoplasmic Reticulum Stress/drug effects , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Transgenic , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Molecular Chaperones/pharmacology , Molecular Chaperones/therapeutic use , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/drug therapy , Muscular Diseases/metabolism , Muscular Diseases/pathology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/pathology , Taurochenodeoxycholic Acid/therapeutic use
4.
EMBO J ; 36(6): 736-750, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28242756

ABSTRACT

The inactivation of S6 kinases mimics several aspects of caloric restriction, including small body size, increased insulin sensitivity and longevity. However, the impact of S6 kinase activity on cellular senescence remains to be established. Here, we show that the constitutive activation of mammalian target of rapamycin complex 1 (mTORC1) by tuberous sclerosis complex (TSC) mutations induces a premature senescence programme in fibroblasts that relies on S6 kinases. To determine novel molecular targets linking S6 kinase activation to the control of senescence, we set up a chemical genetic screen, leading to the identification of the nuclear epigenetic factor ZRF1 (also known as DNAJC2, MIDA1, Mpp11). S6 kinases phosphorylate ZRF1 on Ser47 in cultured cells and in mammalian tissues in vivo Knock-down of ZRF1 or expression of a phosphorylation mutant is sufficient to blunt the S6 kinase-dependent senescence programme. This is traced by a sharp alteration in p16 levels, the cell cycle inhibitor and a master regulator of senescence. Our findings reveal a mechanism by which nutrient sensing pathways impact on cell senescence through the activation of mTORC1-S6 kinases and the phosphorylation of ZRF1.


Subject(s)
Aging , HSP40 Heat-Shock Proteins/metabolism , Protein Processing, Post-Translational , Ribosomal Protein S6 Kinases/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins , Mice , Molecular Chaperones , Phosphorylation , RNA-Binding Proteins
5.
Nature ; 517(7535): 497-500, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25383520

ABSTRACT

Inactivation of APC is a strongly predisposing event in the development of colorectal cancer, prompting the search for vulnerabilities specific to cells that have lost APC function. Signalling through the mTOR pathway is known to be required for epithelial cell proliferation and tumour growth, and the current paradigm suggests that a critical function of mTOR activity is to upregulate translational initiation through phosphorylation of 4EBP1 (refs 6, 7). This model predicts that the mTOR inhibitor rapamycin, which does not efficiently inhibit 4EBP1 (ref. 8), would be ineffective in limiting cancer progression in APC-deficient lesions. Here we show in mice that mTOR complex 1 (mTORC1) activity is absolutely required for the proliferation of Apc-deficient (but not wild-type) enterocytes, revealing an unexpected opportunity for therapeutic intervention. Although APC-deficient cells show the expected increases in protein synthesis, our study reveals that it is translation elongation, and not initiation, which is the rate-limiting component. Mechanistically, mTORC1-mediated inhibition of eEF2 kinase is required for the proliferation of APC-deficient cells. Importantly, treatment of established APC-deficient adenomas with rapamycin (which can target eEF2 through the mTORC1-S6K-eEF2K axis) causes tumour cells to undergo growth arrest and differentiation. Taken together, our data suggest that inhibition of translation elongation using existing, clinically approved drugs, such as the rapalogs, would provide clear therapeutic benefit for patients at high risk of developing colorectal cancer.


Subject(s)
Cell Transformation, Neoplastic/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Multiprotein Complexes/metabolism , Peptide Chain Elongation, Translational , TOR Serine-Threonine Kinases/metabolism , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Elongation Factor 2 Kinase/deficiency , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Enzyme Activation , Genes, APC , Intestinal Neoplasms/genetics , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Oncogene Protein p55(v-myc)/metabolism , Peptide Elongation Factor 2/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Wnt Proteins/metabolism
6.
Am J Respir Cell Mol Biol ; 55(3): 352-67, 2016 09.
Article in English | MEDLINE | ID: mdl-26991739

ABSTRACT

Constitutive activation of the mammalian target of rapamycin (mTOR) complexes mTORC1 and mTORC2 is associated with pulmonary hypertension (PH) and sustained growth of pulmonary artery (PA) smooth muscle cells (SMCs). We investigated whether selective mTORC1 activation in SMCs induced by deleting the negative mTORC1 regulator tuberous sclerosis complex 1 gene (TSC1) was sufficient to produce PH in mice. Mice expressing Cre recombinase under SM22 promoter control were crossed with TSC1(LoxP/LoxP) mice to generate SM22-TSC1(-/-) mice. At 8 weeks of age, SM22-TSC1(-/-) mice exhibited PH with marked increases in distal PA muscularization and Ki67-positive PASMC counts, without systemic hypertension or cardiac dysfunction. Marked activation of the mTORC1 substrates S6 kinase and 4E-BP and the mTORC2 substrates p-Akt(Ser473) and glycogen synthase kinase 3 was found in the lungs and pulmonary vessels of SM22-TSC1(-/-) mice when compared with control mice. Treatment with 5 mg/kg rapamycin for 3 weeks to inhibit mTORC1 and mTORC2 fully reversed PH in SM22-TSC1(-/-) mice. In chronically hypoxic mice and SM22-5HTT(+) mice exhibiting PH associated with mTORC1 and mTORC2 activation, PH was maximally attenuated by low-dose rapamycin associated with selective mTORC1 inhibition. Cultured PASMCs from SM22-TSC1(-/-), SM22-5HTT(+), and chronically hypoxic mice exhibited similar sustained growth-rate enhancement and constitutive mTORC1 and mTORC2 activation; both effects were abolished by rapamycin. Deletion of the downstream mTORC1 effectors S6 kinase 1/2 in mice also activated mTOR signaling and induced PH. We concluded that activation of mTORC1 signaling leads to increased PASMC proliferation and subsequent PH development.


Subject(s)
Gene Deletion , Hypertension, Pulmonary/metabolism , Muscle, Smooth/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chronic Disease , Hyperplasia , Hypertension, Pulmonary/diagnostic imaging , Hypoxia/complications , Hypoxia/metabolism , Hypoxia/pathology , Lung/blood supply , Lung/pathology , Male , Metformin/pharmacology , Mice , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/pathology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Tuberous Sclerosis Complex 1 Protein
7.
Neurobiol Dis ; 89: 180-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26873552

ABSTRACT

DEP-domain containing 5 (DEPDC5), encoding a repressor of the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway, has recently emerged as a major gene mutated in familial focal epilepsies and focal cortical dysplasia. Here we established a global knockout rat using TALEN technology to investigate in vivo the impact of Depdc5-deficiency. Homozygous Depdc5(-/-) embryos died from embryonic day 14.5 due to a global growth delay. Constitutive mTORC1 hyperactivation was evidenced in the brains and in cultured fibroblasts of Depdc5(-/-) embryos, as reflected by enhanced phosphorylation of its downstream effectors S6K1 and rpS6. Consistently, prenatal treatment with mTORC1 inhibitor rapamycin rescued the phenotype of Depdc5(-/-) embryos. Heterozygous Depdc5(+/-) rats developed normally and exhibited no spontaneous electroclinical seizures, but had altered cortical neuron excitability and firing patterns. Depdc5(+/-) rats displayed cortical cytomegalic dysmorphic neurons and balloon-like cells strongly expressing phosphorylated rpS6, indicative of mTORC1 upregulation, and not observed after prenatal rapamycin treatment. These neuropathological abnormalities are reminiscent of the hallmark brain pathology of human focal cortical dysplasia. Altogether, Depdc5 knockout rats exhibit multiple features of rodent models of mTORopathies, and thus, stand as a relevant model to study their underlying pathogenic mechanisms.


Subject(s)
Cerebral Cortex/abnormalities , Disease Models, Animal , Embryonic Development/genetics , Multiprotein Complexes/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Animals, Genetically Modified , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Embryonic Development/drug effects , Fibroblasts/metabolism , Gene Knockout Techniques , Genotype , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/antagonists & inhibitors , Neurons/pathology , Neurons/physiology , Phosphorylation , Rats , Rats, Inbred F344 , Rats, Wistar , Repressor Proteins/metabolism , Signal Transduction/drug effects , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors
8.
Biochim Biophys Acta ; 1832(12): 2103-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23928362

ABSTRACT

Lipin-1 deficiency is associated with massive rhabdomyolysis episodes in humans, precipitated by febrile illnesses. Despite well-known roles of lipin-1 in lipid biosynthesis and transcriptional regulation, the pathogenic mechanisms leading to rhabdomyolysis remain unknown. Here we show that primary myoblasts from lipin-1-deficient patients exhibit a dramatic decrease in LPIN1 expression and phosphatidic acid phosphatase 1 activity, and a significant accumulation of lipid droplets (LD). The expression levels of LPIN1-target genes [peroxisome proliferator-activated receptors delta and alpha (PPARδ, PPARα), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), acyl-coenzyme A dehydrogenase, very long (ACADVL), carnitine palmitoyltransferase IB and 2 (CPT1B and CPT2)] were not affected while lipin-2 protein level, a closely related member of the family, was increased. Microarray analysis of patients' myotubes identified 19 down-regulated and 51 up-regulated genes, indicating pleiotropic effects of lipin-1 deficiency. Special attention was paid to the up-regulated ACACB (acetyl-CoA carboxylase beta), a key enzyme in the fatty acid synthesis/oxidation balance. We demonstrated that overexpression of ACACB was associated with free fatty acid accumulation in patients' myoblasts whereas malonyl-carnitine (as a measure of malonyl-CoA) and CPT1 activity were in the normal range in basal conditions accordingly to the normal daily activity reported by the patients. Remarkably ACACB invalidation in patients' myoblasts decreased LD number and size while LPIN1 invalidation in controls induced LD accumulation. Further, pro-inflammatory treatments tumor necrosis factor alpha+Interleukin-1beta(TNF1α+IL-1ß) designed to mimic febrile illness, resulted in increased malonyl-carnitine levels, reduced CPT1 activity and enhanced LD accumulation, a phenomenon reversed by dexamethasone and TNFα or IL-1ß inhibitors. Our data suggest that the pathogenic mechanism of rhabdomyolysis in lipin-1-deficient patients combines the predisposing constitutive impairment of lipid metabolism and its exacerbation by pro-inflammatory cytokines.


Subject(s)
Cytokines/pharmacology , Inflammation Mediators/pharmacology , Lipid Metabolism Disorders/etiology , Lipids , Muscle Fibers, Skeletal/pathology , Myoblasts/pathology , Phosphatidate Phosphatase/genetics , Biomarkers/metabolism , Blotting, Western , Case-Control Studies , Cell Cycle , Cell Proliferation , Child , Child, Preschool , Endoplasmic Reticulum Stress , Female , Gene Expression Profiling , Humans , Lipid Metabolism Disorders/metabolism , Lipid Metabolism Disorders/pathology , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation/genetics , Myoblasts/drug effects , Myoblasts/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatitis-Associated Proteins , Phosphatidate Phosphatase/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyolysis/etiology , Rhabdomyolysis/metabolism , Rhabdomyolysis/pathology
9.
Nat Metab ; 6(2): 323-342, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38409325

ABSTRACT

Cellular senescence affects many physiological and pathological processes and is characterized by durable cell cycle arrest, an inflammatory secretory phenotype and metabolic reprogramming. Here, by using dynamic transcriptome and metabolome profiling in human fibroblasts with different subtypes of senescence, we show that a homoeostatic switch that results in glycerol-3-phosphate (G3P) and phosphoethanolamine (pEtN) accumulation links lipid metabolism to the senescence gene expression programme. Mechanistically, p53-dependent glycerol kinase activation and post-translational inactivation of phosphate cytidylyltransferase 2, ethanolamine regulate this metabolic switch, which promotes triglyceride accumulation in lipid droplets and induces the senescence gene expression programme. Conversely, G3P phosphatase and ethanolamine-phosphate phospho-lyase-based scavenging of G3P and pEtN acts in a senomorphic way by reducing G3P and pEtN accumulation. Collectively, our study ties G3P and pEtN accumulation to controlling lipid droplet biogenesis and phospholipid flux in senescent cells, providing a potential therapeutic avenue for targeting senescence and related pathophysiology.


Subject(s)
Glycerol , Glycerophosphates , Lipid Metabolism , Humans , Glycerol/metabolism , Ethanolamines , Phosphates
10.
Biochem Soc Trans ; 41(4): 917-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863156

ABSTRACT

To sustain increased growth, rapidly proliferating cells, such as tumour cells, undergo metabolic adaptations. In recent years, the mechanisms of glycolysis activation as a key metabolic adaptation in proliferating cells became the topic of intense research. Although this phenomenon was described more than 50 years ago by Otto Warburg, the molecular mechanisms remained elusive. Only recently, it was demonstrated that the expression of specific glycolytic enzymes, namely PKM2 (pyruvate kinase M2) and HK2 (hexokinase 2), occurs simultaneously with the glycolytic addiction of cancer cells. The PI3K (phosphoinositide 3-kinase)/mTOR [mammalian (or mechanistic) target of rapamycin] signalling pathway is a central signalling hub co-ordinating the growth in response to growth factor signalling and nutrient availability. Not surprisingly, it is found to be activated in the majority of the tumour cells. In the present article, we discuss the requirement of different PI3K/mTOR downstream effectors for the metabolic adaptation in liver cancer cells driven by this signalling pathway. We provide evidence for a selective involvement of the mTOR target Akt2 in tumoral growth. In addition, PTEN (phosphatase and tensin homologue deleted on chromosome 10)-negative human hepatocellular carcinoma cell lines display an up-regulation of PKM2 expression in an Akt2-dependent manner, providing an advantage for cell proliferation and anchorage-independent growth. Our data have implications on the link between the metabolic action of insulin signal transduction and tumorigenesis, identifying Akt2 as a potential therapeutical target in liver malignancies depending on cancer genotype.


Subject(s)
Carrier Proteins/metabolism , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Thyroid Hormones/metabolism , Cell Line, Tumor , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Thyroid Hormone-Binding Proteins
11.
Nat Cell Biol ; 7(3): 286-94, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15723049

ABSTRACT

The mammalian target of rapamycin (mTOR) and Akt proteins regulate various steps of muscle development and growth, but the physiological relevance and the downstream effectors are under investigation. Here we show that S6 kinase 1 (S6K1), a protein kinase activated by nutrients and insulin-like growth factors (IGFs), is essential for the control of muscle cytoplasmic volume by Akt and mTOR. Deletion of S6K1 does not affect myoblast cell proliferation but reduces myoblast size to the same extent as that observed with mTOR inhibition by rapamycin. In the differentiated state, S6K1(-/-) myotubes have a normal number of nuclei but are smaller, and their hypertrophic response to IGF1, nutrients and membrane-targeted Akt is blunted. These growth defects reveal that mTOR requires distinct effectors for the control of muscle cell cycle and size, potentially opening new avenues of therapeutic intervention against neoplasia or muscle atrophy.


Subject(s)
Muscle, Skeletal/metabolism , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Animals , Atrophy , Body Weight , Cell Differentiation , Cell Line , Cells, Cultured , Collagen/pharmacology , Drug Combinations , Gene Deletion , Genetic Vectors , Genotype , Green Fluorescent Proteins/metabolism , Homozygote , Humans , Immunoblotting , Laminin/pharmacology , Mice , Mice, Inbred C57BL , Models, Biological , Muscles/pathology , Plasmids/metabolism , Protein Binding , Proteoglycans/pharmacology , Retroviridae/genetics , Signal Transduction , Somatomedins/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transfection
12.
Front Cell Dev Biol ; 10: 949196, 2022.
Article in English | MEDLINE | ID: mdl-36036012

ABSTRACT

Genetic evidence in living organisms from yeast to plants and animals, including humans, unquestionably identifies the Target Of Rapamycin kinase (TOR or mTOR for mammalian/mechanistic) signal transduction pathway as a master regulator of growth through the control of cell size and cell number. Among the mTOR targets, the activation of p70 S6 kinase 1 (S6K1) is exquisitely sensitive to nutrient availability and rapamycin inhibition. Of note, in vivo analysis of mutant flies and mice reveals that S6K1 predominantly regulates cell size versus cell proliferation. Here we review the putative mechanisms of S6K1 action on cell size by considering the main functional categories of S6K1 targets: substrates involved in nucleic acid and protein synthesis, fat mass accumulation, retrograde control of insulin action, senescence program and cytoskeleton organization. We discuss how S6K1 may be involved in the observed interconnection between cell size, regenerative and ageing responses.

13.
Sci Adv ; 8(49): eade7823, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36490341

ABSTRACT

PIK3CA-related overgrowth syndrome (PROS) is a genetic disorder caused by somatic mosaic gain-of-function mutations of PIK3CA. Clinical presentation of patients is diverse and associated with endocrine disruption. Adipose tissue is frequently involved, but its role in disease development and progression has not been elucidated. Here, we created a mouse model of PIK3CA-related adipose tissue overgrowth that recapitulates patient phenotype. We demonstrate that PIK3CA mutation leads to GLUT4 membrane accumulation with a negative feedback loop on insulin secretion, a burst of liver IGFBP1 synthesis with IGF-1 sequestration, and low circulating levels. Mouse phenotype was mainly driven through AKT2. We also observed that PIK3CA mutation induces metabolic reprogramming with Warburg-like effect and protein and lipid synthesis, hallmarks of cancer cells, in vitro, in vivo, and in patients. We lastly show that alpelisib is efficient at preventing and improving PIK3CA-adipose tissue overgrowth and reversing metabolomic anomalies in both animal models and patients.


Subject(s)
Adipose Tissue , Class I Phosphatidylinositol 3-Kinases , Gain of Function Mutation , Animals , Mice , Adipose Tissue/metabolism , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Gain of Function Mutation/genetics , Mutation , Phenotype
14.
J Clin Invest ; 118(11): 3629-38, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18846252

ABSTRACT

Factors that promote pancreatic beta cell growth and function are potential therapeutic targets for diabetes mellitus. In mice, genetic experiments suggest that signaling cascades initiated by insulin and IGFs positively regulate beta cell mass and insulin secretion. Akt and S6 kinase (S6K) family members are activated as part of these signaling cascades, but how the interplay between these proteins controls beta cell growth and function has not been determined. Here, we found that although transgenic mice overexpressing the constitutively active form of Akt1 under the rat insulin promoter (RIP-MyrAkt1 mice) had enlarged beta cells and high plasma insulin levels, leading to improved glucose tolerance, a substantial proportion of the mice developed insulinomas later in life, which caused decreased viability. This oncogenic transformation tightly correlated with nuclear exclusion of the tumor suppressor PTEN. To address the role of the mammalian target of rapamycin (mTOR) substrate S6K1 in the MyrAkt1-mediated phenotype, we crossed RIP-MyrAkt1 and S6K1-deficient mice. The resulting mice displayed reduced insulinemia and glycemia compared with RIP-MyrAkt1 mice due to a combined effect of improved insulin secretion and insulin sensitivity. Importantly, although the increase in beta cell size in RIP-MyrAkt1 mice was not affected by S6K1 deficiency, the hyperplastic transformation required S6K1. Our results therefore identify S6K1 as a critical element for MyrAkt1-induced tumor formation and suggest that it may represent a useful target for anticancer therapy downstream of mTOR.


Subject(s)
Insulinoma/metabolism , Pancreas/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , Animals , Cell Size , Crosses, Genetic , Disease Models, Animal , Insulin/blood , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulinoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/genetics , Rats , Ribosomal Protein S6 Kinases/genetics
15.
Biochem Soc Trans ; 39(2): 495-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428927

ABSTRACT

In recent years several reports have linked mTORC1 (mammalian target of rapamycin complex 1) to lipogenesis via the SREBPs (sterol-regulatory-element-binding proteins). SREBPs regulate the expression of genes encoding enzymes required for fatty acid and cholesterol biosynthesis. Lipid metabolism is perturbed in some diseases and SREBP target genes, such as FASN (fatty acid synthase), have been shown to be up-regulated in some cancers. We have previously shown that mTORC1 plays a role in SREBP activation and Akt/PKB (protein kinase B)-dependent de novo lipogenesis. Our findings suggest that mTORC1 plays a crucial role in the activation of SREBP and that the activation of lipid biosynthesis through the induction of SREBP could be part of a regulatory pathway that co-ordinates protein and lipid biosynthesis during cell growth. In the present paper, we discuss the increasing amount of data supporting the potential mechanisms of mTORC1-dependent activation of SREBP as well as the implications of this signalling pathway in cancer.


Subject(s)
Proteins/physiology , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Models, Biological , Multiprotein Complexes , Oncogene Protein v-akt/physiology , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Sterol Regulatory Element Binding Proteins/genetics , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism
16.
FASEB J ; 24(9): 3555-61, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20460585

ABSTRACT

Skeletal muscle mass is regulated by signaling pathways that govern protein synthesis and cell proliferation, and the mammalian target of rapamycin (mTOR) plays a key role in these processes. Recent studies suggested the crucial role of AMP-activated protein kinase (AMPK) in the inhibition of protein synthesis and cell growth. Here, we address the role of AMPK in the regulation of muscle cell size in vitro and in vivo. The size of AMPK-deficient myotubes was 1.5-fold higher than for controls. A marked increase in p70S6K Thr(389) and rpS6 Ser-235/236 phosphorylation was observed concomitantly with an up-regulation of protein synthesis rate. Treatment with rapamycin prevented p70S6K phosphorylation and rescued cell size control in AMPK-deficient cells. Importantly, myotubes lacking AMPK were resistant to further cell size increase beyond AMPK deletion alone, as MyrAkt-induced hypertrophy was absent in these cells. Moreover, in skeletal muscle-specific deficient AMPKalpha1/alpha2 KO mice, soleus muscle showed a higher mass with myofibers of larger size and was associated with increased p70S6K and rpS6 phosphorylation. Our results uncover the role of AMPK in the maintenance of muscle cell size control and highlight the crosstalk between AMPK and mTOR/p70S6K signaling pathways coordinating a metabolic checkpoint on cell growth.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Size , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Immunoblotting , In Vitro Techniques , Metformin/pharmacology , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Ribonucleotides/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
17.
Nat Commun ; 12(1): 3660, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34135321

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) integrates cellular nutrient signaling and hormonal cues to control metabolism. We have previously shown that constitutive nutrient signaling to mTORC1 by means of genetic activation of RagA (expression of GTP-locked RagA, or RagAGTP) in mice resulted in a fatal energetic crisis at birth. Herein, we rescue neonatal lethality in RagAGTP mice and find morphometric and metabolic alterations that span glucose, lipid, ketone, bile acid and amino acid homeostasis in adults, and a median lifespan of nine months. Proteomic and metabolomic analyses of livers from RagAGTP mice reveal a failed metabolic adaptation to fasting due to a global impairment in PPARα transcriptional program. These metabolic defects are partially recapitulated by restricting activation of RagA to hepatocytes, and revert by pharmacological inhibition of mTORC1. Constitutive hepatic nutrient signaling does not cause hepatocellular damage and carcinomas, unlike genetic activation of growth factor signaling upstream of mTORC1. In summary, RagA signaling dictates dynamic responses to feeding-fasting cycles to tune metabolism so as to match the nutritional state.


Subject(s)
Fasting/metabolism , Liver/metabolism , Monomeric GTP-Binding Proteins/metabolism , Signal Transduction , Animals , Disease Models, Animal , Glucose/metabolism , Homeostasis , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Monomeric GTP-Binding Proteins/genetics , Nutrients/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Phenotype , Proteomics , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , Transcription, Genetic/drug effects , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism
18.
FASEB J ; 23(7): 2264-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19237506

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) inhibits protein synthesis through the suppression of the mammalian target of rapamycin complex 1 (mTORC1), a critical regulator of muscle growth. The purpose of this investigation was to determine the role of the AMPKalpha1 catalytic subunit on muscle cell size control and adaptation to muscle hypertrophy. We found that AMPKalpha1(-/-) primary cultured myotubes and myofibers exhibit larger cell size compared with control cells in response to chronic Akt activation. We next subjected the plantaris muscle of AMPKalpha1(-/-) and control mice to mechanical overloading to induce muscle hypertrophy. We observed significant elevations of AMPKalpha1 activity in the control muscle at days 7 and 21 after the overload. Overloading-induced muscle hypertrophy was significantly accelerated in AMPKalpha1(-/-) mice than in control mice [+32 vs. +53% at day 7 and +57 vs. +76% at day 21 in control vs. AMPKalpha1(-/-) mice, respectively]. This enhanced growth of AMPKalpha1-deficient muscle was accompanied by increased phosphorylation of mTOR signaling downstream targets and decreased phosphorylation of eukaryotic elongation factor 2. These results demonstrate that AMPKalpha1 plays an important role in limiting skeletal muscle overgrowth during hypertrophy through inhibition of the mTOR-signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/physiology , Carrier Proteins/metabolism , Hypertrophy , Muscle, Skeletal/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , AMP-Activated Protein Kinases/deficiency , Animals , Cell Enlargement , Mice , Mice, Knockout , Peptide Elongation Factor 2/metabolism , Phosphorylation , Protective Agents , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Mechanical , TOR Serine-Threonine Kinases
19.
Nat Commun ; 11(1): 3200, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32581239

ABSTRACT

mTOR activation is essential and sufficient to cause polycystic kidneys in Tuberous Sclerosis Complex (TSC) and other genetic disorders. In disease models, a sharp increase of proliferation and cyst formation correlates with a dramatic loss of oriented cell division (OCD). We find that OCD distortion is intrinsically due to S6 kinase 1 (S6K1) activation. The concomitant loss of S6K1 in Tsc1-mutant mice restores OCD but does not decrease hyperproliferation, leading to non-cystic harmonious hyper growth of kidneys. Mass spectrometry-based phosphoproteomics for S6K1 substrates revealed Afadin, a known component of cell-cell junctions required to couple intercellular adhesions and cortical cues to spindle orientation. Afadin is directly phosphorylated by S6K1 and abnormally decorates the apical surface of Tsc1-mutant cells with E-cadherin and α-catenin. Our data reveal that S6K1 hyperactivity alters centrosome positioning in mitotic cells, affecting oriented cell division and promoting kidney cysts in conditions of mTOR hyperactivity.


Subject(s)
Cell Division , Kinesins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Myosins/metabolism , Polycystic Kidney Diseases/pathology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Cell Line , Kinesins/genetics , Mice , Mice, Mutant Strains , Mutation , Myosins/genetics , Phosphorylation , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism
20.
Dev Cell ; 49(3): 425-443.e9, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063758

ABSTRACT

Merlin/NF2 is a bona fide tumor suppressor whose mutations underlie inherited tumor syndrome neurofibromatosis type 2 (NF2), as well as various sporadic cancers including kidney cancer. Multiple Merlin/NF2 effector pathways including the Hippo-YAP/TAZ pathway have been identified. However, the molecular mechanisms underpinning the growth and survival of NF2-mutant tumors remain poorly understood. Using an inducible orthotopic kidney tumor model, we demonstrate that YAP/TAZ silencing is sufficient to induce regression of pre-established NF2-deficient tumors. Mechanistically, YAP/TAZ depletion diminishes glycolysis-dependent growth and increases mitochondrial respiration and reactive oxygen species (ROS) buildup, resulting in oxidative-stress-induced cell death when challenged by nutrient stress. Furthermore, we identify lysosome-mediated cAMP-PKA/EPAC-dependent activation of RAF-MEK-ERK signaling as a resistance mechanism to YAP/TAZ inhibition. Finally, unbiased analysis of TCGA primary kidney tumor transcriptomes confirms a positive correlation of a YAP/TAZ signature with glycolysis and inverse correlations with oxidative phosphorylation and lysosomal gene expression, supporting the clinical relevance of our findings.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Neurofibromin 2/deficiency , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Glycolysis , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MAP Kinase Signaling System , Mice , Mice, SCID , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Oxidative Phosphorylation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL