Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Biochemistry ; 60(23): 1808-1821, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34080844

ABSTRACT

Tuberous sclerosis protein complex (pTSC) nucleates a proteinaceous signaling hub that integrates information about the internal and external energy status of the cell in the regulation of growth and energy consumption. Biochemical and cryo-electron microscopy studies of recombinant pTSC have revealed its structure and stoichiometry and hinted at the possibility that the complex may form large oligomers. Here, we have partially purified endogenous pTSC from fasted mammalian brains of rat and pig by leveraging a recombinant antigen binding fragment (Fab) specific for the TSC2 subunit of pTSC. We demonstrate Fab-dependent purification of pTSC from membrane-solubilized fractions of the brain homogenates. Negative stain electron microscopy of the samples purified from pig brain demonstrates rod-shaped protein particles with a width of 10 nm, a variable length as small as 40 nm, and a high degree of conformational flexibility. Larger filaments are evident with a similar 10 nm width and a ≤1 µm length in linear and weblike organizations prepared from pig brain. Immunogold labeling experiments demonstrate linear aggregates of pTSC purified from mammalian brains. These observations suggest polymerization of endogenous pTSC into filamentous superstructures.


Subject(s)
Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/ultrastructure , Tuberous Sclerosis/metabolism , Animals , Cryoelectron Microscopy/methods , Cytoskeleton/metabolism , Humans , Protein Binding/physiology , Rats , Recombinant Proteins/metabolism , Signal Transduction/genetics , Swine , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism , Tumor Suppressor Proteins/metabolism
2.
Nat Commun ; 12(1): 339, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436626

ABSTRACT

Tuberous sclerosis complex (TSC) integrates upstream stimuli and regulates cell growth by controlling the activity of mTORC1. TSC complex functions as a GTPase-activating protein (GAP) towards small GTPase Rheb and inhibits Rheb-mediated activation of mTORC1. Mutations in TSC genes cause tuberous sclerosis. In this study, the near-atomic resolution structure of human TSC complex reveals an arch-shaped architecture, with a 2:2:1 stoichiometry of TSC1, TSC2, and TBC1D7. This asymmetric complex consists of two interweaved TSC1 coiled-coil and one TBC1D7 that spans over the tail-to-tail TSC2 dimer. The two TSC2 GAP domains are symmetrically cradled within the core module formed by TSC2 dimerization domain and central coiled-coil of TSC1. Structural and biochemical analyses reveal TSC2 GAP-Rheb complimentary interactions and suggest a catalytic mechanism, by which an asparagine thumb (N1643) stabilizes γ-phosphate of GTP and accelerate GTP hydrolysis of Rheb. Our study reveals mechanisms of TSC complex assembly and GAP activity.


Subject(s)
GTPase-Activating Proteins/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Tuberous Sclerosis Complex 1 Protein/chemistry , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/metabolism , Biocatalysis , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Multimerization , Tuberous Sclerosis Complex 1 Protein/ultrastructure , Tuberous Sclerosis Complex 2 Protein/ultrastructure
3.
Biochem Biophys Res Commun ; 541: 84-89, 2021 02 19.
Article in English | MEDLINE | ID: mdl-33482580

ABSTRACT

Tuberous sclerosis complex 2 (TSC2) is a tumor-suppressor protein that is partially regulated by insulin, energy, oxygen, and growth factors. Mutations in the TSC2 gene and loss of TSC2 promote cell growth by the mammalian target of rapamycin complex 1 (mTORC1) activation. Furthermore, S-adenosylmethionine (SAM) sensor upstream of mTORC1 indirectly inhibits mTORC1 activity via the methionine metabolite SAM. Here, we investigated the effects of methionine on insulin/TSC2/mTORC1 activity. Our results showed that methionine affected TSC2 stability and abolished TSC2 localization to the lysosome. Moreover, activation of insulin signaling contributed to TSC2 degradation in a methionine deprivation-dependent manner. Thus, methionine and insulin crosstalk occurred via TSC2.


Subject(s)
Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Methionine/metabolism , Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/metabolism , HEK293 Cells , HeLa Cells , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Phosphorylation , Protein Stability , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
4.
J Mol Biol ; 433(2): 166743, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33307091

ABSTRACT

The Tuberous Sclerosis Complex (TSC) protein complex (TSCC), comprising TSC1, TSC2, and TBC1D7, is widely recognised as a key integration hub for cell growth and intracellular stress signals upstream of the mammalian target of rapamycin complex 1 (mTORC1). The TSCC negatively regulates mTORC1 by acting as a GTPase-activating protein (GAP) towards the small GTPase Rheb. Both human TSC1 and TSC2 are important tumour suppressors, and mutations in them underlie the disease tuberous sclerosis. We used single-particle cryo-EM to reveal the organisation and architecture of the complete human TSCC. We show that TSCC forms an elongated scorpion-like structure, consisting of a central "body", with a "pincer" and a "tail" at the respective ends. The "body" is composed of a flexible TSC2 HEAT repeat dimer, along the surface of which runs the TSC1 coiled-coil backbone, breaking the symmetry of the dimer. Each end of the body is structurally distinct, representing the N- and C-termini of TSC1; a "pincer" is formed by the highly flexible N-terminal TSC1 core domains and a barbed "tail" makes up the TSC1 coiled-coil-TBC1D7 junction. The TSC2 GAP domain is found abutting the centre of the body on each side of the dimerisation interface, poised to bind a pair of Rheb molecules at a similar separation to the pair in activated mTORC1. Our architectural dissection reveals the mode of association and topology of the complex, casts light on the recruitment of Rheb to the TSCC, and also hints at functional higher order oligomerisation, which has previously been predicted to be important for Rheb-signalling suppression.


Subject(s)
Models, Molecular , Multiprotein Complexes/chemistry , Protein Conformation , Tuberous Sclerosis Complex 1 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/chemistry , Cryoelectron Microscopy , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins , Structure-Activity Relationship , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
5.
Structure ; 28(8): 933-942.e4, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32502382

ABSTRACT

The TSC complex is the cognate GTPase-activating protein (GAP) for the small GTPase Rheb and a crucial regulator of the mechanistic target of rapamycin complex 1 (mTORC1). Mutations in the TSC1 and TSC2 subunits of the complex cause tuberous sclerosis complex (TSC). We present the crystal structure of the catalytic asparagine-thumb GAP domain of TSC2. A model of the TSC2-Rheb complex and molecular dynamics simulations suggest that TSC2 Asn1643 and Rheb Tyr35 are key active site residues, while Rheb Arg15 and Asp65, previously proposed as catalytic residues, contribute to the TSC2-Rheb interface and indirectly aid catalysis. The TSC2 GAP domain is further stabilized by interactions with other TSC2 domains. We characterize TSC2 variants that partially affect TSC2 functionality and are associated with atypical symptoms in patients, suggesting that mutations in TSC1 and TSC2 might predispose to neurological and vascular disorders without fulfilling the clinical criteria for TSC.


Subject(s)
Catalytic Domain , Mutation, Missense , Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis/genetics , HEK293 Cells , Humans , Molecular Dynamics Simulation , Ras Homolog Enriched in Brain Protein/chemistry , Ras Homolog Enriched in Brain Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
6.
Hum Mutat ; 41(4): 759-773, 2020 04.
Article in English | MEDLINE | ID: mdl-31799751

ABSTRACT

The TSC1 and TSC2 gene products interact to form the tuberous sclerosis complex (TSC), an important negative regulator of the mechanistic target of rapamycin complex 1 (TORC1). Inactivating mutations in TSC1 or TSC2 cause TSC, and the identification of a pathogenic TSC1 or TSC2 variant helps establish a diagnosis of TSC. However, it is not always clear whether TSC1 and TSC2 variants are inactivating. To determine whether TSC1 and TSC2 variants of uncertain clinical significance affect TSC complex function and cause TSC, in vitro assays of TORC1 activity can be employed. Here we combine genetic, functional, and structural approaches to try and classify a series of 15 TSC2 VUS. We investigated the effects of the variants on the formation of the TSC complex, on TORC1 activity and on TSC2 pre-mRNA splicing. In 13 cases (87%), the functional data supported the hypothesis that the identified TSC2 variant caused TSC. Our results illustrate the benefits and limitations of functional testing for TSC.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Phenotype , Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/genetics , Amino Acid Substitution , Gene Knockdown Techniques , Genetic Association Studies/methods , Genotype , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , RNA Splicing , Structure-Activity Relationship , Tuberous Sclerosis Complex 2 Protein/metabolism
7.
Nature ; 566(7743): 264-269, 2019 02.
Article in English | MEDLINE | ID: mdl-30700906

ABSTRACT

The mechanistic target of rapamycin complex-1 (mTORC1) coordinates regulation of growth, metabolism, protein synthesis and autophagy1. Its hyperactivation contributes to disease in numerous organs, including the heart1,2, although broad inhibition of mTORC1 risks interference with its homeostatic roles. Tuberin (TSC2) is a GTPase-activating protein and prominent intrinsic regulator of mTORC1 that acts through modulation of RHEB (Ras homologue enriched in brain). TSC2 constitutively inhibits mTORC1; however, this activity is modified by phosphorylation from multiple signalling kinases that in turn inhibits (AMPK and GSK-3ß) or stimulates (AKT, ERK and RSK-1) mTORC1 activity3-9. Each kinase requires engagement of multiple serines, impeding analysis of their role in vivo. Here we show that phosphorylation or gain- or loss-of-function mutations at either of two adjacent serine residues in TSC2 (S1365 and S1366 in mice; S1364 and S1365 in humans) can bidirectionally control mTORC1 activity stimulated by growth factors or haemodynamic stress, and consequently modulate cell growth and autophagy. However, basal mTORC1 activity remains unchanged. In the heart, or in isolated cardiomyocytes or fibroblasts, protein kinase G1 (PKG1) phosphorylates these TSC2 sites. PKG1 is a primary effector of nitric oxide and natriuretic peptide signalling, and protects against heart disease10-13. Suppression of hypertrophy and stimulation of autophagy in cardiomyocytes by PKG1 requires TSC2 phosphorylation. Homozygous knock-in mice that express a phosphorylation-silencing mutation in TSC2 (TSC2(S1365A)) develop worse heart disease and have higher mortality after sustained pressure overload of the heart, owing to mTORC1 hyperactivity that cannot be rescued by PKG1 stimulation. However, cardiac disease is reduced and survival of heterozygote Tsc2S1365A knock-in mice subjected to the same stress is improved by PKG1 activation or expression of a phosphorylation-mimicking mutation (TSC2(S1365E)). Resting mTORC1 activity is not altered in either knock-in model. Therefore, TSC2 phosphorylation is both required and sufficient for PKG1-mediated cardiac protection against pressure overload. The serine residues identified here provide a genetic tool for bidirectional regulation of the amplitude of stress-stimulated mTORC1 activity.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Heart Diseases/prevention & control , Heart Diseases/physiopathology , Mechanistic Target of Rapamycin Complex 1/metabolism , Tuberous Sclerosis Complex 2 Protein/chemistry , Tuberous Sclerosis Complex 2 Protein/metabolism , Animals , Autophagy , Cells, Cultured , Disease Progression , Enzyme Activation , Everolimus/pharmacology , Female , Gene Knock-In Techniques , HEK293 Cells , Heart Diseases/genetics , Heart Diseases/pathology , Humans , Hypertrophy/drug therapy , Hypertrophy/pathology , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Mutation , Myocytes, Cardiac/pathology , Phosphorylation , Phosphoserine/metabolism , Pressure , Rats , Rats, Wistar , Serine/genetics , Serine/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...