Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
2.
Nat Commun ; 13(1): 7108, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36402791

ABSTRACT

The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Mice , Animals , Dystrophin/genetics , Dystrophin/metabolism , Mice, Inbred mdx , Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Acetylation , Transforming Growth Factor beta/metabolism , Muscle, Skeletal/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/metabolism , Fibrosis , Phenotype , Muscular Atrophy/pathology , Glycoproteins/metabolism
3.
Nucleic Acids Res ; 48(9): 4601-4613, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32266374

ABSTRACT

While the histone variant H2A.Z is known to be required for mitosis, it is also enriched in nucleosomes surrounding the transcription start site of active promoters, implicating H2A.Z in transcription. However, evidence obtained so far mainly rely on correlational data generated in actively dividing cells. We have exploited a paradigm in which transcription is uncoupled from the cell cycle by developing an in vivo system to inactivate H2A.Z in terminally differentiated post-mitotic muscle cells. ChIP-seq, RNA-seq and ATAC-seq experiments performed on H2A.Z KO post-mitotic muscle cells show that this histone variant is neither required to maintain nor to activate transcription. Altogether, this study provides in vivo evidence that in the absence of mitosis H2A.Z is dispensable for transcription and that the enrichment of H2A.Z on active promoters is a marker but not an active driver of transcription.


Subject(s)
Histones/physiology , Muscle, Skeletal/metabolism , Transcription, Genetic , Transcriptional Activation , Animals , Cell Differentiation , Cells, Cultured , Chromatin , Chromatin Immunoprecipitation Sequencing , Histones/genetics , Histones/metabolism , Mice , Muscle Fibers, Skeletal , Muscle, Skeletal/cytology , RNA-Seq , Repetitive Sequences, Nucleic Acid , Transcription Initiation Site
4.
J Cachexia Sarcopenia Muscle ; 10(1): 35-53, 2019 02.
Article in English | MEDLINE | ID: mdl-30461220

ABSTRACT

BACKGROUND: The protein kinase mechanistic target of rapamycin (mTOR) controls cellular growth and metabolism. Although balanced mTOR signalling is required for proper muscle homeostasis, partial mTOR inhibition by rapamycin has beneficial effects on various muscle disorders and age-related pathologies. Besides, more potent mTOR inhibitors targeting mTOR catalytic activity have been developed and are in clinical trials. However, the physiological impact of loss of mTOR catalytic activity in skeletal muscle is currently unknown. METHODS: We have generated the mTORmKOKI mouse model in which conditional loss of mTOR is concomitant with expression of kinase inactive mTOR in skeletal muscle. We performed a comparative phenotypic and biochemical analysis of mTORmKOKI mutant animals with muscle-specific mTOR knockout (mTORmKO) littermates. RESULTS: In striking contrast with mTORmKO littermates, mTORmKOKI mice developed an early onset rapidly progressive myopathy causing juvenile lethality. More than 50% mTORmKOKI mice died before 8 weeks of age, and none survived more than 12 weeks, while mTORmKO mice died around 7 months of age. The growth rate of mTORmKOKI mice declined beyond 1 week of age, and the animals showed profound alterations in body composition at 4 weeks of age. At this age, their body weight was 64% that of mTORmKO mice (P < 0.001) due to significant reduction in lean and fat mass. The mass of isolated muscles from mTORmKOKI mice was remarkably decreased by 38-56% (P < 0.001) as compared with that from mTORmKO mice. Histopathological analysis further revealed exacerbated dystrophic features and metabolic alterations in both slow/oxidative and fast/glycolytic muscles from mTORmKOKI mice. We show that the severity of the mTORmKOKI as compared with the mild mTORmKO phenotype is due to more robust suppression of muscle mTORC1 signalling leading to stronger alterations in protein synthesis, oxidative metabolism, and autophagy. This was accompanied with stronger feedback activation of PKB/Akt and dramatic down-regulation of glycogen phosphorylase expression (0.16-fold in tibialis anterior muscle, P < 0.01), thus causing features of glycogen storage disease type V. CONCLUSIONS: Our study demonstrates a critical role for muscle mTOR catalytic activity in the regulation of whole-body growth and homeostasis. We suggest that skeletal muscle targeting with mTOR catalytic inhibitors may have detrimental effects. The mTORmKOKI mutant mouse provides an animal model for the pathophysiological understanding of muscle mTOR activity inhibition as well as for mechanistic investigation of the influence of skeletal muscle perturbations on whole-body homeostasis.


Subject(s)
Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Disease Models, Animal , Homeostasis , Humans , Male , Mice, Transgenic , Muscular Diseases/metabolism
5.
J Cachexia Sarcopenia Muscle ; 9(5): 929-946, 2018 10.
Article in English | MEDLINE | ID: mdl-29989354

ABSTRACT

BACKGROUND: Glucocorticoids (GC) play a major role in muscle atrophy. As skeletal muscle is a secretory organ, characterization of the muscle secretome elicited by muscle atrophy should allow to better understand the cellular mechanisms and to identify circulating biomarkers of this condition. Our project aimed to identify the changes in the muscle secretome associated with GC-induced muscle atrophy and susceptible to translate into circulation. METHODS: We have identified the GC-induced changes in the secretome of C2 C12 muscle cells by proteomic analysis, and then, we have determined how these changes translate into the circulation of mice or human subjects exposed to high concentrations of GC. RESULTS: This approach led us to identify Serpina3n as one of the most markedly secreted protein in response to GC. Our original in vitro results were confirmed in vivo by an increased expression of Serpina3n in skeletal muscle (3.9-fold; P < 0.01) and in the serum (two-fold; P < 0.01) of mice treated with GC. We also observed increased levels of the human orthologue Serpina3 in the serum of Cushing's syndrome patients compared with healthy controls matched for age and sex (n = 9/group, 2.5-fold; P < 0.01). An increase of Serpina3n was also demonstrated in muscle atrophy models mediated by GC such as cancer cachexia (four-fold; P < 0.01), sepsis (12.5-fold; P < 0.001), or diabetes (two-fold; P < 0.01). In contrast, levels of Serpina3n both in skeletal muscle and in the circulation were reduced in several models of muscle hypertrophy induced by myostatin inhibition (P < 0.01). Furthermore, a cluster of data suggests that the regulation of muscle Serpina3n involves mTOR, an essential determinant of the muscle cell size. CONCLUSIONS: Taken together, these data suggest that Serpina3n may represent a circulating biomarker of muscle atrophy associated to GC and, broadly, a reflection of dynamic changes in muscle mass.


Subject(s)
Glucocorticoids/adverse effects , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Serpins/metabolism , Animals , Case-Control Studies , Cell Line , Cell Survival/drug effects , Cells, Cultured , Chromatography, Liquid , Cushing Syndrome/complications , Dexamethasone/adverse effects , Disease Models, Animal , Gene Expression , Humans , Male , Mice , Muscular Atrophy/pathology , Myoblasts , Proteome , Proteomics/methods , Serpins/blood , Tandem Mass Spectrometry
6.
Sci Rep ; 7(1): 5028, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694500

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a central mediator of protein synthesis in skeletal muscle. We utilized immunofluorescence approaches to study mTOR cellular distribution and protein-protein co-localisation in human skeletal muscle in the basal state as well as immediately, 1 and 3 h after an acute bout of resistance exercise in a fed (FED; 20 g Protein/40 g carbohydrate/1 g fat) or energy-free control (CON) state. mTOR and the lysosomal protein LAMP2 were highly co-localised in basal samples. Resistance exercise resulted in rapid translocation of mTOR/LAMP2 towards the cell membrane. Concurrently, resistance exercise led to the dissociation of TSC2 from Rheb and increased in the co-localisation of mTOR and Rheb post exercise in both FED and CON. In addition, mTOR co-localised with Eukaryotic translation initiation factor 3 subunit F (eIF3F) at the cell membrane post-exercise in both groups, with the response significantly greater at 1 h of recovery in the FED compared to CON. Collectively our data demonstrate that cellular trafficking of mTOR occurs in human muscle in response to an anabolic stimulus, events that appear to be primarily influenced by muscle contraction. The translocation and association of mTOR with positive regulators (i.e. Rheb and eIF3F) is consistent with an enhanced mRNA translational capacity after resistance exercise.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Muscle, Skeletal/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Resistance Training/methods , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Adult , Cell Membrane/metabolism , Dietary Carbohydrates/administration & dosage , Dietary Proteins/administration & dosage , Female , Humans , Male , Muscle Contraction , Protein Binding , Protein Transport , Young Adult
7.
J Mol Cell Cardiol ; 97: 213-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27133769

ABSTRACT

Mechanistic target of rapamycin (mTOR) is a central regulator of cell growth, proliferation, survival and metabolism, as part of mTOR complex 1 (mTORC1) and mTORC2. While partial inhibition of mTORC1 using rapamycin was shown to be cardioprotective, genetic studies in mouse models revealed that mTOR is essential for embryonic heart development and cardiac function in adults. However, the physiological role of mTOR during postnatal cardiac maturation is not fully elucidated. We have therefore generated a mouse model in which cardiac mTOR was inactivated at an early postnatal stage. Mutant mTORcmKO mice rapidly developed a dilated cardiomyopathy associated with cardiomyocyte growth defects, apoptosis and fibrosis, and died during their third week. Here, we show that reduced cardiomyocyte growth results from impaired protein translation efficiency through both 4E-BP1-dependent and -independent mechanisms. In addition, infant mTORcmKO hearts displayed markedly increased apoptosis linked to stretch-induced ANKRD1 (Ankyrin repeat-domain containing protein 1) up-regulation, JNK kinase activation and p53 accumulation. Pharmacological inhibition of p53 with pifithrin-α attenuated caspase-3 activation. Cardiomyocyte death did not result from activation of the MST1/Hippo pro-apoptotic pathway as reported in adult rictor/mTORC2 KO hearts. As well, mTORcmKO hearts showed a strong downregulation of myoglobin content, thereby leading to a hypoxic environment. Nevertheless, they lacked a HIF1α-mediated adaptive response, as mTOR is required for hypoxia-induced HIF-1α activation. Altogether, our results demonstrate that mTOR is critically required for cardiomyocyte growth, viability and oxygen supply in early postnatal myocardium and provide insight into the molecular mechanisms involved in apoptosis of mTOR-depleted cardiomyocytes.


Subject(s)
Apoptosis/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Biomarkers , Biopsy , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cardiopulmonary Bypass , Disease Models, Animal , Echocardiography , Energy Metabolism/genetics , Gene Expression Profiling , Gene Expression Regulation , Heart Function Tests , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Knockout , Muscle Proteins/metabolism , Myoglobin/metabolism , Nuclear Proteins/metabolism , Proteolysis , Repressor Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/metabolism
8.
J Clin Invest ; 125(7): 2736-47, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26075820

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a worldwide heath problem that is characterized by insulin resistance and the eventual loss of ß cell function. As recent studies have shown that loss of ribosomal protein (RP) S6 kinase 1 (S6K1) increases systemic insulin sensitivity, S6K1 inhibitors are being pursued as potential agents for improving insulin resistance. Here we found that S6K1 deficiency in mice also leads to decreased ß cell growth, intrauterine growth restriction (IUGR), and impaired placental development. IUGR is a common complication of human pregnancy that limits the supply of oxygen and nutrients to the developing fetus, leading to diminished embryonic ß cell growth and the onset of T2DM later in life. However, restoration of placental development and the rescue of IUGR by tetraploid embryo complementation did not restore ß cell size or insulin levels in S6K1-/- embryos, suggesting that loss of S6K1 leads to an intrinsic ß cell lesion. Consistent with this hypothesis, reexpression of S6K1 in ß cells of S6K1-/- mice restored embryonic ß cell size, insulin levels, glucose tolerance, and RPS6 phosphorylation, without rescuing IUGR. Together, these data suggest that a nutrient-mediated reduction in intrinsic ß cell S6K1 signaling, rather than IUGR, during fetal development may underlie reduced ß cell growth and eventual development of T2DM later in life.


Subject(s)
Fetal Growth Retardation/enzymology , Fetal Growth Retardation/pathology , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Animals , Cell Size , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/pathology , Female , Genetic Complementation Test , Humans , Insulin/metabolism , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Placentation/genetics , Placentation/physiology , Pregnancy , Pregnancy in Diabetics/enzymology , Pregnancy in Diabetics/pathology , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Tetraploidy
9.
Nat Immunol ; 15(8): 749-757, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973821

ABSTRACT

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


Subject(s)
Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , TOR Serine-Threonine Kinases/immunology , Animals , Cell Proliferation , Cells, Cultured , Herpesviridae Infections/immunology , Humans , Immunosuppressive Agents/pharmacology , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Killer Cells, Natural/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Muromegalovirus/immunology , Orthomyxoviridae Infections/immunology , Poly I-C/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
10.
J Neurosci ; 32(5): 1817-25, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302821

ABSTRACT

In developing peripheral nerves, differentiating Schwann cells sort individual axons from bundles and ensheath them to generate multiple layers of myelin. In recent years, there has been an increased understanding of the extracellular and intracellular factors that initiate and stimulate Schwann cell myelination, together with a growing appreciation of some of the signaling pathways involved. However, our knowledge of how Schwann cell growth is regulated during myelination is still incomplete. The mammalian target of rapamycin (mTOR) is a core kinase in two major complexes, mTORC1 and mTORC2, that regulate cell growth and differentiation in a variety of mammalian cells. Here we show that elimination of mTOR from murine Schwann cells prevented neither radial sorting nor the initiation of myelination. However, normal postnatal growth of myelinating Schwann cells, both radially and longitudinally, was highly retarded. The myelin sheath in the mutant was much thinner than normal; nevertheless, sheath thickness relative to axon diameter (g-ratio) remained constant in both wild-type and mutant nerves from P14 to P90. Although axon diameters were normal in the mutant at the initiation of myelination, further growth as myelination proceeded was retarded, and this was associated with reduced phosphorylation of neurofilaments. Consistent with thinner axonal diameters and internodal lengths, conduction velocities in mutant quadriceps nerves were also reduced. These data establish a critical role for mTOR signaling in both the longitudinal and radial growth of the myelinating Schwann cell.


Subject(s)
Axons/pathology , Myelin Sheath/pathology , Schwann Cells/pathology , TOR Serine-Threonine Kinases/deficiency , Animals , Axons/metabolism , Axons/ultrastructure , Cell Enlargement , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Organ Culture Techniques , Schwann Cells/metabolism , Schwann Cells/ultrastructure , TOR Serine-Threonine Kinases/genetics
11.
Proc Natl Acad Sci U S A ; 108(51): 20808-13, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143799

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) is central to the control of cell, organ, and body size. Skeletal muscle-specific inactivation of mTORC1 in mice results in smaller muscle fibers, fewer mitochondria, increased glycogen stores, and a progressive myopathy that causes premature death. In mTORC1-deficient muscles, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), which regulates mitochondrial biogenesis and glucose homeostasis, is strongly down-regulated. Here we tested whether induction of mitochondrial biogenesis pharmacologically or by the overexpression of PGC-1α is sufficient to reverse the phenotype of mice deficient for mTORC1. We show that both approaches normalize mitochondrial function, such as oxidative capacity and expression of mitochondrial genes. However, they do not prevent or delay the progressive myopathy. In addition, we find that mTORC1 has a much stronger effect than PGC-1α on the glycogen content in muscle. This effect is based on the strong activation of PKB/Akt in mTORC1-deficient mice. We also show that activation of PKB/Akt not only affects glycogen synthesis but also diminishes glycogen degradation. Thus, our work provides strong functional evidence that mitochondrial dysfunction in mice with inactivated mTORC1 signaling is caused by the down-regulation of PGC-1α. However, our data also show that the impairment of mitochondria does not lead directly to the lethal myopathy.


Subject(s)
Bezafibrate/pharmacology , Gene Expression Regulation , Muscular Diseases/metabolism , Proteins/metabolism , Animals , Glycogen/chemistry , Glycogen/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Models, Genetic , Multiprotein Complexes , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , TOR Serine-Threonine Kinases , Trans-Activators/metabolism , Transcription Factors
12.
J Cell Biol ; 187(6): 859-74, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-20008564

ABSTRACT

Mammalian target of rapamycin (mTOR) is a key regulator of cell growth that associates with raptor and rictor to form the mTOR complex 1 (mTORC1) and mTORC2, respectively. Raptor is required for oxidative muscle integrity, whereas rictor is dispensable. In this study, we show that muscle-specific inactivation of mTOR leads to severe myopathy, resulting in premature death. mTOR-deficient muscles display metabolic changes similar to those observed in muscles lacking raptor, including impaired oxidative metabolism, altered mitochondrial regulation, and glycogen accumulation associated with protein kinase B/Akt hyperactivation. In addition, mTOR-deficient muscles exhibit increased basal glucose uptake, whereas whole body glucose homeostasis is essentially maintained. Importantly, loss of mTOR exacerbates the myopathic features in both slow oxidative and fast glycolytic muscles. Moreover, mTOR but not raptor and rictor deficiency leads to reduced muscle dystrophin content. We provide evidence that mTOR controls dystrophin transcription in a cell-autonomous, rapamycin-resistant, and kinase-independent manner. Collectively, our results demonstrate that mTOR acts mainly via mTORC1, whereas regulation of dystrophin is raptor and rictor independent.


Subject(s)
Carrier Proteins/metabolism , Dystrophin/metabolism , Muscle, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Dystrophin/genetics , Electroporation , Energy Metabolism , Enzyme Activation , Female , Glucose/metabolism , Glycogen/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/enzymology , Muscle Contraction , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Mutation , Oxidation-Reduction , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Rats , Regulatory-Associated Protein of mTOR , Severity of Illness Index , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transduction, Genetic , Utrophin/metabolism
13.
Mol Cell Biol ; 24(21): 9508-16, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485918

ABSTRACT

The mammalian target of rapamycin (mTOR) is a key component of a signaling pathway which integrates inputs from nutrients and growth factors to regulate cell growth. Recent studies demonstrated that mice harboring an ethylnitrosourea-induced mutation in the gene encoding mTOR die at embryonic day 12.5 (E12.5). However, others have shown that the treatment of E4.5 blastocysts with rapamycin blocks trophoblast outgrowth, suggesting that the absence of mTOR should lead to embryonic lethality at an earlier stage. To resolve this discrepancy, we set out to disrupt the mTOR gene and analyze the outcome in both heterozygous and homozygous settings. Heterozygous mTOR (mTOR(+/-)) mice do not display any overt phenotype, although mouse embryonic fibroblasts derived from these mice show a 50% reduction in mTOR protein levels and phosphorylation of S6 kinase 1 T389, a site whose phosphorylation is directly mediated by mTOR. However, S6 phosphorylation, raptor levels, cell size, and cell cycle transit times are not diminished in these cells. In contrast to the situation in mTOR(+/-) mice, embryonic development of homozygous mTOR(-/-) mice appears to be arrested at E5.5; such embryos are severely runted and display an aberrant developmental phenotype. The ability of these embryos to implant corresponds to a limited level of trophoblast outgrowth in vitro, reflecting a maternal mRNA contribution, which has been shown to persist during preimplantation development. Moreover, mTOR(-/-) embryos display a lesion in inner cell mass proliferation, consistent with the inability to establish embryonic stem cells from mTOR(-/-) embryos.


Subject(s)
Embryo Loss/genetics , Embryo Loss/pathology , Embryonic Development/physiology , Protein Kinases/deficiency , Protein Kinases/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Alleles , Animals , Cell Differentiation , Cell Division , Cell Size , Cells, Cultured , Female , Fibroblasts , Gene Deletion , Heterozygote , Mice , Mice, Knockout , Pregnancy , Protein Kinases/genetics , TOR Serine-Threonine Kinases
14.
J Biol Chem ; 277(47): 45510-7, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12237303

ABSTRACT

Yeast TFIID comprises the TATA binding protein and 14 TBP-associated factors (TAF(II)s), nine of which contain histone-fold domains (HFDs). The C-terminal region of the TFIID-specific yTAF4 (yTAF(II)48) containing the HFD shares strong sequence similarity with Drosophila (d)TAF4 (dTAF(II)110) and human TAF4 (hTAF(II)135). A structure/function analysis of yTAF4 demonstrates that the HFD, a short conserved C-terminal domain (CCTD), and the region separating them are all required for yTAF4 function. Temperature-sensitive mutations in the yTAF4 HFD alpha2 helix or the CCTD can be suppressed upon overexpression of yTAF12 (yTAF(II)68). Moreover, coexpression in Escherichia coli indicates direct yTAF4-yTAF12 heterodimerization optimally requires both the yTAF4 HFD and CCTD. The x-ray crystal structure of the orthologous hTAF4-hTAF12 histone-like heterodimer indicates that the alpha3 region within the predicted TAF4 HFD is unstructured and does not correspond to the bona fide alpha3 helix. Our functional and biochemical analysis of yTAF4, rather provides strong evidence that the HFD alpha3 helix of the TAF4 family lies within the CCTD. These results reveal an unexpected and novel HFD organization in which the alpha3 helix is separated from the alpha2 helix by an extended loop containing a conserved functional domain.


Subject(s)
Protein Structure, Secondary , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Genetic Complementation Test , Humans , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics
15.
J Biol Chem ; 277(47): 45502-9, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12237304

ABSTRACT

The crystal structure is presented of a complex formed by the interacting domains from two subunits of the general transcription factor TFIID, the human TATA binding protein-associated factors hTAF4 (hTAF(II)135) and hTAF12 (hTAF(II)20). In agreement with predictions, hTAF12 forms a histone fold that is very similar to that of histone H2B, yet unexpected differences are observed between the structures of the hTAF12 interaction domain of hTAF4 and histone H2A. Most importantly, the hTAF4 fragment forms only the first two helices of a classical histone fold, which are followed by a 26-residue disordered region. This indicates that either full-length TAF4 contains an unusually long connecting loop between its second and third helix, and this helix is not required for stable interaction with TAF12, or that TAF4 represents a novel class of partial histone fold motifs. Structural models and structure-based sequence alignments support a role for TAF4b and hSTAF42/yADA1 as alternative partners for TAF12 and are consistent with the formation of nucleosome-like histone-fold octamers through interaction of TAF12 with a TAF6-TAF9 tetramer, yet argue against involvement of TAF12-containing histone-fold pairs in DNA binding.


Subject(s)
Protein Structure, Quaternary , Protein Structure, Tertiary , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Dimerization , Histones/chemistry , Histones/genetics , Humans , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Sequence Alignment , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
16.
Mol Cell Biol ; 22(9): 3178-93, 2002 May.
Article in English | MEDLINE | ID: mdl-11940675

ABSTRACT

The RNA polymerase II transcription factor TFIID, composed of the TATA-binding protein (TBP) and TBP-associated factors (TAF(II)s), nucleates preinitiation complex formation at protein-coding gene promoters. SAGA, a second TAF(II)-containing multiprotein complex, is involved in transcription regulation in Saccharomyces cerevisiae. One of the essential protein components common to SAGA and TFIID is yTAF(II)25. We define a minimal evolutionarily conserved 91-amino-acid region of TAF(II)25 containing a histone fold domain that is necessary and sufficient for growth in vivo. Different temperature-sensitive mutations of yTAF(II)25 or chimeras with the human homologue TAF(II)30 arrested cell growth at either the G(1) or G(2)/M cell cycle phase and displayed distinct phenotypic changes and gene expression patterns. Immunoprecipitation studies revealed that TAF(II)25 mutation-dependent gene expression and phenotypic changes correlated at least partially with the integrity of SAGA and TFIID. Genome-wide expression analysis revealed that the five TAF(II)25 temperature-sensitive mutant alleles individually affect the expression of between 18 and 33% of genes, whereas taken together they affect 64% of all class II genes. Thus, different yTAF(II)25 mutations induce distinct phenotypes and affect the regulation of different subsets of genes, demonstrating that no individual TAF(II) mutant allele reflects the full range of its normal functions.


Subject(s)
DNA-Binding Proteins/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Mutation/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , TATA-Binding Protein Associated Factors , Transcription Factors, TFII/chemistry , Transcription Factors, TFII/genetics , Base Composition , Blotting, Western , Cell Cycle , Chromosome Mapping , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Flow Cytometry , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Humans , Macromolecular Substances , Multiprotein Complexes , Oligonucleotide Array Sequence Analysis , Phenotype , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Temperature , Transcription Factor TFIID , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors, TFII/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...