Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 30(3): 235-247.e12, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36863346

ABSTRACT

Malignant tumors can evade destruction by the immune system by attracting immune-suppressive regulatory T cells (Treg) cells. The IKZF2 (Helios) transcription factor plays a crucial role in maintaining function and stability of Treg cells, and IKZF2 deficiency reduces tumor growth in mice. Here we report the discovery of NVP-DKY709, a selective molecular glue degrader of IKZF2 that spares IKZF1/3. We describe the recruitment-guided medicinal chemistry campaign leading to NVP-DKY709 that redirected the degradation selectivity of cereblon (CRBN) binders from IKZF1 toward IKZF2. Selectivity of NVP-DKY709 for IKZF2 was rationalized by analyzing the DDB1:CRBN:NVP-DKY709:IKZF2(ZF2 or ZF2-3) ternary complex X-ray structures. Exposure to NVP-DKY709 reduced the suppressive activity of human Treg cells and rescued cytokine production in exhausted T-effector cells. In vivo, treatment with NVP-DKY709 delayed tumor growth in mice with a humanized immune system and enhanced immunization responses in cynomolgus monkeys. NVP-DKY709 is being investigated in the clinic as an immune-enhancing agent for cancer immunotherapy.


Subject(s)
Neoplasms , Transcription Factors , Animals , Humans , Mice , Ikaros Transcription Factor , Immunotherapy , Neoplasms/therapy , Neoplasms/metabolism , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/metabolism
2.
Proc Natl Acad Sci U S A ; 116(2): 631-640, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30578319

ABSTRACT

A hallmark of prostate cancer progression is dysregulation of lipid metabolism via overexpression of fatty acid synthase (FASN), a key enzyme in de novo fatty acid synthesis. Metastatic castration-resistant prostate cancer (mCRPC) develops resistance to inhibitors of androgen receptor (AR) signaling through a variety of mechanisms, including the emergence of the constitutively active AR variant V7 (AR-V7). Here, we developed an FASN inhibitor (IPI-9119) and demonstrated that selective FASN inhibition antagonizes CRPC growth through metabolic reprogramming and results in reduced protein expression and transcriptional activity of both full-length AR (AR-FL) and AR-V7. Activation of the reticulum endoplasmic stress response resulting in reduced protein synthesis was involved in IPI-9119-mediated inhibition of the AR pathway. In vivo, IPI-9119 reduced growth of AR-V7-driven CRPC xenografts and human mCRPC-derived organoids and enhanced the efficacy of enzalutamide in CRPC cells. In human mCRPC, both FASN and AR-FL were detected in 87% of metastases. AR-V7 was found in 39% of bone metastases and consistently coexpressed with FASN. In patients treated with enzalutamide and/or abiraterone FASN/AR-V7 double-positive metastases were found in 77% of cases. These findings provide a compelling rationale for the use of FASN inhibitors in mCRPCs, including those overexpressing AR-V7.


Subject(s)
Lipogenesis , Neoplasm Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Humans , Male , Mice , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Xenograft Model Antitumor Assays
3.
Nat Med ; 24(4): 497-504, 2018 05.
Article in English | MEDLINE | ID: mdl-29457796

ABSTRACT

Genomic analyses of cancer have identified recurrent point mutations in the RNA splicing factor-encoding genes SF3B1, U2AF1, and SRSF2 that confer an alteration of function. Cancer cells bearing these mutations are preferentially dependent on wild-type (WT) spliceosome function, but clinically relevant means to therapeutically target the spliceosome do not currently exist. Here we describe an orally available modulator of the SF3b complex, H3B-8800, which potently and preferentially kills spliceosome-mutant epithelial and hematologic tumor cells. These killing effects of H3B-8800 are due to its direct interaction with the SF3b complex, as evidenced by loss of H3B-8800 activity in drug-resistant cells bearing mutations in genes encoding SF3b components. Although H3B-8800 modulates WT and mutant spliceosome activity, the preferential killing of spliceosome-mutant cells is due to retention of short, GC-rich introns, which are enriched for genes encoding spliceosome components. These data demonstrate the therapeutic potential of splicing modulation in spliceosome-mutant cancers.


Subject(s)
Neoplasms/drug therapy , Neoplasms/genetics , Piperazines/pharmacology , Pyridines/pharmacology , RNA Splicing/genetics , Small Molecule Libraries/therapeutic use , Spliceosomes/genetics , Administration, Oral , Animals , Base Sequence , Humans , Introns/genetics , K562 Cells , Leukemia/genetics , Leukemia/pathology , Mice , Mutation , Neoplasms/pathology , Piperazines/administration & dosage , Pyridines/administration & dosage , RNA Splicing/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Small Molecule Libraries/pharmacology , Tumor Burden , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 112(1): E21-9, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25524627

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) integrates multiple signals from growth factors, nutrients, and cellular energy status to control a wide range of metabolic processes, including mRNA biogenesis; protein, nucleotide, and lipid synthesis; and autophagy. Deregulation of the mTORC1 pathway is found in cancer as well as genetic disorders such as tuberous sclerosis complex (TSC) and sporadic lymphangioleiomyomatosis. Recent studies have shown that the mTORC1 inhibitor rapamycin and its analogs generally suppress proliferation rather than induce apoptosis. Therefore, it is critical to use alternative strategies to induce death of cells with activated mTORC1. In this study, a small-molecule screen has revealed that the combination of glutaminase (GLS) and heat shock protein 90 (Hsp90) inhibitors selectively triggers death of TSC2-deficient cells. At a mechanistic level, high mTORC1-driven translation rates in TSC1/2-deficient cells, unlike wild-type cells, sensitizes these cells to endoplasmic reticulum (ER) stress. Thus, Hsp90 inhibition drives accumulation of unfolded protein and ER stress. When combining proteotoxic stress with oxidative stress by depletion of the intracellular antioxidant glutathione by GLS inhibition, acute cell death is observed in cells with activated mTORC1 signaling. This study suggests that this combination strategy may have the potential to be developed into a therapeutic use for the treatment of mTORC1-driven tumors.


Subject(s)
Glutaminase/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Animals , Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival/drug effects , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/metabolism , Glutaminase/metabolism , Glutamine/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Oxidation-Reduction/drug effects , Phenotype , Sirolimus/pharmacology , Small Molecule Libraries/pharmacology , Sulfides/pharmacology , Thiadiazoles/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
5.
Curr Biol ; 24(19): 2274-80, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25220053

ABSTRACT

Growth-promoting signaling molecules, including the mammalian target of rapamycin complex 1 (mTORC1), drive the metabolic reprogramming of cancer cells required to support their biosynthetic needs for rapid growth and proliferation. Glutamine is catabolyzed to α-ketoglutarate (αKG), a tricarboxylic acid (TCA) cycle intermediate, through two deamination reactions, the first requiring glutaminase (GLS) to generate glutamate and the second occurring via glutamate dehydrogenase (GDH) or transaminases. Activation of the mTORC1 pathway has been shown previously to promote the anaplerotic entry of glutamine to the TCA cycle via GDH. Moreover, mTORC1 activation also stimulates the uptake of glutamine, but the mechanism is unknown. It is generally thought that rates of glutamine utilization are limited by mitochondrial uptake via GLS, suggesting that, in addition to GDH, mTORC1 could regulate GLS. Here we demonstrate that mTORC1 positively regulates GLS and glutamine flux through this enzyme. We show that mTORC1 controls GLS levels through the S6K1-dependent regulation of c-Myc (Myc). Molecularly, S6K1 enhances Myc translation efficiency by modulating the phosphorylation of eukaryotic initiation factor eIF4B, which is critical to unwind its structured 5' untranslated region (5'UTR). Finally, our data show that the pharmacological inhibition of GLS is a promising target in pancreatic cancers expressing low levels of PTEN.


Subject(s)
DNA-Binding Proteins/genetics , Eukaryotic Initiation Factors/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Transcription Factors/genetics , Cell Line , DNA-Binding Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Glutaminase/metabolism , Glutamine/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mitochondria/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Real-Time Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism
6.
J Exp Med ; 211(1): 15-28, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24395886

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a progressive neoplastic disorder that leads to lung destruction and respiratory failure primarily in women. LAM is typically caused by tuberous sclerosis complex 2 (TSC2) mutations resulting in mTORC1 activation in proliferative smooth muscle-like cells in the lung. The female predominance of LAM suggests that estradiol contributes to disease development. Metabolomic profiling identified an estradiol-enhanced prostaglandin biosynthesis signature in Tsc2-deficient (TSC(-)) cells, both in vitro and in vivo. Estradiol increased the expression of cyclooxygenase-2 (COX-2), a rate-limiting enzyme in prostaglandin biosynthesis, which was also increased at baseline in TSC-deficient cells and was not affected by rapamycin treatment. However, both Torin 1 treatment and Rictor knockdown led to reduced COX-2 expression and phospho-Akt-S473. Prostaglandin production was also increased in TSC-deficient cells. In preclinical models, both Celecoxib and aspirin reduced tumor development. LAM patients had significantly higher serum prostaglandin levels than healthy women. 15-epi-lipoxin-A4 was identified in exhaled breath condensate from LAM subjects and was increased by aspirin treatment, indicative of functional COX-2 expression in the LAM airway. In vitro, 15-epi-lipoxin-A4 reduced the proliferation of LAM patient-derived cells in a dose-dependent manner. Targeting COX-2 and prostaglandin pathways may have therapeutic value in LAM and TSC-related diseases, and possibly in other conditions associated with mTOR hyperactivation.


Subject(s)
Carcinogenesis/metabolism , Estradiol/metabolism , Gene Expression Regulation, Enzymologic/physiology , Lymphangioleiomyomatosis/metabolism , Multiprotein Complexes/metabolism , Prostaglandins/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Analysis of Variance , Animals , Aspirin/pharmacology , Breath Tests , Carrier Proteins/genetics , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Knockdown Techniques , Humans , Immunoblotting , Immunohistochemistry , Lipoxins/analysis , Mechanistic Target of Rapamycin Complex 2 , Metabolomics , Mice , Mice, SCID , Microscopy, Confocal , Naphthyridines/pharmacology , Prostaglandins/blood , Rapamycin-Insensitive Companion of mTOR Protein , Real-Time Polymerase Chain Reaction , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency
7.
Int J Biochem Cell Biol ; 45(10): 2158-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23769948

ABSTRACT

The eukaryotic initiation factor 3 subunit f (eIF3f) is one of the 13 subunits of the translation initiation factor complex eIF3 required for several steps in the initiation of mRNA translation. In skeletal muscle, recent studies have demonstrated that eIF3f plays a central role in skeletal muscle size maintenance. Accordingly, eIF3f overexpression results in hypertrophy through modulation of protein synthesis via the mTORC1 pathway. Importantly, eIF3f was described as a target of the E3 ubiquitin ligase MAFbx/atrogin-1 for proteasome-mediated breakdown under atrophic conditions. The biological importance of the MAFbx/atrogin-1-dependent targeting of eFI3f is highlighted by the finding that expression of an eIF3f mutant insensitive to MAFbx/atrogin-1 polyubiquitination is associated with enhanced protection against starvation-induced muscle atrophy. A better understanding of the precise role of this subunit should lead to the development of new therapeutic approaches to prevent or limit muscle wasting that prevails in numerous physiological and pathological states such as immobilization, aging, denervated conditions, neuromuscular diseases, AIDS, cancer, diabetes. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Animals , Cell Proliferation , Eukaryotic Initiation Factor-3/genetics , Humans , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Protein Biosynthesis , Signal Transduction
8.
Cancer Res ; 73(14): 4429-38, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23687346

ABSTRACT

Metformin inhibits cancer cell proliferation, and epidemiology studies suggest an association with increased survival in patients with cancer taking metformin; however, the mechanism by which metformin improves cancer outcomes remains controversial. To explore how metformin might directly affect cancer cells, we analyzed how metformin altered the metabolism of prostate cancer cells and tumors. We found that metformin decreased glucose oxidation and increased dependency on reductive glutamine metabolism in both cancer cell lines and in a mouse model of prostate cancer. Inhibition of glutamine anaplerosis in the presence of metformin further attenuated proliferation, whereas increasing glutamine metabolism rescued the proliferative defect induced by metformin. These data suggest that interfering with glutamine may synergize with metformin to improve outcomes in patients with prostate cancer.


Subject(s)
Glucose/metabolism , Glutamine/metabolism , Metformin/pharmacology , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle/drug effects , Disease Models, Animal , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
9.
Cancer Cell ; 23(4): 450-63, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23562301

ABSTRACT

DNA damage elicits a cellular signaling response that initiates cell cycle arrest and DNA repair. Here, we find that DNA damage triggers a critical block in glutamine metabolism, which is required for proper DNA damage responses. This block requires the mitochondrial SIRT4, which is induced by numerous genotoxic agents and represses the metabolism of glutamine into tricarboxylic acid cycle. SIRT4 loss leads to both increased glutamine-dependent proliferation and stress-induced genomic instability, resulting in tumorigenic phenotypes. Moreover, SIRT4 knockout mice spontaneously develop lung tumors. Our data uncover SIRT4 as an important component of the DNA damage response pathway that orchestrates a metabolic block in glutamine metabolism, cell cycle arrest, and tumor suppression.


Subject(s)
DNA Damage , Glutamine/antagonists & inhibitors , Glutamine/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Neoplasms, Experimental/genetics , Sirtuins/genetics , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , DNA Repair , Female , Glutamine/genetics , HEK293 Cells , Hep G2 Cells , Humans , Male , Mice , Mice, Knockout , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Proteins/metabolism , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Signal Transduction , Sirtuins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Am J Physiol Cell Physiol ; 303(5): C475-85, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22700795

ABSTRACT

The AMP-activated protein kinase (AMPK) is a serine/threonine protein kinase that acts as a sensor of cellular energy status switch regulating several systems including glucose and lipid metabolism. Recently, AMPK has been implicated in the control of skeletal muscle mass by decreasing mTORC1 activity and increasing protein degradation through regulation of ubiquitin-proteasome and autophagy pathways. In this review, we give an overview of the central role of AMPK in the control of skeletal muscle plasticity. We detail particularly its implication in the control of the hypertrophic and atrophic signaling pathways. In the light of these cumulative and attractive results, AMPK appears as a key player in regulating muscle homeostasis and the modulation of its activity may constitute a therapeutic potential in treating muscle wasting syndromes in humans.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism/physiology , Gene Expression Regulation, Enzymologic/physiology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Humans
11.
J Cell Biochem ; 113(2): 695-710, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22006269

ABSTRACT

In skeletal muscle, protein levels are determined by relative rates of protein synthesis and breakdown. The balance between synthesis and degradation of intracellular components determines the overall muscle fiber size. AMP-activated protein kinase (AMPK), a sensor of cellular energy status, was recently shown to increase myofibrillar protein degradation through the expression of MAFbx and MuRF1. In the present study, the effect of AMPK activation by AICAR on autophagy was investigated in muscle cells. Our results show that FoxO3a transcription factor activation by AMPK induces the expression of the autophagy-related proteins LC3B-II, Gabarapl1, and Beclin1 in primary mouse skeletal muscle myotubes and in the Tibialis anterior (TA) muscle. Time course studies reveal that AMPK activation by AICAR leads to a transient nuclear relocalization of FoxO3a followed by an increase of its cytosolic level. Moreover, AMPK activation leads to the inhibition of mTORC1 and its subsequent dissociation of Ulk1, Atg13, and FIP200 complex. Interestingly, we identify Ulk1 as a new interacting partner of AMPK in muscle cells and we show that Ulk1 is associated with AMPK under normal conditions and dissociates from AMPK during autophagy process. Moreover, we find that AMPK phosphorylates FoxO3a and Ulk1. In conclusion, our data show that AMPK activation stimulates autophagy in skeletal muscle cells through its effects on the transcriptional function of FoxO3a and takes part in the initiation of autophagosome formation by interacting with Ulk1. Here, we present new evidences that AMPK plays a crucial role in the fine tuning of protein expression programs that control skeletal muscle mass.


Subject(s)
Adenylate Kinase/physiology , Autophagy , Forkhead Transcription Factors/metabolism , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/pathology , Protein Serine-Threonine Kinases/metabolism , Adenylate Kinase/metabolism , Amino Acid Sequence , Aminoimidazole Carboxamide/analogs & derivatives , Animals , Autophagy-Related Protein-1 Homolog , Cell Line , Conserved Sequence , Enzyme Activation , Forkhead Box Protein O3 , Forkhead Transcription Factors/chemistry , Gene Expression , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes , Muscle Fibers, Skeletal/enzymology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Transport , Proteins/metabolism , Ribonucleotides , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
BMC Biol ; 9: 69, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22018140

ABSTRACT

The elevated metabolic requirements of cancer cells reflect their rapid growth and proliferation and are met through mutations in oncogenes and tumor suppressor genes that reprogram cellular processes. For example, in tuberous sclerosis complex (TSC)-related tumors, the loss of TSC1/2 function causes constitutive mTORC1 activity, which stimulates glycolysis, resulting in glucose addiction in vitro. In research published in Cell and Bioscience, Jiang and colleagues show that pharmacological restriction of glucose metabolism decreases tumor progression in a TSC xenograft model.


Subject(s)
Glycolysis , Neoplasms/metabolism , Neoplasms/therapy , Tuberous Sclerosis/metabolism , Animals , Humans , Mutation/genetics , Signal Transduction
13.
PLoS One ; 5(4): e10070, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20383279

ABSTRACT

Angiotensin II (Ang II) plays a major role in the pathogenesis of insulin resistance and diabetes by inhibiting insulin's metabolic and potentiating its trophic effects. Whereas the precise mechanisms involved remain ill-defined, they appear to be associated with and dependent upon increased oxidative stress. We found Ang II to block insulin-dependent GLUT4 translocation in L6 myotubes in an NO- and O(2)(*-)-dependent fashion suggesting the involvement of peroxynitrite. This hypothesis was confirmed by the ability of Ang II to induce tyrosine nitration of the MAP kinases ERK1/2 and of protein kinase B/Akt (Akt). Tyrosine nitration of ERK1/2 was required for their phosphorylation on Thr and Tyr and their subsequent activation, whereas it completely inhibited Akt phosphorylation on Ser(473) and Thr(308) as well as its activity. The inhibitory effect of nitration on Akt activity was confirmed by the ability of SIN-1 to completely block GSK3alpha phosphorylation in vitro. Inhibition of nitric oxide synthase and NAD(P)Hoxidase and scavenging of free radicals with myricetin restored insulin-stimulated Akt phosphorylation and GLUT4 translocation in the presence of Ang II. Similar restoration was obtained by inhibiting the ERK activating kinase MEK, indicating that these kinases regulate Akt activation. We found a conserved nitration site of ERK1/2 to be located in their kinase domain on Tyr(156/139), close to their active site Asp(166/149), in agreement with a permissive function of nitration for their activation. Taken together, our data show that Ang II inhibits insulin-mediated GLUT4 translocation in this skeletal muscle model through at least two pathways: first through the transient activation of ERK1/2 which inhibit IRS-1/2 and second through a direct inhibitory nitration of Akt. These observations indicate that not only oxidative but also nitrative stress play a key role in the pathogenesis of insulin resistance. They underline the role of protein nitration as a major mechanism in the regulation of Ang II and insulin signaling pathways and more particularly as a key regulator of protein kinase activity.


Subject(s)
Angiotensin II/pharmacology , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Nitrates/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tyrosine/metabolism , Animals , Cells, Cultured , Glucose Transporter Type 4/antagonists & inhibitors , Humans , Insulin Receptor Substrate Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Protein Transport , Rats
14.
PLoS One ; 5(2): e8994, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20126553

ABSTRACT

The mTORC1 pathway is required for both the terminal muscle differentiation and hypertrophy by controlling the mammalian translational machinery via phosphorylation of S6K1 and 4E-BP1. mTOR and S6K1 are connected by interacting with the eIF3 initiation complex. The regulatory subunit eIF3f plays a major role in muscle hypertrophy and is a key target that accounts for MAFbx function during atrophy. Here we present evidence that in MAFbx-induced atrophy the degradation of eIF3f suppresses S6K1 activation by mTOR, whereas an eIF3f mutant insensitive to MAFbx polyubiquitination maintained persistent phosphorylation of S6K1 and rpS6. During terminal muscle differentiation a conserved TOS motif in eIF3f connects mTOR/raptor complex, which phosphorylates S6K1 and regulates downstream effectors of mTOR and Cap-dependent translation initiation. Thus eIF3f plays a major role for proper activity of mTORC1 to regulate skeletal muscle size.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Myoblasts, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Animals , Binding Sites/genetics , Blotting, Western , Cell Differentiation , Cell Enlargement , Cells, Cultured , Eukaryotic Initiation Factor-3/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lysine/genetics , Lysine/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutation , Myoblasts, Skeletal/cytology , Protein Binding , Protein Biosynthesis , Protein Serine-Threonine Kinases/genetics , Proteins , RNA Interference , Ribosomal Protein S6 Kinases, 90-kDa/genetics , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
15.
PLoS One ; 4(3): e4973, 2009.
Article in English | MEDLINE | ID: mdl-19319192

ABSTRACT

Ubiquitin ligase Atrogin1/Muscle Atrophy F-box (MAFbx) up-regulation is required for skeletal muscle atrophy but substrates and function during the atrophic process are poorly known. The transcription factor MyoD controls myogenic stem cell function and differentiation, and seems necessary to maintain the differentiated phenotype of adult fast skeletal muscle fibres. We previously showed that MAFbx mediates MyoD proteolysis in vitro. Here we present evidence that MAFbx targets MyoD for degradation in several models of skeletal muscle atrophy. In cultured myotubes undergoing atrophy, MAFbx expression increases, leading to a cytoplasmic-nuclear shuttling of MAFbx and a selective suppression of MyoD. Conversely, transfection of myotubes with sh-RNA-mediated MAFbx gene silencing (shRNAi) inhibited MyoD proteolysis linked to atrophy. Furthermore, overexpression of a mutant MyoDK133R lacking MAFbx-mediated ubiquitination prevents atrophy of mouse primary myotubes and skeletal muscle fibres in vivo. Regarding the complex role of MyoD in adult skeletal muscle plasticity and homeostasis, its rapid suppression by MAFbx seems to be a major event leading to skeletal muscle wasting. Our results point out MyoD as the second MAFbx skeletal muscle target by which powerful therapies could be developed.


Subject(s)
Muscle Proteins/metabolism , MyoD Protein/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cells, Cultured , Gene Knockout Techniques , Mice , Muscle Fibers, Skeletal/cytology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscular Atrophy/prevention & control , Mutation , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/genetics
16.
J Biol Chem ; 284(7): 4413-21, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19073596

ABSTRACT

We recently presented evidence that the subunit eIF3-f of the eukaryotic initiation translation factor eIF3 that interacts with the E3-ligase Atrogin-1/muscle atrophy F-box (MAFbx) for polyubiquitination and proteasome-mediated degradation is a key target that accounts for MAFbx function during muscle atrophy. To understand this process, deletion analysis was used to identify the region of eIF3-f that is required for its proteolysis. Here, we report that the highly conserved C-terminal domain of eIF3-f is implicated for MAFbx-directed polyubiquitination and proteasomal degradation. Site-directed mutagenesis of eIF3-f revealed that the six lysine residues within this domain are required for full polyubiquitination and degradation by the proteasome. In addition, mutation of these six lysines (mutant K(5-10)R) displayed hypertrophic activity in cellulo and in vivo and was able to protect against starvation-induced muscle atrophy. Taken together, our data demonstrate that the C-terminal modifications, believed to be critical for proper eIF3-f regulation, are essential and contribute to a fine-tuning mechanism that plays an important role for eIF3-f function in skeletal muscle.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Lysine/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-3/genetics , Lysine/genetics , Mice , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Mutagenesis, Site-Directed , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary/genetics , SKP Cullin F-Box Protein Ligases/genetics , Starvation/genetics , Starvation/metabolism , Starvation/pathology , Ubiquitination/genetics
17.
Cell Cycle ; 7(12): 1698-701, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18583931

ABSTRACT

The control of muscle cell size is a physiological process balanced by a fine tuning between protein synthesis and protein degradation. MAFbx/Atrogin-1 is a muscle specific E3 ubiquitin ligase upregulated during disuse, immobilization and fasting or systemic diseases such as diabetes, cancer, AIDS and renal failure. This response is necessary to induce a rapid and functional atrophy. To date, the targets of MAFbx/Atrogin-1 in skeletal muscle remain to be identified. We have recently presented evidence that eIF3-f, a regulatory subunit of the eukaryotic translation factor eIF3 is a key target that accounts for MAFbx/Atrogin-1 function in muscle atrophy. More importantly, we showed that eIF3-f acts as a "translational enhancer" that increases the efficiency of the structural muscle proteins synthesis leading to both in vitro and in vivo muscle hypertrophy. We propose that eIF3-f subunit, a mTOR/S6K1 scaffolding protein in the IGF-1/Akt/mTOR dependent control of protein translation, is a positive actor essential to the translation of specific mRNAs probably implicated in muscle hypertrophy. The central role of eIF3-f in both the atrophic and hypertrophic pathways will be discussed in the light of its promising potential in muscle wasting therapy.


Subject(s)
Eukaryotic Initiation Factor-3/chemistry , Eukaryotic Initiation Factor-3/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Protein Biosynthesis , Animals , Eukaryotic Initiation Factor-3/antagonists & inhibitors , Humans , Hypertrophy , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Muscle, Skeletal/growth & development , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Protein Kinases/metabolism , Protein Subunits/physiology , Ribosomal Protein S6 Kinases/metabolism , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Wasting Syndrome/therapy
18.
EMBO J ; 27(8): 1266-76, 2008 Apr 23.
Article in English | MEDLINE | ID: mdl-18354498

ABSTRACT

In response to cancer, AIDS, sepsis and other systemic diseases inducing muscle atrophy, the E3 ubiquitin ligase Atrogin1/MAFbx (MAFbx) is dramatically upregulated and this response is necessary for rapid atrophy. However, the precise function of MAFbx in muscle wasting has been questioned. Here, we present evidence that during muscle atrophy MAFbx targets the eukaryotic initiation factor 3 subunit 5 (eIF3-f) for ubiquitination and degradation by the proteasome. Ectopic expression of MAFbx in myotubes induces atrophy and degradation of eIF3-f. Conversely, blockade of MAFbx expression by small hairpin RNA interference prevents eIF3-f degradation in myotubes undergoing atrophy. Furthermore, genetic activation of eIF3-f is sufficient to cause hypertrophy and to block atrophy in myotubes, whereas genetic blockade of eIF3-f expression induces atrophy in myotubes. Finally, eIF3-f induces increasing expression of muscle structural proteins and hypertrophy in both myotubes and mouse skeletal muscle. We conclude that eIF3-f is a key target that accounts for MAFbx function during muscle atrophy and has a major role in skeletal muscle hypertrophy. Thus, eIF3-f seems to be an attractive therapeutic target.


Subject(s)
Eukaryotic Initiation Factor-3/metabolism , Muscle Proteins/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , SKP Cullin F-Box Protein Ligases/physiology , Animals , Cell Line , Disease Models, Animal , Female , Humans , Hypertrophy/enzymology , Hypertrophy/metabolism , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/enzymology , Muscular Atrophy/enzymology , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/physiology , Protein Interaction Mapping , Ubiquitination
19.
Arch. venez. farmacol. ter ; 26(2): 98-104, 2007. ilus, graf
Article in Spanish | LILACS | ID: lil-516927

ABSTRACT

La adrenomedulina (AM) y el péptido relacionado con el gen de la calcitonina (CGRP) pertenecen a la superfamilia de los péptidos de CGRP. En el SNC, los sitios de unión para la AM y el CGRP se encuentran presentes en áreas hipotalámicas y en la corteza cerebelosa de la rata. La administración central de AM o de CGRP en ratas induce diuresis, natriuresis e incremento de la presión arterial. El papel de la AM en el cerebelo se desconoce. Con el fin de establecer la posible relación de la AM y CGRP cerebelosa y la regulación cardiovascular, en el presente estudio evaluamos la densidad de sitios de unión para la AM y el CGRP en el cerebelo de ratas espontáneamente hipertensas (SHR) y sus controles normotensos Wistar Kyoto (WKY) adultos de 16 semanas, mediante el uso de técnicas autoradiografícas y empleando 125I-hCGRPα y 125I-hAM13-52 como radioligandos. Los cortes coronales de cerebelo fueron incubados con 35 pM de [125I]-hCGRPα o [125I]-hAM13-52, durante 90 y 120 minutos, respectivamente. La unión no específica fue determinada en presencia de 1µM del ligando no marcado. El análisis densitométrico demostró que existe una colocalización de los sitios de unión para el [125I]-hCGRPα y la [125I]-hAM13-52 en la corteza cerebelosa. En el cerebelo la unión de la [125I]-hAM13-52 en las ratas SHR fue significativamente mayor que las WKY, indicando una mayor expresión de los receptores para la AM en el cerebelo de animales hipertensos. En relación a la unión de [125I]-hCGRPα, se observó también un pequeño incremento significativo en las ratas SHR en relación a las WKY. Con el fin de establecer la posible vía de señalización de la AM en la corteza cerebelosa, se evaluó la actividad de la óxido nítrico sintasa inducida por la AM.


Subject(s)
Male , Animals , Rats , Adrenomedullin/physiology , Cerebellum/physiology , Hypertension/physiopathology , Nitric Oxide/metabolism , Calcitonin Gene-Related Peptide/physiology , Disease Models, Animal , Rats, Sprague-Dawley , Receptors, Calcitonin Gene-Related Peptide/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...