Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38565265

ABSTRACT

A cell committed to proliferation must reshape its metabolism to enable robust yet balanced production of building blocks for the assembly of proteins, lipids, nucleic acids, and other macromolecules, from which two functional daughter cells can be produced. The metabolic remodeling associated with proliferation is orchestrated by a number of pro-proliferative signaling nodes, which include phosphatidylinositol-3 kinase (PI3K), the RAS family of small GTPases, and transcription factor c-myc In metazoan cells, these signals are activated in a paracrine manner via growth factor-mediated activation of receptor (or receptor-associated) tyrosine kinases. Such stimuli are limited in duration and therefore allow the metabolism of target cells to return to the resting state once the proliferation demands have been satisfied. Cancer cells acquire activating genetic alterations within common pro-proliferative signaling nodes. These alterations lock cellular nutrient uptake and utilization into a perpetual progrowth state, leading to the aberrant accumulation and spread of cancer cells.

2.
Cardiovasc Res ; 118(16): 3198-3210, 2022 12 29.
Article in English | MEDLINE | ID: mdl-35388887

ABSTRACT

AIMS: Cardiomyopathy and arrhythmias can be severe presentations in patients with inherited defects of mitochondrial long-chain fatty acid ß-oxidation (FAO). The pathophysiological mechanisms that underlie these cardiac abnormalities remain largely unknown. We investigated the molecular adaptations to a FAO deficiency in the heart using the long-chain acyl-CoA dehydrogenase (LCAD) knockout (KO) mouse model. METHODS AND RESULTS: We observed enrichment of amino acid metabolic pathways and of ATF4 target genes among the upregulated genes in the LCAD KO heart transcriptome. We also found a prominent activation of the eIF2α/ATF4 axis at the protein level that was independent of the feeding status, in addition to a reduction of cardiac protein synthesis during a short period of food withdrawal. These findings are consistent with an activation of the integrated stress response (ISR) in the LCAD KO mouse heart. Notably, charging of several transfer RNAs (tRNAs), such as tRNAGln was decreased in LCAD KO hearts, reflecting a reduced availability of cardiac amino acids, in particular, glutamine. We replicated the activation of the ISR in the hearts of mice with muscle-specific deletion of carnitine palmitoyltransferase 2. CONCLUSIONS: Our results show that perturbations in amino acid metabolism caused by long-chain FAO deficiency impact cardiac metabolic signalling, in particular the ISR. These results may serve as a foundation for investigating the role of the ISR in the cardiac pathology associated with long-chain FAO defects.Translational Perspective: The heart relies mainly on mitochondrial fatty acid ß-oxidation (FAO) for its high energy requirements. The heart disease observed in patients with a genetic defect in this pathway highlights the importance of FAO for cardiac health. We show that the consequences of a FAO defect extend beyond cardiac energy homeostasis and include amino acid metabolism and associated signalling pathways such as the integrated stress response.


Subject(s)
Fatty Acids , Mitochondria , Mice , Animals , Mitochondria/metabolism , Fatty Acids/metabolism , Oxidation-Reduction , Mice, Knockout , Amino Acids/metabolism , RNA, Transfer/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism
3.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35105808

ABSTRACT

Cells acquire essential nutrients from the environment and utilize adaptive mechanisms to survive when nutrients are scarce. How nutrients are trafficked and compartmentalized within cells and whether they are stored in response to stress remain poorly understood. Here, we investigate amino acid trafficking and uncover evidence for the lysosomal transit of numerous essential amino acids. We find that starvation induces the lysosomal retention of leucine in a manner requiring RAG-GTPases and the lysosomal protein complex Ragulator, but that this process occurs independently of mechanistic target of rapamycin complex 1 activity. We further find that stored leucine is utilized in protein synthesis and that inhibition of protein synthesis releases lysosomal stores. These findings identify a regulated starvation response that involves the lysosomal storage of leucine.


Subject(s)
Leucine/metabolism , Lysosomes/metabolism , Signal Transduction , Stress, Physiological , Animals , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , RAW 264.7 Cells
4.
Cell Metab ; 34(3): 355-377, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35123658

ABSTRACT

Metabolism of cancer cells is geared toward biomass production and proliferation. Since the metabolic resources within the local tissue are finite, this can lead to nutrient depletion and accumulation of metabolic waste. To maintain growth in these conditions, cancer cells employ a variety of metabolic adaptations, the nature of which is collectively determined by the physiology of their cell of origin, the identity of transforming lesions, and the tissue in which cancer cells reside. Furthermore, select metabolites not only serve as substrates for energy and biomass generation, but can also regulate gene and protein expression and influence the behavior of non-transformed cells in the tumor vicinity. As they grow and metastasize, tumors can also affect and be affected by the nutrient distribution within the body. In this hallmark update, recent advances are incorporated into a conceptual framework that may help guide further research efforts in exploring cancer cell metabolism.


Subject(s)
Energy Metabolism , Neoplasms , Energy Metabolism/physiology , Humans , Neoplasms/metabolism , Nutrients
5.
Nat Metab ; 3(11): 1484-1499, 2021 11.
Article in English | MEDLINE | ID: mdl-34764457

ABSTRACT

The aberrant production of collagen by fibroblasts is a hallmark of many solid tumours and can influence cancer progression. How the mesenchymal cells in the tumour microenvironment maintain their production of extracellular matrix proteins as the vascular delivery of glutamine and glucose becomes compromised remains unclear. Here we show that pyruvate carboxylase (PC)-mediated anaplerosis in tumour-associated fibroblasts contributes to tumour fibrosis and growth. Using cultured mesenchymal and cancer cells, as well as mouse allograft models, we provide evidence that extracellular lactate can be utilized by fibroblasts to maintain tricarboxylic acid (TCA) cycle anaplerosis and non-essential amino acid biosynthesis through PC activity. Furthermore, we show that fibroblast PC is required for collagen production in the tumour microenvironment. These results establish TCA cycle anaplerosis as a determinant of extracellular matrix collagen production, and identify PC as a potential target to inhibit tumour desmoplasia.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Collagen/biosynthesis , Neoplasms/etiology , Neoplasms/metabolism , Pyruvate Carboxylase/metabolism , Tumor Microenvironment , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line , Citric Acid Cycle , Disease Susceptibility , Enzyme Activation/drug effects , Fibrosis , Gene Expression Regulation, Enzymologic , Glutamine/metabolism , Humans , Lactic Acid/metabolism , Mice , Neoplasms/pathology , Protein Biosynthesis , Pyruvate Carboxylase/genetics , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/genetics
6.
Elife ; 92020 12 08.
Article in English | MEDLINE | ID: mdl-33289483

ABSTRACT

An inadequate supply of amino acids leads to accumulation of uncharged tRNAs, which can bind and activate GCN2 kinase to reduce translation. Here, we show that glutamine-specific tRNAs selectively become uncharged when extracellular amino acid availability is compromised. In contrast, all other tRNAs retain charging of their cognate amino acids in a manner that is dependent upon intact lysosomal function. In addition to GCN2 activation and reduced total translation, the reduced charging of tRNAGln in amino-acid-deprived cells also leads to specific depletion of proteins containing polyglutamine tracts including core-binding factor α1, mediator subunit 12, transcriptional coactivator CBP and TATA-box binding protein. Treating amino-acid-deprived cells with exogenous glutamine or glutaminase inhibitors restores tRNAGln charging and the levels of polyglutamine-containing proteins. Together, these results demonstrate that the activation of GCN2 and the translation of polyglutamine-encoding transcripts serve as key sensors of glutamine availability in mammalian cells.


Subject(s)
Amino Acids/deficiency , Protein Biosynthesis , RNA, Transfer, Gln/metabolism , Transfer RNA Aminoacylation , Animals , Cell Line, Tumor , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Glutamine/metabolism , Humans , Mice , Peptides/metabolism
7.
J Biol Chem ; 294(49): 18674-18684, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31659118

ABSTRACT

Tumor cells adapt to nutrient-limited environments by inducing gene expression that ensures adequate nutrients to sustain metabolic demands. For example, during amino acid limitations, ATF4 in the amino acid response induces expression of asparagine synthetase (ASNS), which provides for asparagine biosynthesis. Acute lymphoblastic leukemia (ALL) cells are sensitive to asparagine depletion, and administration of the asparagine depletion enzyme l-asparaginase is an important therapy option. ASNS expression can counterbalance l-asparaginase treatment by mitigating nutrient stress. Therefore, understanding the mechanisms regulating ASNS expression is important to define the adaptive processes underlying tumor progression and treatment. Here we show that DNA hypermethylation at the ASNS promoter prevents its transcriptional expression following asparagine depletion. Insufficient expression of ASNS leads to asparagine deficiency, which facilitates ATF4-independent induction of CCAAT-enhancer-binding protein homologous protein (CHOP), which triggers apoptosis. We conclude that chromatin accessibility is critical for ATF4 activity at the ASNS promoter, which can switch ALL cells from an ATF4-dependent adaptive response to ATF4-independent apoptosis during asparagine depletion. This work may also help explain why ALL cells are most sensitive to l-asparaginase treatment compared with other cancers.


Subject(s)
Activating Transcription Factor 4/metabolism , Asparagine/metabolism , Aspartate-Ammonia Ligase/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Activating Transcription Factor 4/genetics , Aspartate-Ammonia Ligase/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Promoter Regions, Genetic/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism
8.
Mol Cell Oncol ; 5(3): e1441633, 2018.
Article in English | MEDLINE | ID: mdl-30250896

ABSTRACT

A challenge of targeting glutamine metabolism in cancer is that tumor cells develop various strategies to adapt to glutamine limitation. We found that asparagine plays a critical role in supporting protein synthesis during glutamine starvation, highlighting a possible approach to optimize the therapeutic efficacy of targeting glutamine metabolism in cancer.

9.
Cell Metab ; 27(2): 428-438.e5, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29337136

ABSTRACT

When mammalian cells are deprived of glutamine, exogenous asparagine rescues cell survival and growth. Here we report that this rescue results from use of asparagine in protein synthesis. All mammalian cell lines tested lacked cytosolic asparaginase activity and could not utilize asparagine to produce other amino acids or biosynthetic intermediates. Instead, most glutamine-deprived cell lines are capable of sufficient glutamine synthesis to maintain essential amino acid uptake and production of glutamine-dependent biosynthetic precursors, with the exception of asparagine. While experimental introduction of cytosolic asparaginase could enhance the synthesis of glutamine and increase tricarboxylic acid cycle anaplerosis and the synthesis of nucleotide precursors, cytosolic asparaginase suppressed the growth and survival of cells in glutamine-depleted medium in vitro and severely compromised the in vivo growth of tumor xenografts. These results suggest that the lack of asparaginase activity represents an evolutionary adaptation to allow mammalian cells to survive pathophysiologic variations in extracellular glutamine.


Subject(s)
Amino Acids, Essential/metabolism , Asparagine/metabolism , Extracellular Space/metabolism , Glutamine/metabolism , Animals , Asparaginase/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Female , Glutamine/biosynthesis , Humans , Mammals/metabolism , Mice, Inbred C57BL , Protein Biosynthesis , Substrate Specificity
10.
EMBO J ; 36(10): 1302-1315, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28420743

ABSTRACT

Biochemistry textbooks and cell culture experiments seem to be telling us two different things about the significance of external glutamine supply for mammalian cell growth and proliferation. Despite the fact that glutamine is a nonessential amino acid that can be synthesized by cells from glucose-derived carbons and amino acid-derived ammonia, most mammalian cells in tissue culture cannot proliferate or even survive in an environment that does not contain millimolar levels of glutamine. Not only are the levels of glutamine in standard tissue culture media at least ten-fold higher than other amino acids, but glutamine is also the most abundant amino acid in the human bloodstream, where it is assiduously maintained at approximately 0.5 mM through a combination of dietary uptake, de novo synthesis, and muscle protein catabolism. The complex metabolic logic of the proliferating cancer cells' appetite for glutamine-which goes far beyond satisfying their protein synthesis requirements-has only recently come into focus. In this review, we examine the diversity of biosynthetic and regulatory uses of glutamine and their role in proliferation, stress resistance, and cellular identity, as well as discuss the mechanisms that cells utilize in order to adapt to glutamine limitation.


Subject(s)
Cell Proliferation , Glutamine/metabolism , Neoplasms/metabolism , Animals , Humans
11.
Cell Metab ; 23(1): 27-47, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26771115

ABSTRACT

Tumorigenesis is dependent on the reprogramming of cellular metabolism as both direct and indirect consequence of oncogenic mutations. A common feature of cancer cell metabolism is the ability to acquire necessary nutrients from a frequently nutrient-poor environment and utilize these nutrients to both maintain viability and build new biomass. The alterations in intracellular and extracellular metabolites that can accompany cancer-associated metabolic reprogramming have profound effects on gene expression, cellular differentiation, and the tumor microenvironment. In this Perspective, we have organized known cancer-associated metabolic changes into six hallmarks: (1) deregulated uptake of glucose and amino acids, (2) use of opportunistic modes of nutrient acquisition, (3) use of glycolysis/TCA cycle intermediates for biosynthesis and NADPH production, (4) increased demand for nitrogen, (5) alterations in metabolite-driven gene regulation, and (6) metabolic interactions with the microenvironment. While few tumors display all six hallmarks, most display several. The specific hallmarks exhibited by an individual tumor may ultimately contribute to better tumor classification and aid in directing treatment.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Transport , Energy Metabolism , Humans , Metabolic Networks and Pathways , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction
12.
Cell ; 162(2): 259-270, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26144316

ABSTRACT

Despite being surrounded by diverse nutrients, mammalian cells preferentially metabolize glucose and free amino acids. Recently, Ras-induced macropinocytosis of extracellular proteins was shown to reduce a transformed cell's dependence on extracellular glutamine. Here, we demonstrate that protein macropinocytosis can also serve as an essential amino acid source. Lysosomal degradation of extracellular proteins can sustain cell survival and induce activation of mTORC1 but fails to elicit significant cell accumulation. Unlike its growth-promoting activity under amino-acid-replete conditions, we discovered that mTORC1 activation suppresses proliferation when cells rely on extracellular proteins as an amino acid source. Inhibiting mTORC1 results in increased catabolism of endocytosed proteins and enhances cell proliferation during nutrient-depleted conditions in vitro and within vascularly compromised tumors in vivo. Thus, by preventing nutritional consumption of extracellular proteins, mTORC1 couples growth to availability of free amino acids. These results may have important implications for the use of mTOR inhibitors as therapeutics.


Subject(s)
Embryo, Mammalian/cytology , Multiprotein Complexes/metabolism , Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Albumins/metabolism , Amino Acids/metabolism , Animals , Cell Proliferation , Cell Survival , Eukaryota/classification , Eukaryota/cytology , Eukaryota/metabolism , Fibroblasts/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Pinocytosis , Proteins/chemistry , ras Proteins/metabolism
13.
Elife ; 2: e00358, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23682311

ABSTRACT

During all stages of tumor progression, cancer cells are subjected to inappropriate extracellular matrix environments and must undergo adaptive changes in order to evade growth constraints associated with the loss of matrix attachment. A gain of function screen for genes that enable proliferation independently of matrix anchorage identified a cell adhesion molecule PVRL4 (poliovirus-receptor-like 4), also known as Nectin-4. PVRL4 promotes anchorage-independence by driving cell-to-cell attachment and matrix-independent integrin ß4/SHP-2/c-Src activation. Solid tumors frequently have copy number gains of the PVRL4 locus and some have focal amplifications. We demonstrate that the transformation of breast cancer cells is dependent on PVRL4. Furthermore, growth of orthotopically implanted tumors in vivo is inhibited by blocking PVRL4-driven cell-to-cell attachment with monoclonal antibodies, demonstrating a novel strategy for targeted therapy of cancer. DOI:http://dx.doi.org/10.7554/eLife.00358.001.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Cell Communication , Cell Transformation, Neoplastic/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Copy Number Variations , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Integrin beta4/metabolism , Mice, Nude , Nectins , RNA Interference , Signal Transduction , Time Factors , Tumor Burden , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
14.
Proc Natl Acad Sci U S A ; 110(5): E407-14, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23319639

ABSTRACT

Cancer develops through genetic and epigenetic alterations that allow unrestrained proliferation and increased survival. Using a genetic RNAi screen, we previously identified hundreds of suppressors of tumorigenesis and/or proliferation (STOP) genes that restrain normal cell proliferation. Our STOP gene set was significantly enriched for known and putative tumor suppressor genes. Here, we report a tumor-suppressive role for one STOP gene, phosphatase and actin regulator 4 (PHACTR4). Phactr4 is one of four members of the largely uncharacterized Phactr family of protein phosphatase 1 (PP1)-and actin-binding proteins. Our work suggests that Phactr4 restrains normal cell proliferation and transformation. Depletion of Phactr4 with multiple shRNAs leads to increased proliferation and soft agar colony formation. Phactr4 acts, in part, through an Rb-dependent pathway, because Rb phosphorylation is maintained upon growth factor withdrawal in Phactr4-depleted cells. Examination of tumor copy number analysis and sequencing revealed that PHACTR4 is significantly deleted and mutant in many tumor subtypes. Furthermore,cancer cell lines with reduced Phactr4 expression exhibit tumor suppressor hypersensitivity upon Phactr4 complementation,leading to reduced proliferation, transformation, and tumor formation. Thus, Phactr4 acts as a tumor suppressor that is deleted and mutant in several cancers.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation , Mutation , Tumor Suppressor Proteins/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Doxycycline/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoblotting , MCF-7 Cells , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Transfection , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism
15.
Proc Natl Acad Sci U S A ; 109(45): 18523-8, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23064642

ABSTRACT

Antibody discovery platforms have become an important source of both therapeutic biomolecules and research reagents. Massively parallel DNA sequencing can be used to assist antibody selection by comprehensively monitoring libraries during selection, thus greatly expanding the power of these systems. We have therefore constructed a rationally designed, fully defined single-chain variable fragment (scFv) library and analysis platform optimized for analysis with short-read deep sequencing. Sequence-defined oligonucleotide libraries encoding three complementarity-determining regions (L3 from the light chain, H2 and H3 from the heavy chain) were synthesized on a programmable microarray and combinatorially cloned into a single scFv framework for molecular display. Our unique complementarity-determining region sequence design optimizes for protein binding by utilizing a hidden Markov model that was trained on all antibody-antigen cocrystal structures in the Protein Data Bank. The resultant ~10(12)-member library was produced in ribosome-display format, and comprehensively analyzed over four rounds of antigen selections by multiplex paired-end Illumina sequencing. The hidden Markov model scFv library generated multiple binders against an emerging cancer antigen and is the basis for a next-generation antibody production platform.


Subject(s)
Antibodies/immunology , Peptide Library , Sequence Analysis, DNA , Cell Adhesion Molecules/immunology , Clone Cells , Flow Cytometry , Humans , Markov Chains , Single-Chain Antibodies/immunology
16.
Cell ; 144(5): 703-18, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376233

ABSTRACT

Among breast cancers, triple-negative breast cancer (TNBC) is the most poorly understood and is refractory to current targeted therapies. Using a genetic screen, we identify the PTPN12 tyrosine phosphatase as a tumor suppressor in TNBC. PTPN12 potently suppresses mammary epithelial cell proliferation and transformation. PTPN12 is frequently compromised in human TNBCs, and we identify an upstream tumor-suppressor network that posttranscriptionally controls PTPN12. PTPN12 suppresses transformation by interacting with and inhibiting multiple oncogenic tyrosine kinases, including HER2 and EGFR. The tumorigenic and metastatic potential of PTPN12-deficient TNBC cells is severely impaired upon restoration of PTPN12 function or combined inhibition of PTPN12-regulated tyrosine kinases, suggesting that TNBCs are dependent on the proto-oncogenic tyrosine kinases constrained by PTPN12. Collectively, these data identify PTPN12 as a commonly inactivated tumor suppressor and provide a rationale for combinatorially targeting proto-oncogenic tyrosine kinases in TNBC and other cancers based on their profile of tyrosine-phosphatase activity.


Subject(s)
Breast Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , MicroRNAs/metabolism , Mutation , Neoplasm Metastasis , Protein Processing, Post-Translational
17.
Nature ; 452(7185): 370-4, 2008 Mar 20.
Article in English | MEDLINE | ID: mdl-18354483

ABSTRACT

The RE1-silencing transcription factor (REST, also known as NRSF) is a master repressor of neuronal gene expression and neuronal programmes in non-neuronal lineages. Recently, REST was identified as a human tumour suppressor in epithelial tissues, suggesting that its regulation may have important physiological and pathological consequences. However, the pathways controlling REST have yet to be elucidated. Here we show that REST is regulated by ubiquitin-mediated proteolysis, and use an RNA interference (RNAi) screen to identify a Skp1-Cul1-F-box protein complex containing the F-box protein beta-TRCP (SCF(beta-TRCP)) as an E3 ubiquitin ligase responsible for REST degradation. beta-TRCP binds and ubiquitinates REST and controls its stability through a conserved phospho-degron. During neural differentiation, REST is degraded in a beta-TRCP-dependent manner. beta-TRCP is required for proper neural differentiation only in the presence of REST, indicating that beta-TRCP facilitates this process through degradation of REST. Conversely, failure to degrade REST attenuates differentiation. Furthermore, we find that beta-TRCP overexpression, which is common in human epithelial cancers, causes oncogenic transformation of human mammary epithelial cells and that this pathogenic function requires REST degradation. Thus, REST is a key target in beta-TRCP-driven transformation and the beta-TRCP-REST axis is a new regulatory pathway controlling neurogenesis.


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic , Neurons/cytology , Neurons/pathology , Repressor Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Transcription Factors/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , Cell Line, Tumor , Conserved Sequence , Humans , Mice , Phosphorylation , Protein Processing, Post-Translational , RNA Interference , Repressor Proteins/genetics , Substrate Specificity , Transcription Factors/genetics , Ubiquitin/metabolism , beta-Transducin Repeat-Containing Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...