Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Oncol ; 12: 990672, 2022.
Article in English | MEDLINE | ID: mdl-36176395

ABSTRACT

The sterol regulatory-element binding proteins (SREBPs) are transcription factors controlling cholesterol and fatty acid synthesis and metabolism. There are three SREBP proteins, SREBP1a, SREBP1c and SREBP2, with SREBP1a being the strongest transcription factor. The expression of SREBP1a is restricted to rapidly proliferating cells, including cancer cells. The SREBP proteins are translated as large, inactive precursors bound to the endoplasmic reticulum (ER) membranes. These precursors undergo a two-step cleavage process that releases the amino terminal domains of the proteins, which translocate to the nucleus and function as transcription factors. The nuclear forms of the SREBPs are rapidly degraded by the ubiquitin-proteasome system in a manner dependent on the Fbw7 ubiquitin ligase. Consequently, inactivation of Fbw7 results in the stabilization of active SREBP1 and SREBP2 and enhanced expression of target genes. We report that the inactivation of Fbw7 in cancer cells blocks the proteolytic maturation of SREBP2. The same is true in cells expressing a cancer-specific loss-of-function Fbw7 protein. Interestingly, the activation of SREBP2 is restored in response to cholesterol depletion, suggesting that Fbw7-deficient cells accumulate cholesterol. Importantly, inactivation of SREBP1 in Fbw7-deficient cells also restores the cholesterol-dependent regulation of SREBP2, suggesting that the stabilization of active SREBP1 molecules could be responsible for the blunted activation of SREBP2 in Fbw7-deficient cancer cells. We suggest that this could be an important negative feedback loop in cancer cells with Fbw7 loss-of-function mutations to protect these cells from the accumulation of toxic levels of cholesterol and/or cholesterol metabolites. Surprisingly, we also found that the inactivation of Fbw7 resulted in the activation of AKT. Importantly, the activation of AKT was dependent on SREBP1 and on the accumulation of cholesterol. Thus, we suggest that the loss of Fbw7 rewires lipid metabolism in cancer cells to support cell proliferation and survival.

2.
Front Oncol ; 12: 942386, 2022.
Article in English | MEDLINE | ID: mdl-36091143

ABSTRACT

The sterol regulatory-element binding protein (SREBP) family of transcription factors regulates cholesterol, fatty acid, and triglyceride synthesis and metabolism. However, they are also targeted by the ubiquitin ligase Fbw7, a major tumor suppressor, suggesting that they could regulate cell growth. Indeed, enhanced lipid synthesis is a hallmark of many human tumors. Thus, the SREBP pathway has recently emerged as a potential target for cancer therapy. We have previously demonstrated that one of these transcription factors, SREBP1, is stabilized and remains associated with target promoters during mitosis, suggesting that the expression of these target genes could be important as cells enter G1 and transcription is restored. Activation of cyclin D-cdk4/6 complexes is critical for the phosphorylation and inactivation of the retinoblastoma protein (Rb) family of transcriptional repressors and progression through the G1 phase of the cell cycle. Importantly, the cyclin D-cdk4/6-Rb regulatory axis is frequently dysregulated in human cancer. In the current manuscript, we demonstrate that SREBP1 activates the expression of cyclin D1, a coactivator of cdk4 and cdk6, by binding to an E-box in the cyclin D1 promoter. Consequently, inactivation of SREBP1 in human liver and breast cancer cell lines reduces the expression of cyclin D1 and attenuates Rb phosphorylation. Rb phosphorylation in these cells can be rescued by restoring cyclin D1 expression. On the other hand, expression of active SREBP1 induced the expression of cyclin D1 and increased the phosphorylation of Rb in a manner dependent on cyclin D1 and cdk4/6 activity. Inactivation of SREBP1 resulted in reduced expression of cyclin D1, attenuated phosphorylation of Rb, and reduced proliferation. Inactivation of SREBP1 also reduced the insulin-dependent regulation of the cyclin D1 gene. At the same time, SREBP1 is known to play an important role in supporting lipid synthesis in cancer cells. Thus, we propose that the SREBP1-dependent regulation of cyclin D1 coordinates cell proliferation with the enhanced lipid synthesis required to support cell growth.

3.
Genes Dev ; 27(23): 2531-6, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24298052

ABSTRACT

The Fbw7 tumor suppressor targets a broad network of proteins for ubiquitylation. Here we show critical functions for Fbw7 dimerization in regulating the specificity and robustness of degradation. Dimerization enables Fbw7 to target substrates through concerted binding to two suboptimal and independent recognition sites. Accordingly, an endogenous dimerization-deficient Fbw7 mutation stabilizes suboptimal substrates. Dimerization increases Fbw7's robustness by preserving its function in the setting of mutations that disable Fbw7 monomers, thereby buffering against pathogenic mutations. Finally, dimerization regulates Fbw7 stability, and this likely involves Fbw7 trans-autoubiquitylation. Our study reveals novel functions of Fbw7 dimerization and an unanticipated complexity in substrate degradation.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Cell Cycle Proteins/chemistry , Dimerization , F-Box Proteins/chemistry , F-Box-WD Repeat-Containing Protein 7 , HCT116 Cells , Humans , Ligases/chemistry , Ligases/metabolism , Mutation , Protein Binding , Protein Stability , Substrate Specificity , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
4.
Proc Natl Acad Sci U S A ; 107(26): 11817-22, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20534483

ABSTRACT

Adipose tissue controls body lipid and energy metabolism, as well as food intake, and abnormalities in adipose function play a central role in diseases such as obesity and type-2 diabetes. Adipocyte differentiation is controlled by a transcriptional cascade involving PPARgamma and members of the C/EBP family of transcription factors. Here, we demonstrate that C/EBPalpha is targeted for degradation by the ubiquitin ligase Fbxw7 in a phosphorylation-dependent manner. Importantly, inactivation of Fbxw7 is sufficient to convert mouse preadipocytes into mature adipocytes in a manner dependent on C/EBPalpha. In addition, inactivation of Fbxw7 promotes adipocyte differentiation of human adult stem cells. Taken together, our results suggest that Fbxw7 is a negative regulator of adipogenesis by targeting C/EBPalpha for degradation. This notion is supported by the observation that the expression of Fbxw7 is down-regulated during adipocyte differentiation, resulting in the accumulation of proadipogenic proteins such as C/EBPalpha. Thus, Fbxw7 could be an important regulator of energy and lipid metabolism.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Adipogenesis/physiology , Adult , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Differentiation , F-Box Proteins/antagonists & inhibitors , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Humans , Mice , Models, Biological , NIH 3T3 Cells , RNA, Small Interfering/genetics , Substrate Specificity , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
6.
J Biol Chem ; 284(9): 5885-95, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19126544

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are a family of transcription factors that regulates cholesterol and lipid metabolism. The active forms of these transcription factors are targeted by a number of post-translational modifications, including phosphorylation. Phosphorylation of Thr-426 and Ser-430 in SREBP1a creates a docking site for the ubiquitin ligase Fbw7, resulting in the degradation of the transcription factor. Here, we identify a novel phosphorylation site in SREBP1a, Ser-434, which regulates the Fbw7-dependent degradation of SREBP1. We demonstrate that both SREBP1a and SREBP1c are phosphorylated on this residue (Ser-410 in SREBP1c). Importantly, we demonstrate that the mature form of endogenous SREBP1 is phosphorylated on Ser-434. Glycogen synthase kinase-3 phosphorylates Ser-434, and the phosphorylation of this residue is attenuated in response to insulin signaling. Interestingly, phosphorylation of Ser-434 promotes the glycogen synthase kinase-3-dependent phosphorylation of Thr-426 and Ser-430 and destabilizes SREBP1. Consequently, mutation of Ser-434 blocks the interaction between SREBP1 and Fbw7 and attenuates Fbw7-dependent degradation of SREBP1. Importantly, insulin fails to enhance the levels of mature SREBP1 in cells lacking Fbw7. Thus, the degradation of mature SREBP1 is controlled by cross-talk between multiple phosphorylated residues in its C-terminal domain and the phosphorylation of Ser-434 could function as a molecular switch to control these processes.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Cycle Proteins/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Insulin/metabolism , Luciferases/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination , beta-Galactosidase/metabolism
7.
Curr Opin Cell Biol ; 19(2): 215-22, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17303406

ABSTRACT

The last few years have seen important advances in defining the mechanisms that cells use to monitor changes in cholesterol levels and regulate lipid metabolism. This work has unraveled a feedback system that enables cholesterol and certain sterol intermediates to regulate the proteolysis and transport of specific membrane proteins. The sterol regulatory element-binding protein (SREBP) family of transcription factors is at the center of this feedback system. These membrane-embedded proteins are activated by ER-to-Golgi transport followed by limited proteolysis. In addition, both the activation of the SREBPs and the stability of the rate-limiting enzyme in cholesterol synthesis are regulated by the ubiquitin-proteasome system in a sterol-dependent manner. The ubiquitin-proteasome system also regulates the degradation of active SREBPs. Recent work also highlights the important role of this regulatory system in several organisms, ranging from yeast to humans. In addition, the SREBP pathway has been found to regulate a diverse set of cellular processes, including phagocytosis, cell cycle progression, oxygen sensing and survival in response to bacterial infection. These advances illustrate the wide-ranging roles that SREBPs and membrane biogenesis have in cell biology.


Subject(s)
Cholesterol/metabolism , Signal Transduction , Sterol Regulatory Element Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cholesterol/genetics , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipids/biosynthesis , Models, Biological , Models, Genetic , Molecular Sequence Data , Protein Processing, Post-Translational
8.
Cell Cycle ; 5(15): 1708-18, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16880739

ABSTRACT

Members of the sterol regulatory element-binding protein (SREBP) family of transcription factors control the biosynthesis of cholesterol and other lipids, and lipid synthesis is critical for cell growth and proliferation. We recently found that the mature forms of SREBP1a and SREBP1c are hyperphosphorylated in mitotic cells, giving rise to a phosphoepitope recognized by the mitotic protein monoclonal-2 (MPM-2) antibody. In addition, we found that mature SREBP1 was stabilized in a phosphorylation-dependent manner during mitosis. We have now mapped the major MPM-2 epitope to a serine residue, S439, in the C terminus of mature SREBP1. Using phosphorylation-specific antibodies, we demonstrate that endogenous SREBP1 is phosphorylated on S439 specifically during mitosis. Mature SREBP1 interacts with the Cdk1/cyclin B complex in mitotic cells and we demonstrate that Cdk1 phosphorylates S439, both in vitro and in vivo. Our results suggest that Cdk1-mediated phosphorylation of S439 stabilizes mature SREBP1 during mitosis, thereby preserving a critical pool of active transcription factors to support lipid synthesis. Taken together with our previous work, the current study suggests that SREBP1 may provide a link between lipid synthesis, proliferation and cell growth. This hypothesis was supported by our observation that siRNA-mediated inactivation of SREBP1 arrested cells in the G(1) phase of the cell cycle, thereby attenuating cell growth.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B/metabolism , Mitosis , Sterol Regulatory Element Binding Protein 1/metabolism , Thermodynamics , Amino Acid Sequence , Cells, Cultured , Epitopes/immunology , G1 Phase , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Phosphoserine/metabolism , Protein Interaction Mapping , S Phase , Sterol Regulatory Element Binding Protein 1/chemistry , Sterol Regulatory Element Binding Protein 1/genetics , Transcription, Genetic
9.
J Biol Chem ; 281(35): 25278-86, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16825193

ABSTRACT

Members of the sterol regulatory element-binding protein (SREBP) family of transcription factors control cholesterol and lipid metabolism and play critical roles during adipocyte differentiation. The transcription factor SREBP1 is degraded by the ubiquitin-proteasome system following phosphorylation of Thr426 and Ser430 in its phosphodegron. We now demonstrate that the glycogen synthase kinase (GSK)-3beta-dependent phosphorylation of these residues in SREBP1 is enhanced in response to specific DNA binding. DNA binding enhances the direct interaction between the C-terminal domain of SREBP1 and GSK-3beta. Accordingly, we demonstrate that GSK-3beta is recruited to the promoters of SREBP target genes in vivo. As a result of the phosphorylation of Thr426 and Ser430, the ubiquitin ligase Fbw7 is recruited to SREBP molecules associated with target promoters. Using a reconstituted ubiquitination system, we demonstrate that Fbw7-mediated ubiquitination of SREBP1 is dependent on its DNA binding activity. Thus, DNA binding could provide a mechanistic link between the phosphorylation, ubiquitination, and degradation of active transcription factors.


Subject(s)
DNA/chemistry , Sterol Regulatory Element Binding Protein 1/chemistry , Ubiquitin/chemistry , Cell Line , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Lipid Metabolism , Mutation , Phosphorylation , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Binding
10.
Cell Metab ; 1(6): 379-91, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16054087

ABSTRACT

The sterol regulatory element binding protein (SREBP) family of transcription factors controls cholesterol and lipid metabolism. The nuclear forms of these proteins are rapidly degraded by the ubiquitin-proteasome pathway, but the signals and factors required for this are unknown. Here, we identify a phosphodegron in SREBP1a that serves as a recognition motif for the SCF(Fbw7) ubiquitin ligase. Fbw7 interacts with nuclear SREBP1a and enhances its ubiquitination and degradation in a manner dependent on the phosphorylation of T426 and S430 by GSK-3. Fbw7 also degrades nuclear SREBP1c and SREBP2, and inactivation of endogenous Fbw7 results in stabilization of nuclear SREBP1 and -2, enhanced expression of SREBP target genes, enhanced synthesis of cholesterol and fatty acids, and enhanced receptor-mediated uptake of LDL. Thus, our results suggest that Fbw7 may be a major regulator of lipid metabolism through control of the phosphorylation-dependent degradation of the SREBP family of transcription factors.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Lipid Metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , CCAAT-Enhancer-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Multigene Family , Phosphorylation , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Transcription Factors/genetics , Ubiquitin/metabolism
11.
Proc Natl Acad Sci U S A ; 102(33): 11681-6, 2005 Aug 16.
Article in English | MEDLINE | ID: mdl-16081540

ABSTRACT

The sterol regulatory element-binding protein (SREBP) family of transcription factors controls the biosynthesis of cholesterol and other lipids, and lipid synthesis is critical for cell growth and proliferation. We were, therefore, interested in the expression and activity of SREBPs during the cell cycle. We found that the expression of a number of SREBP-responsive promoter-reporter genes were induced in a SREBP-dependent manner in cells arrested in G2/M. In addition, the mature forms of SREBP1a and SREBP1c were hyperphosphorylated in mitotic cells, giving rise to a phosphoepitope recognized by the mitotic protein monoclonal-2 (MPM-2) antibody. In contrast, SREBP2 was not hyperphosphorylated in mitotic cells and was not recognized by the MPM-2 antibody. The MPM-2 epitope was mapped to the C terminus of mature SREBP1, and the mitosis-specific hyperphosphorylation of SREBP1 depended on this domain of the protein. The transcriptional and DNA-binding activity of SREBP1 was enhanced in cells arrested in G2/M, and these effects depended on the C-terminal domain of the protein. In part, these effects could be explained by our observation that mature SREBP1 was stabilized in G2/M. In agreement with these observations, we found that the synthesis of cholesterol was increased in G2/M-arrested cells. Thus, our results demonstrate that the activity of mature SREBP1 is regulated by phosphorylation during the cell cycle, suggesting that SREBP1 may provide a link between lipid synthesis, proliferation, and cell growth.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Mitosis , Transcription Factors/metabolism , CCAAT-Enhancer-Binding Proteins/chemistry , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Cholesterol/biosynthesis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , G2 Phase , Gene Expression Regulation , Genes, Reporter/genetics , Humans , Phosphorylation , Promoter Regions, Genetic/genetics , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic/genetics
12.
J Biol Chem ; 278(50): 49988-99, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14522984

ABSTRACT

The refolding of mitochondrial aspartate aminotransferase (mAAT; EC 2.6.1.1) has been studied following unfolding in 6 m guanidine hydrochloride for different periods of time. Whereas reactivation of equilibrium-unfolded mAAT is sigmoidal, reactivation of the short term unfolded protein displays a double exponential behavior consistent with the presence of fast and slow refolding species. The amplitude of the fast phase decreases with increasing unfolding times (k approximately 0.75 min(-1) at 20 degrees C) and becomes undetectable at equilibrium unfolding. According to hydrogen exchange and stopped-flow intrinsic fluorescence data, unfolding of mAAT appears to be complete in less than 10 s, but hydrolysis of the Schiff base linking the coenzyme pyridoxal 5'-phosphate (PLP) to the polypeptide is much slower (k approximately 0.08 min(-1)). This implies the existence in short term unfolded samples of unfolded species with PLP still attached. However, since the disappearance of the fast refolding phase is about 10-fold faster than the release of PLP, the fast refolding phase does not correspond to folding of the coenzyme-containing molecules. The fast refolding phase disappears more rapidly in the pyridoxamine and apoenzyme forms of mAAT, both of which lack covalently attached cofactor. Thus, bound PLP increases the kinetic stability of the fast refolding unfolding intermediates. Conversion between fast and slow folding forms also takes place in an early folding intermediate. The presence of cyclophilin has no effect on the reactivation of either equilibrium or short term unfolded mAAT. These results suggest that proline isomerization may not be the only factor determining the slow refolding of this cofactor-dependent protein.


Subject(s)
Aspartate Aminotransferases/chemistry , Animals , Aspartate Aminotransferases/metabolism , Cyclophilins/chemistry , Escherichia coli/metabolism , Guanidine/chemistry , Hydrogen-Ion Concentration , Kinetics , Liver/enzymology , Mass Spectrometry , Microscopy, Fluorescence , Models, Chemical , Peptides/chemistry , Proline/chemistry , Protein Binding , Protein Folding , Pyridoxal Phosphate/chemistry , Rats , Spectrometry, Fluorescence , Temperature , Time Factors , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...