Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Wiley Interdiscip Rev RNA ; : e1814, 2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37671427

ABSTRACT

The budding yeast, Saccharomyces cerevisiae, has been used for decades as a powerful genetic tool to study a broad spectrum of biological topics. With its ease of use, economic utility, well-studied genome, and a highly conserved proteome across eukaryotes, it has become one of the most used model organisms. Due to these advantages, it has been used to study an array of complex human diseases. From broad, complex pathological conditions such as aging and neurodegenerative disease to newer uses such as SARS-CoV-2, yeast continues to offer new insights into how cellular processes are affected by disease and how affected pathways might be targeted in therapeutic settings. At the same time, the roles of RNA and RNA-based processes have become increasingly prominent in the pathology of many of these same human diseases, and yeast has been utilized to investigate these mechanisms, from aberrant RNA-binding proteins in amyotrophic lateral sclerosis to translation regulation in cancer. Here we review some of the important insights that yeast models have yielded into the molecular pathology of complex, RNA-based human diseases. This article is categorized under: RNA in Disease and Development > RNA in Disease.

2.
G3 (Bethesda) ; 13(1)2023 01 12.
Article in English | MEDLINE | ID: mdl-36409020

ABSTRACT

During cellular stress it is essential for cells to alter their gene expression to adapt and survive. Gene expression is regulated at multiple levels, but translation regulation is both a method for rapid changes to the proteome and, as one of the most energy-intensive cellular processes, a way to efficiently redirect cellular resources during stress conditions. Despite this ideal positioning, many of the specifics of how translation is regulated, positively or negatively, during various types of cellular stress remain poorly understood. To further assess this regulation, we examined the essential translation factor Ded1, an RNA helicase that has been previously shown to play important roles in the translational response to cellular stress. In particular, ded1 mutants display an increased resistance to growth inhibition and translation repression induced by the TOR pathway inhibitor, rapamycin, suggesting that normal stress responses are partially defective in these mutants. To gain further insight into Ded1 translational regulation during stress, synthetic genetic array analysis was conducted in the presence of rapamycin with a ded1 mutant and a library of nonessential genes in Saccharomyces cerevisiae to identify positive and negative genetic interactions in an unbiased manner. Here, we report the results of this screen and subsequent network mapping and Gene Ontology-term analysis. Hundreds of candidate interactions were identified, which fell into expected categories, such as ribosomal proteins and amino acid biosynthesis, as well as unexpected ones, including membrane trafficking, sporulation, and protein glycosylation. Therefore, these results provide several specific directions for further comprehensive studies.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomycetales , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Saccharomycetales/genetics , Protein Biosynthesis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
3.
Methods Enzymol ; 673: 103-140, 2022.
Article in English | MEDLINE | ID: mdl-35965004

ABSTRACT

Stress is inevitable, so all organisms have developed response mechanisms to allow for their survival during times of stress. Regulation of gene expression is a critical part of these responses, which allows for the appropriate cohort of proteins to be produced to counter the stress while downregulating others in order to conserve resources. Translation is both highly energy intensive and able to rapidly shift the proteome, thus making it a key target for regulation during stress. Numerous stress pathways converge on translation, and examining the regulatory mechanisms that underlie these pathways is essential for understanding the initial and long-term effects of stress on cells. A number of RNA helicases, including eIF4A, Ded1/DDX3X, and Dhh1/DDX6, have been previously linked to translation, and given their ability to dramatically alter RNA-protein interactions, they are well-positioned to play critical roles in translation regulation during stress. Therefore, assessing the role of helicases in these conditions is vital to the overall understanding of stress. Outlined below are key assays focusing on two areas: assessing cellular phenotypes in growth and survival during stress conditions, and analyzing cellular translation in the presence and absence of stress. The combination of these two approaches will begin to establish the function(s) of a given helicase in the overall stress response.


Subject(s)
DNA Helicases , RNA Helicases , Cell Cycle , DNA Helicases/genetics , DNA Helicases/metabolism , Humans , Protein Processing, Post-Translational , Stress, Physiological
4.
Mol Cell Biol ; 42(1): e0024421, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34723653

ABSTRACT

Ded1 is a conserved RNA helicase that promotes translation initiation in steady-state conditions. Ded1 has also been shown to regulate translation during cellular stress and affect the dynamics of stress granules (SGs), accumulations of RNA and protein linked to translation repression. To better understand its role in stress responses, we examined Ded1 function in two different models: DED1 overexpression and oxidative stress. DED1 overexpression inhibits growth and promotes the formation of SGs. A ded1 mutant lacking the low-complexity C-terminal region (ded1-ΔCT), which mediates Ded1 oligomerization and interaction with the translation factor eIF4G1, suppressed these phenotypes, consistent with other stresses. During oxidative stress, a ded1-ΔCT mutant was defective in growth and in SG formation compared to wild-type cells, although SGs were increased rather than decreased in these conditions. Unlike stress induced by direct TOR inhibition, the phenotypes in both models were only partially dependent on eIF4G1 interaction, suggesting an additional contribution from Ded1 oligomerization. Furthermore, examination of the growth defects and translational changes during oxidative stress suggested that Ded1 plays a role during recovery from stress. Integrating these disparate results, we propose that Ded1 controls multiple aspects of translation and RNP dynamics in both initial stress responses and during recovery.


Subject(s)
DEAD-box RNA Helicases/metabolism , RNA Helicases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Cytoplasmic Granules/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Fungal/genetics , Protein Biosynthesis/physiology , RNA, Messenger/genetics , Saccharomyces cerevisiae/metabolism , Stress, Physiological/physiology
6.
J Biol Chem ; 296: 100296, 2021.
Article in English | MEDLINE | ID: mdl-33460649

ABSTRACT

Medulloblastoma is the most common pediatric brain cancer, and sequencing studies identified frequent mutations in DDX3X, a DEAD-box RNA helicase primarily implicated in translation. Forty-two different sites were identified, suggesting that the functional effects of the mutations are complex. To investigate how these mutations are affecting DDX3X cellular function, we constructed a full set of equivalent mutant alleles in DED1, the Saccharomyces cerevisiae ortholog of DDX3X, and characterized their effects in vivo and in vitro. Most of the medulloblastoma-associated mutants in DDX3X/DED1 (ded1-mam) showed substantial growth defects, indicating that functional effects are conserved in yeast. Further, while translation was affected in some mutants, translation defects affecting bulk mRNA were neither consistent nor correlated with the growth phenotypes. Likewise, increased formation of stress granules in ded1-mam mutants was common but did not correspond to the severity of the mutants' growth defects. In contrast, defects in translating mRNAs containing secondary structure in their 5' untranslated regions (UTRs) were found in almost all ded1-mam mutants and correlated well with growth phenotypes. We thus conclude that these specific translation defects, rather than generalized effects on translation, are responsible for the observed cellular phenotypes and likely contribute to DDX3X-mutant medulloblastoma. Examination of ATPase activity and RNA binding of recombinant mutant proteins also did not reveal a consistent defect, indicating that the translation defects are derived from multiple enzymatic deficiencies. This work suggests that future studies into medulloblastoma pathology should focus on this specific translation defect, while taking into account the wide spectrum of DDX3X mutations.


Subject(s)
Cerebellar Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Medulloblastoma/genetics , Protein Biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , 5' Untranslated Regions , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Substitution , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Conserved Sequence , DEAD-box RNA Helicases/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mutagenesis, Site-Directed , Mutation , Phenotype , Protein Binding , RNA/genetics , RNA/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Red Fluorescent Protein
7.
Biomolecules ; 10(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992901

ABSTRACT

Stress granules (SGs) are hypothesized to facilitate TAR DNA-binding protein 43 (TDP-43) cytoplasmic mislocalization and aggregation, which may underly amyotrophic lateral sclerosis pathology. However, much data for this hypothesis is indirect. Additionally, whether P-bodies (PBs; related mRNA-protein granules) affect TDP-43 phenotypes is unclear. Here, we determine that induction of TDP-43 expression in yeast results in the accumulation of SG-like foci that in >90% of cases become the sites where TDP-43 cytoplasmic foci first appear. Later, TDP-43 foci associate less with SGs and more with PBs, though independent TDP-43 foci also accumulate. However, depleting or over-expressing yeast SG and PB proteins reveals no consistent trend between SG or PB assembly and TDP-43 foci formation, toxicity or protein abundance. In human cells, immunostaining endogenous TDP-43 with different TDP-43 antibodies reveals distinct localization and aggregation behaviors. Following acute arsenite stress, all phospho-TDP-43 foci colocalize with SGs. Finally, formation of TDP-43 cytoplasmic foci following low-dose chronic arsenite stress is impaired, but not completely blocked, in G3BP1/2ΔΔ cells. Collectively, our data suggest that SG and PB assembly may facilitate TDP-43 cytoplasmic localization and aggregation but are likely not essential for these events.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cytoplasmic Granules/genetics , DNA-Binding Proteins/genetics , Stress, Physiological/genetics , Amyotrophic Lateral Sclerosis/pathology , Cytoplasm/genetics , Humans , Protein Aggregates/genetics , RNA, Messenger/genetics
8.
Mol Biol Cell ; 30(17): 2171-2184, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141444

ABSTRACT

Ded1 is a DEAD-box RNA helicase with essential roles in translation initiation. It binds to the eukaryotic initiation factor 4F (eIF4F) complex and promotes 48S preinitiation complex assembly and start-site scanning of 5' untranslated regions of mRNAs. Most prior studies of Ded1 cellular function were conducted in steady-state conditions during nutrient-rich growth. In this work, however, we examine its role in the translational response during target of rapamycin (TOR)C1 inhibition and identify a novel function of Ded1 as a translation repressor. We show that C-terminal mutants of DED1 are defective in down-regulating translation following TORC1 inhibition using rapamycin. Furthermore, following TORC1 inhibition, eIF4G1 normally dissociates from translation complexes and is degraded, and this process is attenuated in mutant cells. Mapping of the functional requirements for Ded1 in this translational response indicates that Ded1 enzymatic activity and interaction with eIF4G1 are required, while homo-oligomerization may be dispensable. Our results are consistent with a model wherein Ded1 stalls translation and specifically removes eIF4G1 from translation preinitiation complexes, thus removing eIF4G1 from the translating mRNA pool and leading to the codegradation of both proteins. Shared features among DED1 orthologues suggest that this role is conserved and may be implicated in pathologies such as oncogenesis.


Subject(s)
DEAD-box RNA Helicases/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , 5' Untranslated Regions , Cytoplasm/metabolism , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4F/metabolism , Nucleic Acid Conformation , Peptide Chain Elongation, Translational , Protein Biosynthesis , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
9.
Mol Cell Biol ; 37(21)2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28784717

ABSTRACT

DEAD-box proteins (DBPs) are required in gene expression to facilitate changes to ribonucleoprotein complexes, but the cellular mechanisms and regulation of DBPs are not fully defined. Gle1 is a multifunctional regulator of DBPs with roles in mRNA export and translation. In translation, Gle1 modulates Ded1, a DBP required for initiation. However, DED1 overexpression causes defects, suggesting that Ded1 can promote or repress translation in different contexts. Here we show that GLE1 expression suppresses the repressive effects of DED1 in vivo and Gle1 counteracts Ded1 in translation assays in vitro Furthermore, both Ded1 and Gle1 affect the assembly of preinitiation complexes. Through mutation analysis and binding assays, we show that Gle1 inhibits Ded1 by reducing its affinity for RNA. Our results are consistent with a model wherein active Ded1 promotes translation but inactive or excess Ded1 leads to translation repression. Gle1 can inhibit either role of Ded1, positioning it as a gatekeeper to optimize Ded1 activity to the appropriate level for translation. This study suggests a paradigm for finely controlling the activity of DEAD-box proteins to optimize their function in RNA-based processes. It also positions the versatile regulator Gle1 as a potential node for the coordination of different steps of gene expression.

10.
J Biol Chem ; 286(46): 39750-9, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21949122

ABSTRACT

DEAD-box protein (Dbp) family members are essential for gene expression; however, their precise roles and regulation are not fully defined. During messenger (m)RNA export, Gle1 bound to inositol hexakisphosphate (IP(6)) acts via Dbp5 to facilitate remodeling of mRNA-protein complexes. In contrast, here we define a novel Gle1 role in translation initiation through regulation of a different DEAD-box protein, the initiation factor Ded1. We find that Gle1 physically and genetically interacts with Ded1. Surprisingly, whereas Gle1 stimulates Dbp5, it inhibits Ded1 ATPase activity in vitro, and IP(6) does not affect this inhibition. Functionally, a gle1-4 mutant specifically suppresses initiation defects in a ded1-120 mutant, and ded1 and gle1 mutants have complementary perturbations in AUG start site recognition. Consistent with this role in initiation, Gle1 inhibits translation in vitro in competent extracts. These results indicate that Gle1 has a direct role in initiation and negatively regulates Ded1. Together, the differential regulation of two distinct DEAD-box proteins by a common factor (Gle1) establishes a new paradigm for controlling gene expression and coupling translation with mRNA export.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Fungal/physiology , Nuclear Pore Complex Proteins/metabolism , Peptide Chain Initiation, Translational/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport, Active/physiology , DEAD-box RNA Helicases/genetics , Mutation , Nuclear Pore Complex Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
11.
J Biol Chem ; 285(22): 16683-92, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20371601

ABSTRACT

The unidirectional translocation of messenger RNA (mRNA) through the aqueous channel of the nuclear pore complex (NPC) is mediated by interactions between soluble mRNA export factors and distinct binding sites on the NPC. At the cytoplasmic side of the NPC, the conserved mRNA export factors Gle1 and inositol hexakisphosphate (IP(6)) play an essential role in mRNA export by activating the ATPase activity of the DEAD-box protein Dbp5, promoting localized messenger ribonucleoprotein complex remodeling, and ensuring the directionality of the export process. In addition, Dbp5, Gle1, and IP(6) are also required for proper translation termination. However, the specificity of the IP(6)-Gle1 interaction in vivo is unknown. Here, we characterize the biochemical interaction between Gle1 and IP(6) and the relationship to Dbp5 binding and stimulation. We identify Gle1 residues required for IP(6) binding and show that these residues are needed for IP(6)-dependent Dbp5 stimulation in vitro. Furthermore, we demonstrate that Gle1 is the primary target of IP(6) for both mRNA export and translation termination in vivo. In Saccharomyces cerevisiae cells, the IP(6)-binding mutants recapitulate all of the mRNA export and translation termination defects found in mutants depleted of IP(6). We conclude that Gle1 specifically binds IP(6) and that this interaction is required for the full potentiation of Dbp5 ATPase activity during both mRNA export and translation termination.


Subject(s)
Nuclear Pore Complex Proteins/chemistry , Phytic Acid/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Biological Transport , DEAD-box RNA Helicases/metabolism , Glutathione Transferase/metabolism , Inositol/metabolism , Kinetics , Models, Biological , Molecular Sequence Data , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Protein Binding , Protein Biosynthesis , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism
12.
Cell ; 134(4): 624-33, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18724935

ABSTRACT

Gene expression requires proper messenger RNA (mRNA) export and translation. However, the functional links between these consecutive steps have not been fully defined. Gle1 is an essential, conserved mRNA export factor whose export function is dependent on the small molecule inositol hexakisphosphate (IP(6)). Here, we show that both Gle1 and IP(6) are required for efficient translation termination in Saccharomyces cerevisiae and that Gle1 interacts with termination factors. In addition, Gle1 has a conserved physical association with the initiation factor eIF3, and gle1 mutants display genetic interactions with the eIF3 mutant nip1-1. Strikingly, gle1 mutants have defects in initiation, whereas strains lacking IP(6) do not. We propose that Gle1 functions together with IP(6) and the DEAD-box protein Dbp5 to regulate termination. However, Gle1 also independently mediates initiation. Thus, Gle1 is uniquely positioned to coordinate the mRNA export and translation mechanisms. These results directly impact models for perturbation of Gle1 function in pathophysiology.


Subject(s)
Carrier Proteins/metabolism , Phytic Acid/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-3/metabolism , Nuclear Pore Complex Proteins , Nucleocytoplasmic Transport Proteins/metabolism , Peptide Termination Factors/metabolism , Protein Isoforms/metabolism , Saccharomyces cerevisiae/genetics
14.
J Biol Chem ; 282(40): 29186-92, 2007 Oct 05.
Article in English | MEDLINE | ID: mdl-17646162

ABSTRACT

Ataxin-1 is a neurodegenerative disorder protein whose mutant form causes spinocerebellar ataxia type-1 (SCA1). Evidence suggests that ataxin-1 may function as a transcription repressor. However, neither the importance of this putative transcriptional repression activity in neural cytotoxicity nor the transcriptional targets of ataxin-1 are known. Here we identify the MEF2-HDAC4 transcriptional complex involved in neuron survival as a target of ataxin-1. We show that ataxin-1 binds specifically to histone deacetylase-4 (HDAC4) and MEF2 and colocalizes with them in nuclear inclusion bodies. Significantly, these interactions are greatly reduced by the S776A mutation, which largely abrogates the cytotoxicity of ataxin-1. Supporting the importance of these interactions, we show that wild type ataxin-1 represses MEF2-dependent transcription, whereas the S776A mutant is less potent. Furthermore, overexpression of MEF2 can partially reverse cytotoxicity caused by ataxin-1. Our results identify the MEF2-HDAC4 complex as a target for ataxin-1 transcriptional repression activity and suggest a novel pathogenic mechanism whereby ataxin-1 sequesters and inhibits the neuronal survival factor MEF2.


Subject(s)
Myogenic Regulatory Factors/metabolism , Nerve Tissue Proteins/physiology , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Nuclear Proteins/physiology , Animals , Ataxin-1 , Ataxins , COS Cells , Cerebellum/cytology , Chlorocebus aethiops , Histone Deacetylases/metabolism , MEF2 Transcription Factors , Mice , Microscopy, Fluorescence , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding , Protein Transport , Transfection
15.
J Neurosci ; 25(41): 9544-53, 2005 Oct 12.
Article in English | MEDLINE | ID: mdl-16221865

ABSTRACT

Histone deacetylase 4 (HDAC4) undergoes signal-dependent shuttling between the cytoplasm and nucleus, which is regulated in part by calcium/calmodulin-dependent kinase (CaMK)-mediated phosphorylation. Here, we report that HDAC4 intracellular trafficking is important in regulating neuronal cell death. HDAC4 is normally localized to the cytoplasm in brain tissue and cultured cerebellar granule neurons (CGNs). However, in response to low-potassium or excitotoxic glutamate conditions that induce neuronal cell death, HDAC4 rapidly translocates into the nucleus of cultured CGNs. Treatment with the neuronal survival factor BDNF suppresses HDAC4 nuclear translocation, whereas a proapoptotic CaMK inhibitor stimulates HDAC4 nuclear accumulation. Moreover, ectopic expression of nuclear-localized HDAC4 promotes neuronal apoptosis and represses the transcriptional activities of myocyte enhancer factor 2 and cAMP response element-binding protein, survival factors in neurons. In contrast, inactivation of HDAC4 by small interfering RNA or HDAC inhibitors suppresses neuronal cell death. Finally, an increase of nuclear HDAC4 in granule neurons is also observed in weaver mice, which harbor a mutation that promotes CGN apoptosis. Our data identify HDAC4 and its intracellular trafficking as key effectors of multiple pathways that regulate neuronal cell death.


Subject(s)
Histone Deacetylases/biosynthesis , Intracellular Fluid/enzymology , Neurons/enzymology , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histone Deacetylases/genetics , Intracellular Fluid/drug effects , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Neurons/cytology , Neurons/drug effects , Protein Transport/drug effects , Protein Transport/physiology
16.
Mol Cell Biol ; 25(19): 8456-64, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166628

ABSTRACT

The class II deacetylase histone deacetylase 4 (HDAC4) negatively regulates the transcription factor MEF2. HDAC4 is believed to repress MEF2 transcriptional activity by binding to MEF2 and catalyzing local histone deacetylation. Here we report that HDAC4 also controls MEF2 by a novel SUMO E3 ligase activity. We show that HDAC4 interacts with the SUMO E2 conjugating enzyme Ubc9 and is itself sumoylated. The overexpression of HDAC4 leads to prominent MEF2 sumoylation in vivo, whereas recombinant HDAC4 stimulates MEF2 sumoylation in a reconstituted system in vitro. Importantly, HDAC4 promotes sumoylation on a lysine residue that is also subject to acetylation by a MEF2 coactivator, the acetyltransferase CBP, suggesting a possible interplay between acetylation and sumoylation in regulating MEF2 activity. Indeed, MEF2 acetylation is correlated with MEF2 activation and dynamically induced upon muscle cell differentiation, while sumoylation inhibits MEF2 transcriptional activity. Unexpectedly, we found that HDAC4 does not function as a MEF2 deacetylase. Instead, the NAD+-dependent deacetylase SIRT1 can potently induce MEF2 deacetylation. Our studies reveal a novel regulation of MEF2 transcriptional activity by two distinct classes of deacetylases that affect MEF2 sumoylation and acetylation.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Lysine/chemistry , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Repressor Proteins/metabolism , Sirtuins/metabolism , Acetylation , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation , Cell Line , DNA/chemistry , Fibroblasts/metabolism , Genetic Vectors , Humans , Immunoprecipitation , Luciferases/metabolism , MEF2 Transcription Factors , Mice , Models, Biological , Molecular Sequence Data , Mutation , Myogenic Regulatory Factors/chemistry , Plasmids/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sirtuin 1 , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic , Transfection , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...