Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biol Open ; 12(11)2023 11 15.
Article in English | MEDLINE | ID: mdl-37850739

ABSTRACT

The chromatin environment has a significant impact on gene expression. Chromatin structure is highly regulated by histone modifications and RNA polymerase II binding dynamics. The SIN3 histone modifying complex regulates the chromatin environment leading to changes in gene expression. In Drosophila melanogaster, the Sin3A gene is alternatively spliced to produce different protein isoforms, two of which include SIN3 220 and SIN3 187. Both SIN3 isoforms are scaffolding proteins that interact with several other factors to regulate the chromatin landscape. The mechanism through which the SIN3 isoforms regulate chromatin is not well understood. Here, we analyze publicly available data sets to allow us to ask specific questions on how SIN3 isoforms regulate chromatin and gene activity. We determined that genes repressed by the SIN3 isoforms exhibited enrichment in histone H3K4me2, H3K4me3, H3K14ac and H3K27ac near the transcription start site. We observed an increase in the amount of paused RNA polymerase II on the promoter of genes repressed by the isoforms as compared to genes that require SIN3 for maximum activation. Furthermore, we analyzed a subset of genes regulated by SIN3 187 that suggest a mechanism in which SIN3 187 might exhibit hard regulation as well as soft regulation. Data presented here expand our knowledge of how the SIN3 isoforms regulate the chromatin environment and RNA polymerase II binding dynamics.


Subject(s)
Chromatin , Histones , Animals , Chromatin/genetics , Chromatin/metabolism , Histones/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
2.
Enzymes ; 53: 33-68, 2023.
Article in English | MEDLINE | ID: mdl-37748836

ABSTRACT

Post-translational modifications of histone proteins control the expression of genes. Metabolites from central and one-carbon metabolism act as donor moieties to modify histones and regulate gene expression. Thus, histone modification and gene regulation are connected to the metabolite status of the cell. Histone modifiers, such as the SIN3 complex, regulate genes involved in proliferation and metabolism. The SIN3 complex contains a histone deacetylase and a histone demethylase, which regulate the chromatin landscape and gene expression. In this chapter, we review the cross-talk between metabolic pathways that produce donor moieties, and epigenetic complexes regulating proliferation and metabolic genes. This cross-talk between gene regulation and metabolism is tightly controlled, and disruption of this cross-talk leads to metabolic diseases. We discuss promising therapeutics that directly regulate histone modifiers, and can affect the metabolic status of the cell, alleviating some metabolic diseases.


Subject(s)
Epigenesis, Genetic , Histones , Chromatin , Histone Deacetylases , Sin3 Histone Deacetylase and Corepressor Complex , Humans , Animals
3.
Nat Commun ; 13(1): 6548, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36319643

ABSTRACT

Aberrant expression of the Forkhead box transcription factor, FOXQ1, is a prevalent mechanism of epithelial-mesenchymal transition (EMT) and metastasis in multiple carcinoma types. However, it remains unknown how FOXQ1 regulates gene expression. Here, we report that FOXQ1 initiates EMT by recruiting the MLL/KMT2 histone methyltransferase complex as a transcriptional coactivator. We first establish that FOXQ1 promoter recognition precedes MLL complex assembly and histone-3 lysine-4 trimethylation within the promoter regions of critical genes in the EMT program. Mechanistically, we identify that the Forkhead box in FOXQ1 functions as a transactivation domain directly binding the MLL core complex subunit RbBP5 without interrupting FOXQ1 DNA binding activity. Moreover, genetic disruption of the FOXQ1-RbBP5 interaction or pharmacologic targeting of KMT2/MLL recruitment inhibits FOXQ1-dependent gene expression, EMT, and in vivo tumor progression. Our study suggests that targeting the FOXQ1-MLL epigenetic axis could be a promising strategy to combat triple-negative breast cancer metastatic progression.


Subject(s)
Breast Neoplasms , Neoplasms, Second Primary , Female , Humans , Breast Neoplasms/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms, Second Primary/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Melanoma, Cutaneous Malignant
4.
Biochim Biophys Acta Mol Cell Res ; 1869(10): 119322, 2022 10.
Article in English | MEDLINE | ID: mdl-35820484

ABSTRACT

The SIN3 scaffolding protein is a conserved transcriptional regulator known to fine-tune gene expression. In Drosophila, there are two major isoforms of SIN3, SIN3 220 and SIN3 187, which each assemble into multi-subunit histone modifying complexes. The isoforms have distinct developmental expression patterns and non-redundant functions. Gene regulatory network analyses indicate that both isoforms affect genes encoding proteins in pathways such as the cell cycle and cell morphogenesis. Interestingly, the SIN3 187 isoform uniquely regulates a subset of pathways including post-embryonic development, phosphate metabolism and apoptosis. Target genes in the phosphate metabolism pathway include nuclear-encoded mitochondrial genes coding for proteins responsible for oxidative phosphorylation. Here, we investigate the physiological effects of SIN3 isoforms on energy metabolism and cell survival. We find that ectopic expression of SIN3 187 represses expression of several nuclear-encoded mitochondrial genes affecting production of ATP and generation of reactive oxygen species (ROS). Forced expression of SIN3 187 also activates several pro-apoptotic and represses a few anti-apoptotic genes. In the SIN3 187 expressing cells, these gene expression patterns are accompanied with an increased sensitivity to paraquat-mediated oxidative stress. These findings indicate that SIN3 187 influences the regulation of mitochondrial function, apoptosis and oxidative stress response in ways that are dissimilar from SIN3 220. The data suggest that the distinct SIN3 histone modifying complexes are deployed in different cellular contexts to maintain cellular homeostasis.


Subject(s)
Drosophila Proteins , Animals , Cell Survival/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Energy Metabolism/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Phosphates/metabolism , Protein Isoforms/genetics , Sin3 Histone Deacetylase and Corepressor Complex/metabolism
5.
Bioessays ; 43(2): e2000231, 2021 02.
Article in English | MEDLINE | ID: mdl-33215731

ABSTRACT

Pleiotropically acting eukaryotic corepressors such as retinoblastoma and SIN3 have been found to physically interact with many widely expressed "housekeeping" genes. Evidence suggests that their roles at these loci are not to provide binary on/off switches, as is observed at many highly cell-type specific genes, but rather to serve as governors, directly modulating expression within certain bounds, while not shutting down gene expression. This sort of regulation is challenging to study, as the differential expression levels can be small. We hypothesize that depending on context, corepressors mediate "soft repression," attenuating expression in a less dramatic but physiologically appropriate manner. Emerging data indicate that such regulation is a pervasive characteristic of most eukaryotic systems, and may reflect the mechanistic differences between repressor action at promoter and enhancer locations. Soft repression may represent an essential component of the cybernetic systems underlying metabolic adaptations, enabling modest but critical adjustments on a continual basis.


Subject(s)
Repressor Proteins , Transcription, Genetic , Gene Expression Regulation , Histone Deacetylases/genetics , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism
6.
J Biol Chem ; 295(2): 375-389, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31776190

ABSTRACT

The SIN3 histone-modifying complex regulates the expression of multiple methionine catabolic genes, including SAM synthetase (Sam-S), as well as SAM levels. To further dissect the relationship between methionine catabolism and epigenetic regulation by SIN3, we sought to identify genes and metabolic pathways controlled by SIN3 and SAM synthetase (SAM-S) in Drosophila melanogaster Using several approaches, including RNAi-mediated gene silencing, RNA-Seq- and quantitative RT-PCR-based transcriptomics, and ultra-high-performance LC-MS/MS- and GC/MS-based metabolomics, we found that, as a global transcriptional regulator, SIN3 impacted a wide range of genes and pathways. In contrast, SAM-S affected only a narrow range of genes and pathways. The expression and levels of additional genes and metabolites, however, were altered in Sin3A+Sam-S dual knockdown cells. This analysis revealed that SIN3 and SAM-S regulate overlapping pathways, many of which involve one-carbon and central carbon metabolisms. In some cases, the factors acted independently; in some others, redundantly; and for a third set, in opposition. Together, these results, obtained from experiments with the chromatin regulator SIN3 and the metabolic enzyme SAM-S, uncover a complex relationship between metabolism and epigenetic regulation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Methionine Adenosyltransferase/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epigenesis, Genetic , Gene Regulatory Networks , Metabolome , Methionine Adenosyltransferase/genetics , RNA Interference , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Transcriptional Activation
7.
Sci Rep ; 8(1): 17048, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451916

ABSTRACT

Establishment and maintenance of histone acetylation levels are critical for metazoan development and viability. Disruption of the balance between acetylation and deacetylation by treatment with chemical histone deacetylase (HDAC) inhibitors results in loss of cell proliferation, differentiation and/or apoptosis. Histone deacetylation by the SIN3 complex is essential in Drosophila and mice, as loss of the scaffolding factor SIN3 or the associated HDAC results in lethality. The objective of this study is to elucidate contributions of SIN3 complex components to these essential processes. We used the Drosophila model organism to carry out a systematic functional analysis of the SIN3 complex. We find that SIN3 associated proteins are essential for viability and cell proliferation during development. Additionally, tissue specific reduction of SIN3 complex components results in abnormal wing development. Interestingly, while knockdown of each factor resulted in similar phenotypes, their individual effects on recruitment of SIN3 to polytene chromosomes are distinct. Reduction of some factors leads to large changes in the morphology of the chromosome and/or greatly reduced SIN3 binding. These findings suggest that while individual SIN3 complex components work through distinct molecular mechanisms, they each make a substantial contribution to the overall function of this highly conserved histone deacetylase complex.


Subject(s)
Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Animals , Cell Proliferation , Cell Survival , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Histones/metabolism , Male
8.
Epigenetics Chromatin ; 11(1): 17, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29665841

ABSTRACT

SIN3 is a global transcriptional coregulator that governs expression of a large repertoire of gene targets. It is an important player in gene regulation, which can repress or activate diverse gene targets in a context-dependent manner. SIN3 is required for several vital biological processes such as cell proliferation, energy metabolism, organ development, and cellular senescence. The functional flexibility of SIN3 arises from its ability to interact with a large variety of partners through protein interaction domains that are conserved across species, ranging from yeast to mammals. Several isoforms of SIN3 are present in these different species that can perform common and specialized functions through interactions with distinct enzymes and DNA-binding partners. Although SIN3 has been well studied due to its wide-ranging functions and highly conserved interaction domains, precise roles of individual SIN3 isoforms have received less attention. In this review, we discuss the differences in structure and function of distinct SIN3 isoforms and provide possible avenues to understand the complete picture of regulation by SIN3.


Subject(s)
Sin3 Histone Deacetylase and Corepressor Complex/chemistry , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Transcription, Genetic , Animals , Conserved Sequence , Gene Expression Regulation , Humans , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Saccharomyces cerevisiae/metabolism
9.
J Biol Chem ; 292(5): 1970-1976, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28028175

ABSTRACT

Chromatin modification and cellular metabolism are tightly connected. Chromatin modifiers regulate the expression of genes involved in metabolism and, in turn, the levels of metabolites. The generated metabolites are utilized by chromatin modifiers to affect epigenetic modification. The mechanism for this cross-talk, however, remains incompletely understood. The corepressor SIN3 controls histone acetylation through association with the histone deacetylase RPD3. The SIN3 complex is known to regulate genes involved in a number of metabolic processes. Here, we find that Drosophila SIN3 binds to the promoter region of genes involved in methionine catabolism and that this binding affects histone modification, which in turn influences gene expression. Specifically, we observe that reduced expression of SIN3 leads to an increase in S-adenosylmethionine (SAM), which is the major cellular donor of methyl groups for protein modification. Additionally, Sin3A knockdown results in an increase in global histone H3K4me3 levels. Furthermore, decreased H3K4me3 caused by knockdown of either SAM synthetase (Sam-S) or the histone methyltransferase Set1 is restored to near normal levels when SIN3 is also reduced. Taken together, these results indicate that knockdown of Sin3A directly alters the expression of methionine metabolic genes to increase SAM, which in turn leads to an increase in global H3K4me3. Our study reveals that SIN3 is an important epigenetic regulator directly connecting methionine metabolism and histone modification.


Subject(s)
Drosophila Proteins/metabolism , Epigenesis, Genetic/physiology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methionine/metabolism , Repressor Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Histone-Lysine N-Methyltransferase/genetics , Methionine/genetics , Methylation , Repressor Proteins/genetics , Sin3 Histone Deacetylase and Corepressor Complex
10.
J Biol Chem ; 291(22): 11566-71, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27129248

ABSTRACT

SIN3 is a transcriptional corepressor that acts as a scaffold for a histone deacetylase (HDAC) complex. The SIN3 complex regulates various biological processes, including organ development, cell proliferation, and energy metabolism. Little is known, however, about the regulation of SIN3 itself. There are two major isoforms of Drosophila SIN3, 187 and 220, which are differentially expressed. Intrigued by the developmentally timed exchange of SIN3 isoforms, we examined whether SIN3 187 controls the fate of the 220 counterpart. Here, we show that in developing tissue, there is interplay between SIN3 isoforms: when SIN3 187 protein levels increase, SIN3 220 protein decreases concomitantly. SIN3 187 has a dual effect on SIN3 220. Expression of 187 leads to reduced 220 transcript, while also increasing the turnover of SIN3 220 protein by the proteasome. These data support the presence of a novel, inter-isoform-dependent mechanism that regulates the amount of SIN3 protein, and potentially the level of specific SIN3 complexes, during distinct developmental stages.


Subject(s)
Drosophila/metabolism , Gene Expression Regulation, Developmental , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Western , Cells, Cultured , Drosophila/growth & development , Immunoenzyme Techniques , Protein Isoforms , Proteolysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sin3 Histone Deacetylase and Corepressor Complex
11.
BMC Genomics ; 17: 111, 2016 Feb 13.
Article in English | MEDLINE | ID: mdl-26872827

ABSTRACT

BACKGROUND: The multisubunit SIN3 complex is a global transcriptional regulator. In Drosophila, a single Sin3A gene encodes different isoforms of SIN3, of which SIN3 187 and SIN3 220 are the major isoforms. Previous studies have demonstrated functional non-redundancy of SIN3 isoforms. The role of SIN3 isoforms in regulating distinct biological processes, however, is not well characterized. RESULTS: We established a Drosophila S2 cell culture model system in which cells predominantly express either SIN3 187 or SIN3 220. To identify genomic targets of SIN3 isoforms, we performed chromatin immunoprecipitation followed by deep sequencing. Our data demonstrate that upon overexpression of SIN3 187, the level of SIN3 220 decreased and the large majority of genomic sites bound by SIN3 220 were instead bound by SIN3 187. We used RNA-seq to identify genes regulated by the expression of one isoform or the other. In S2 cells, which predominantly express SIN3 220, we found that SIN3 220 directly regulates genes involved in metabolism and cell proliferation. We also determined that SIN3 187 regulates a unique set of genes and likely modulates expression of many genes also regulated by SIN3 220. Interestingly, biological pathways enriched for genes specifically regulated by SIN3 187 strongly suggest that this isoform plays an important role during the transition from the embryonic to the larval stage of development. CONCLUSION: These data establish the role of SIN3 isoforms in regulating distinct biological processes. This study substantially contributes to our understanding of the complexity of gene regulation by SIN3.


Subject(s)
Gene Expression Regulation , Genome-Wide Association Study , Histone Deacetylases/metabolism , Signal Transduction , Animals , Binding Sites , Drosophila/genetics , Drosophila/metabolism , Gene Expression Profiling , Histone Deacetylases/genetics , Models, Biological , Protein Binding , Protein Isoforms , Transcriptome
12.
Article in English | MEDLINE | ID: mdl-26848313

ABSTRACT

BACKGROUND: Regulation of gene expression by histone-modifying enzymes is essential to control cell fate decisions and developmental processes. Two histone-modifying enzymes, RPD3, a deacetylase, and dKDM5/LID, a demethylase, are present in a single complex, coordinated through the SIN3 scaffold protein. While the SIN3 complex has been demonstrated to have functional histone deacetylase activity, the role of the demethylase dKDM5/LID as part of the complex has not been investigated. RESULTS: Here, we analyzed the developmental and transcriptional activities of dKDM5/LID in relation to SIN3. Knockdown of either Sin3A or lid resulted in decreased cell proliferation in S2 cells and wing imaginal discs. Conditional knockdown of either Sin3A or lid resulted in flies that displayed wing developmental defects. Interestingly, overexpression of dKDM5/LID rescued the wing developmental defect due to reduced levels of SIN3 in female flies, indicating a major role for dKDM5/LID in cooperation with SIN3 during development. Together, these observed phenotypes strongly suggest that dKDM5/LID as part of the SIN3 complex can impact previously uncharacterized transcriptional networks. Transcriptome analysis revealed that SIN3 and dKDM5/LID regulate many common genes. While several genes implicated in cell cycle and wing developmental pathways were affected upon altering the level of these chromatin factors, a significant affect was also observed on genes required to mount an effective stress response. Further, under conditions of induced oxidative stress, reduction of SIN3 and/or dKDM5/LID altered the expression of a greater number of genes involved in cell cycle-related processes relative to normal conditions. This highlights an important role for SIN3 and dKDM5/LID proteins to maintain proper progression through the cell cycle in environments of cellular stress. Further, we find that target genes are bound by both SIN3 and dKDM5/LID, however, histone acetylation, not methylation, plays a predominant role in gene regulation by the SIN3 complex. CONCLUSIONS: We have provided genetic evidence to demonstrate functional cooperation between the histone demethylase dKDM5/LID and SIN3. Biochemical and transcriptome data further support functional links between these proteins. Together, the data provide a solid framework for analyzing the gene regulatory pathways through which SIN3 and dKDM5/LID control diverse biological processes in the organism.

13.
G3 (Bethesda) ; 6(1): 121-32, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26546310

ABSTRACT

Histone methylation levels, which are determined by the action of both histone demethylases and methyltransferases, impact multiple biological processes by affecting gene expression activity. Methionine metabolism generates the major methyl donor S-adenosylmethionine (SAM) for histone methylation. The functions of methionine metabolic enzymes in regulating biological processes as well as the interaction between the methionine pathway and histone methylation, however, are still not fully understood. Here, we report that reduced levels of some enzymes involved in methionine metabolism and histone demethylases lead to lethality as well as wing development and cell proliferation defects in Drosophila melanogaster. Additionally, disruption of methionine metabolism can directly affect histone methylation levels. Reduction of little imaginal discs (LID) histone demethylase, but not lysine-specific demethylase 2 (KDM2) demethylase, is able to counter the effects on histone methylation due to reduction of SAM synthetase (SAM-S). Taken together, these results reveal an essential role of key enzymes that control methionine metabolism and histone methylation. Additionally, these findings are an indication of a strong connection between metabolism and epigenetics.


Subject(s)
Drosophila melanogaster/metabolism , Histones/metabolism , Methionine/metabolism , Animals , Drosophila melanogaster/genetics , Gene Knockdown Techniques , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Phenotype
14.
Fly (Austin) ; 9(1): 36-44, 2015.
Article in English | MEDLINE | ID: mdl-26207949

ABSTRACT

Epigenetic regulation of chromatin structure is a fundamental process for eukaryotes. Regulators include DNA methylation, microRNAs and chromatin modifications. Within the chromatin modifiers, one class of enzymes that can functionally bind and modify chromatin, through the removal of methyl marks, is the histone lysine demethylases. Here, we summarize the current findings of the 13 known histone lysine demethylases in Drosophila melanogaster, and discuss the critical role of these histone-modifying enzymes in the maintenance of genomic functions. Additionally, as histone demethylase dysregulation has been identified in cancer, we discuss the advantages for using Drosophila as a model system to study tumorigenesis.


Subject(s)
Drosophila melanogaster/enzymology , Histone Demethylases/metabolism , Animals , Carcinogenesis , Disease Models, Animal , Jumonji Domain-Containing Histone Demethylases/metabolism , Neoplasms/enzymology
15.
Aging (Albany NY) ; 6(8): 645-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25133314

ABSTRACT

Coordinate control of gene activity is critical for fitness and longevity of an organism. The SIN3 histone deacetylase (HDAC) complex functions as a transcriptional repressor of many genes. SIN3-regulated genes include those that encode proteins affecting multiple aspects of mitochondrial function, such as energy production and stress responsiveness, important for health maintenance. Here we used Drosophila melanogaster as a model organism to examine the role of SIN3 in the regulation of fitness and longevity. Adult flies with RNA interference (RNAi) induced knockdown expression of Sin3A have reduced climbing ability; an activity that likely requires fully functional mitochondria. Additionally, compared to wild type, adult Sin3A knockdown flies were more sensitive to oxidative stress. Interestingly, media supplementation with the antioxidant glutathione largely restored fly tolerance to oxidative stress. Although Sin3A knockdown flies exhibited decreased longevity compared to wild type, no significant changes in expression of many well-categorized aging genes were observed. We found, however, that Sin3A knockdown corresponded to a significant reduction in expression of genes encoding proteins involved in the de novo synthesis of glutathione. Taken together, the data support a model whereby SIN3 regulates a gene expression program required for proper mitochondrial function and effective stress response during adulthood.


Subject(s)
Drosophila Proteins/metabolism , Longevity/physiology , Oxidative Stress/physiology , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Stress, Physiological/physiology , Animals , Behavior, Animal/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Motor Activity/physiology , RNA Interference , Sin3 Histone Deacetylase and Corepressor Complex/genetics
16.
PLoS One ; 7(11): e49563, 2012.
Article in English | MEDLINE | ID: mdl-23166712

ABSTRACT

The role of the Sin3A transcriptional corepressor in regulating the cell cycle is established in various metazoans. Little is known, however, about the signaling pathways that trigger or are triggered by Sin3A function. To discover genes that work in similar or opposing pathways to Sin3A during development, we have performed an unbiased screen of deficiencies of the Drosophila third chromosome. Additionally, we have performed a targeted loss of function screen to identify cell cycle genes that genetically interact with Sin3A. We have identified genes that encode proteins involved in regulation of gene expression, signaling pathways and cell cycle that can suppress the curved wing phenotype caused by the knockdown of Sin3A. These data indicate that Sin3A function is quite diverse and impacts a wide variety of cellular processes.


Subject(s)
Drosophila melanogaster/genetics , Phenotype , Repressor Proteins/genetics , Wings, Animal/growth & development , Animals , Body Patterning/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomes, Insect , Cyclin-Dependent Kinase 8/metabolism , Drosophila melanogaster/metabolism , Epistasis, Genetic , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Male , RNA Interference , Repressor Proteins/metabolism , Signal Transduction , Sin3 Histone Deacetylase and Corepressor Complex , Transcription, Genetic , Wings, Animal/anatomy & histology , Wings, Animal/pathology
17.
Front Biosci (Landmark Ed) ; 17(3): 909-37, 2012 01 01.
Article in English | MEDLINE | ID: mdl-22201781

ABSTRACT

Post-translational modification of histones is a major mechanism of epigenetic regulation of eukaryotic transcription. Drosophila has proven to be an important model system for the study of histone modifying enzymes and the cross talk that occurs between the various modifications. Polytene chromosome analysis and genome-wide chromatin immunoprecipitation (ChIP) studies have provided much insight into the location of marks and many of the enzymes that perform the catalytic reactions. Gene specific effects have been determined through study of flies carrying mutations in histone modifying enzymes. This review will highlight classic studies and present recent progress on both the localization data and mutant analyses. This information has been used to assign function to the marks and to the enzymes that place or remove them, critical for the process of transcriptional regulation.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Animals , Epigenesis, Genetic , Genome, Insect , Heterochromatin/genetics , Histones/chemistry , Histones/metabolism , Male , Models, Genetic , Protein Processing, Post-Translational , Sex Chromosomes/genetics , Transcription, Genetic
18.
BMC Biochem ; 11: 26, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20618965

ABSTRACT

BACKGROUND: SIN3 is a transcriptional repressor protein known to regulate many genes, including a number of those that encode mitochondrial components. RESULTS: By monitoring RNA levels, we find that loss of SIN3 in Drosophila cultured cells results in up-regulation of not only nuclear encoded mitochondrial genes, but also those encoded by the mitochondrial genome. The up-regulation of gene expression is accompanied by a perturbation in ATP levels in SIN3-deficient cells, suggesting that the changes in mitochondrial gene expression result in altered mitochondrial activity. In support of the hypothesis that SIN3 is necessary for normal mitochondrial function, yeast sin3 null mutants exhibit very poor growth on non-fermentable carbon sources and show lower levels of ATP and reduced respiration rates. CONCLUSIONS: The findings that both yeast and Drosophila SIN3 affect mitochondrial activity suggest an evolutionarily conserved role for SIN3 in the control of cellular energy production.


Subject(s)
Drosophila Proteins/metabolism , Mitochondria/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Drosophila/metabolism , Drosophila Proteins/genetics , Gene Knockdown Techniques , RNA Interference , Respiratory Rate , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics
19.
J Biol Chem ; 285(35): 27457-27467, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20566628

ABSTRACT

The SIN3 corepressor serves as a scaffold for the assembly of histone deacetylase (HDAC) complexes. SIN3 and its associated HDAC have been shown to have critical roles in both development and the regulation of cell cycle progression. Although multiple SIN3 isoforms have been reported in simple to complex eukaryotic organisms, the mechanisms by which such isoforms regulate specific biological processes are still largely uncharacterized. To gain insight into how SIN3 isoform-specific function contributes to the growth and development of a metazoan organism, we have affinity-purified two SIN3 isoform-specific complexes, SIN3 187 and 220, from Drosophila S2 cells and embryos. We have identified a number of proteins common to the complexes, including the HDAC RPD3, as well as orthologs of several proteins known to have roles in regulating cell proliferation in other organisms. We additionally identified factors, including the histone demethylase little imaginal discs and histone-interacting protein p55, that exhibited a preferential interaction with the largest SIN3 isoform. Our experiments indicate that the isoforms are associated with distinct HDAC activity and are recruited to unique and shared sites along polytene chromosome arms. Furthermore, although expression of SIN3 220 can substitute for genetic loss of other isoforms, expression of SIN3 187 does not support Drosophila viability. Together our findings suggest that SIN3 isoforms serve distinct roles in transcriptional regulation by partnering with different histone-modifying enzymes.


Subject(s)
Drosophila Proteins/metabolism , Histone Deacetylase 1/metabolism , Multiprotein Complexes/metabolism , Repressor Proteins/metabolism , Retinoblastoma-Binding Protein 4/metabolism , Animals , Body Patterning/physiology , Cell Line , Cell Proliferation , Chromosomes/genetics , Chromosomes/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Embryo, Nonmammalian/metabolism , Histone Deacetylase 1/genetics , Histones/genetics , Histones/metabolism , Multiprotein Complexes/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins/genetics , Retinoblastoma-Binding Protein 4/genetics , Sin3 Histone Deacetylase and Corepressor Complex , Transcription, Genetic/physiology
20.
Mech Dev ; 127(1-2): 96-106, 2010.
Article in English | MEDLINE | ID: mdl-19825413

ABSTRACT

The transcriptional corepressor SIN3 is an essential gene in metazoans. In cell culture experiments, loss of SIN3 leads to defects in cell proliferation. Whether and how SIN3 may regulate the cell cycle during development has not been explored. To gain insight into this relationship, we have generated conditional knock down of Drosophila SIN3 and analyzed effects on growth and development in the wing imaginal disc. We find that loss of SIN3 affects normal cell growth and leads to down regulation of expression of the cell cycle regulator gene String (STG). A SIN3 knock down phenotype can be suppressed by overexpression either of STG or of Cdk1, the target of STG phosphatase. These data link SIN3 and STG in a genetic pathway that affects cell cycle progression in a developing tissue.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Protein Tyrosine Phosphatases/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Wings, Animal/embryology , Alleles , Animals , Animals, Genetically Modified , Cell Cycle , Cell Proliferation , Cloning, Molecular , Green Fluorescent Proteins/metabolism , Models, Biological , Models, Genetic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...