Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 17(11): e1009668, 2021 11.
Article in English | MEDLINE | ID: mdl-34807910

ABSTRACT

The Spt/Ada-Gcn5 Acetyltransferase (SAGA) coactivator complex has multiple modules with different enzymatic and non-enzymatic functions. How each module contributes to gene expression is not well understood. During Drosophila oogenesis, the enzymatic functions are not equally required, which may indicate that different genes require different enzymatic functions. An analogy for this phenomenon is the handyman principle: while a handyman has many tools, which tool he uses depends on what requires maintenance. Here we analyzed the role of the non-enzymatic core module during Drosophila oogenesis, which interacts with TBP. We show that depletion of SAGA-specific core subunits blocked egg chamber development at earlier stages than depletion of enzymatic subunits. These results, as well as additional genetic analyses, point to an interaction with TBP and suggest a differential role of SAGA modules at different promoter types. However, SAGA subunits co-occupied all promoter types of active genes in ChIP-seq and ChIP-nexus experiments, and the complex was not specifically associated with distinct promoter types in the ovary. The high-resolution genomic binding profiles were congruent with SAGA recruitment by activators upstream of the start site, and retention on chromatin by interactions with modified histones downstream of the start site. Our data illustrate that a distinct genetic requirement for specific components may conceal the fact that the entire complex is physically present and suggests that the biological context defines which module functions are critical.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Histone Acetyltransferases/metabolism , Oogenesis/physiology , Promoter Regions, Genetic , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Histone Acetyltransferases/genetics , Oogenesis/genetics
2.
Proc Natl Acad Sci U S A ; 117(50): 31861-31870, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257578

ABSTRACT

Streamlined characterization of protein complexes remains a challenge for the study of protein interaction networks. Here we describe serial capture affinity purification (SCAP), in which two separate proteins are tagged with either the HaloTag or the SNAP-tag, permitting a multistep affinity enrichment of specific protein complexes. The multifunctional capabilities of this protein-tagging system also permit in vivo validation of interactions using acceptor photobleaching Förster resonance energy transfer and fluorescence cross-correlation spectroscopy quantitative imaging. By coupling SCAP to cross-linking mass spectrometry, an integrative structural model of the complex of interest can be generated. We demonstrate this approach using the Spindlin1 and SPINDOC protein complex, culminating in a structural model with two SPINDOC molecules docked on one SPIN1 molecule. In this model, SPINDOC interacts with the SPIN1 interface previously shown to bind a lysine and arginine methylated sequence of histone H3. Our approach combines serial affinity purification, live cell imaging, and cross-linking mass spectrometry to build integrative structural models of protein complexes.


Subject(s)
Chromatography, Affinity/methods , Mass Spectrometry/methods , Models, Molecular , Cell Cycle Proteins/genetics , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , Co-Repressor Proteins/genetics , Co-Repressor Proteins/isolation & purification , Co-Repressor Proteins/metabolism , Feasibility Studies , Fluorescent Dyes/chemistry , HEK293 Cells , Humans , Intravital Microscopy , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/isolation & purification , Microtubule-Associated Proteins/metabolism , Molecular Imaging/methods , Molecular Probes/chemistry , Phosphoproteins/genetics , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
3.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-31965995

ABSTRACT

Under nutritional stress, cells undergo metabolic rewiring that results in changes of various cellular processes that include gene transcription. This transcriptional regulation requires dynamic chromatin remodeling that involves histone post-translational modifications. There are several histone marks that may act as switches upon starvation for stress-response pathways.


Subject(s)
Chromatin/genetics , Glucose/metabolism , Histone Code/genetics , Histones/genetics , Nucleosomes/genetics , Humans , Methylation , Protein Processing, Post-Translational/genetics , Signal Transduction/genetics , Starvation/genetics , Starvation/metabolism
4.
Cell Res ; 29(9): 694-695, 2019 09.
Article in English | MEDLINE | ID: mdl-31399698
5.
Nucleic Acids Res ; 47(7): 3383-3394, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30715476

ABSTRACT

The Gcn5 acetyltransferase functions in multiple acetyltransferase complexes in yeast and metazoans. Yeast Gcn5 is part of the large SAGA (Spt-Ada-Gcn5 acetyltransferase) complex and a smaller ADA acetyltransferase complex. In flies and mammals, Gcn5 (and its homolog pCAF) is part of various versions of the SAGA complex and another large acetyltransferase complex, ATAC (Ada2A containing acetyltransferase complex). However, a complex analogous to the small ADA complex in yeast has never been described in metazoans. Previous studies in Drosophila hinted at the existence of a small complex which contains Ada2b, a partner of Gcn5 in the SAGA complex. Here we have purified and characterized the composition of this complex and show that it is composed of Gcn5, Ada2b, Ada3 and Sgf29. Hence, we have named it the metazoan 'ADA complex'. We demonstrate that the fly ADA complex has histone acetylation activity on histones and nucleosome substrates. Moreover, ChIP-Sequencing experiments identified Ada2b peaks that overlap with another SAGA subunit, Spt3, as well as Ada2b peaks that do not overlap with Spt3 suggesting that the ADA complex binds chromosomal sites independent of the larger SAGA complex.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Histone Acetyltransferases/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Nuclear Proteins/metabolism , Animals , Cell Line , Chromatin/metabolism , Drosophila Proteins/isolation & purification , Drosophila melanogaster/cytology , Histone Acetyltransferases/isolation & purification , Multienzyme Complexes/isolation & purification , Nuclear Proteins/isolation & purification , Trans-Activators/isolation & purification , Trans-Activators/metabolism
6.
Genes Dev ; 31(15): 1588-1600, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28887412

ABSTRACT

The Spt-Ada-Gcn5-acetyltransferase (SAGA) chromatin-modifying complex is a transcriptional coactivator that contains four different modules of subunits. The intact SAGA complex has been well characterized for its function in transcription regulation and development. However, little is known about the roles of individual modules within SAGA and whether they have any SAGA-independent functions. Here we demonstrate that the two enzymatic modules of Drosophila SAGA are differently required in oogenesis. Loss of the histone acetyltransferase (HAT) activity blocks oogenesis, while loss of the H2B deubiquitinase (DUB) activity does not. However, the DUB module regulates a subset of genes in early embryogenesis, and loss of the DUB subunits causes defects in embryogenesis. ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) analysis revealed that both the DUB and HAT modules bind most SAGA target genes even though many of these targets do not require the DUB module for expression. Furthermore, we found that the DUB module can bind to chromatin and regulate transcription independently of the HAT module. Our results suggest that the DUB module has functions within SAGA and independent functions.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Histone Acetyltransferases/metabolism , Oogenesis/genetics , Animals , Ataxin-7/genetics , Chromatin/metabolism , Deubiquitinating Enzymes/metabolism , Drosophila Proteins/genetics , Female , High-Throughput Nucleotide Sequencing , Histone Acetyltransferases/genetics , Histones/metabolism , Microscopy, Confocal , Ovary/growth & development , Protein Binding , Zygote/physiology
7.
Cell Rep ; 18(9): 2124-2134, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28249159

ABSTRACT

The 12-subunit Swi/Snf chromatin remodeling complex is conserved from yeast to humans. It functions to alter nucleosome positions by either sliding nucleosomes on DNA or evicting histones. Interestingly, 20% of all human cancers carry mutations in subunits of the Swi/Snf complex. Many of these mutations cause protein instability and loss, resulting in partial Swi/Snf complexes. Although several studies have shown that histone acetylation and activator-dependent recruitment of Swi/Snf regulate its function, it is less well understood how subunits regulate stability and function of the complex. Using functional proteomic and genomic approaches, we have assembled the network architecture of yeast Swi/Snf. In addition, we find that subunits of the Swi/Snf complex regulate occupancy of the catalytic subunit Snf2, thereby modulating gene transcription. Our findings have direct bearing on how cancer-causing mutations in orthologous subunits of human Swi/Snf may lead to aberrant regulation of gene expression by this complex.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Mutation/genetics , Transcription Factors/genetics , Adenosine Triphosphatases/genetics , Catalysis , DNA-Binding Proteins/genetics , Gene Expression/genetics , Genomics/methods , Histones/genetics , Nucleosomes/genetics , Protein Subunits/genetics , Proteomics/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic/genetics
8.
J Mol Biol ; 429(13): 2093-2107, 2017 06 30.
Article in English | MEDLINE | ID: mdl-27984043

ABSTRACT

Mutations that affect myelodysplasia/myeloid leukemia factor (MLF) proteins are associated with leukemia and several other cancers. However, with no strong homology to other proteins of known function, the role of MLF proteins in the cell has remained elusive. Here, we describe a proteomics approach that identifies MLF as a member of a nuclear chaperone complex containing a DnaJ protein, BCL2-associated anthanogene 2, and Hsc70. This complex associates with chromatin and regulates the expression of target genes. The MLF complex is bound to sites of nucleosome depletion and sites containing active chromatin marks (e.g., H3K4me3 and H3K4me1). Hence, MLF binding is enriched at promoters and enhancers. Additionally, the MLF-chaperone complex functions to regulate transcription factor stability, including the RUNX transcription factor involved in hematopoiesis. Although Hsc70 and other co-chaperones have been shown to play a role in nuclear translocation of a variety of proteins including transcription factors, our findings suggest that MLF and the associated co-chaperones play a direct role in modulating gene transcription.


Subject(s)
Gene Expression , Molecular Chaperones , Protein Multimerization , Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Cell Cycle Proteins , Chromatin/metabolism , DNA-Binding Proteins , Drosophila Proteins/metabolism , Protein Binding
10.
Mol Cell ; 63(4): 547-552, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27540855

ABSTRACT

Modifications of histones play important roles in balancing transcriptional output. The discovery of acyl marks, besides histone acetylation, has added to the functional diversity of histone modifications. Since all modifications use metabolic intermediates as substrates for chromatin-modifying enzymes, the prevalent landscape of histone modifications in any cell type is a snapshot of its metabolic status. Here, we review some of the current findings of how differential use of histone acylations regulates gene expression as response to metabolic changes and differentiation programs.


Subject(s)
Chromatin Assembly and Disassembly , Energy Metabolism , Histones/metabolism , Protein Processing, Post-Translational , Acylation , Animals , Humans , Signal Transduction , Transcription, Genetic
12.
Mol Cell Biol ; 36(14): 1900-7, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27185879

ABSTRACT

The lysine acetyltransferase 6 (KAT6) histone acetyltransferase (HAT) complexes are highly conserved from yeast to higher organisms. They acetylate histone H3 and other nonhistone substrates and are involved in cell cycle regulation and stem cell maintenance. In addition, the human KAT6 HATs are recurrently mutated in leukemia and solid tumors. Therefore, it is important to understand the mechanisms underlying the regulation of KAT6 HATs and their roles in cell cycle progression. In this minireview, we summarize the identification and analysis of the KAT6 complexes and discuss the regulatory mechanisms governing their enzymatic activities and substrate specificities. We further focus on the roles of KAT6 HATs in regulating cell proliferation and stem cell maintenance and review recent insights that aid in understanding their involvement in human diseases.


Subject(s)
Histone Acetyltransferases/metabolism , Neoplasms/enzymology , Neurodevelopmental Disorders/enzymology , Acetylation , Cell Cycle , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Mutation , Neoplasms/genetics , Neurodevelopmental Disorders/genetics , Stem Cells/cytology , Stem Cells/enzymology , Substrate Specificity
13.
Genes Dev ; 30(10): 1198-210, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27198229

ABSTRACT

KAT6 histone acetyltransferases (HATs) are highly conserved in eukaryotes and are involved in cell cycle regulation. However, information regarding their roles in regulating cell cycle progression is limited. Here, we report the identification of subunits of the Drosophila Enok complex and demonstrate that all subunits are important for its HAT activity. We further report a novel interaction between the Enok complex and the Elg1 proliferating cell nuclear antigen (PCNA)-unloader complex. Depletion of Enok in S2 cells resulted in a G1/S cell cycle block, and this block can be partially relieved by depleting Elg1. Furthermore, depletion of Enok reduced the chromatin-bound levels of PCNA in both S2 cells and early embryos, suggesting that the Enok complex may interact with the Elg1 complex and down-regulate its PCNA-unloading function to promote the G1/S transition. Supporting this hypothesis, depletion of Enok also partially rescued the endoreplication defects in Elg1-depleted nurse cells. Taken together, our study provides novel insights into the roles of KAT6 HATs in cell cycle regulation through modulating PCNA levels on chromatin.


Subject(s)
Drosophila Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Histone Acetyltransferases/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cells, Cultured , Chromatin/metabolism , Down-Regulation/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Histone Acetyltransferases/genetics , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism
14.
Methods Mol Biol ; 1313: 149-64, 2015.
Article in English | MEDLINE | ID: mdl-25947663

ABSTRACT

Myoblast fusion in the Drosophila embryo is a highly elaborate process that is initiated by Founder Cells and Fusion-Competent Myoblasts (FCMs). It occurs through an asymmetric event in which actin foci assemble in the FCMs at points of cell-cell contact and direct the formation of membrane protrusions that drive fusion. Herein, we describe the approach that we have used to image in living embryos the highly dynamic actin foci and actin-rich projections that precede myoblast fusion. We discuss resources currently available for imaging actin and myogenesis, and our experience with these resources if available. This technical report is not intended to be comprehensive on providing instruction on standard microscopy practices or software utilization. However, we discuss microscope parameters that we have used in data collection, and our experience with image processing tools in data analysis.


Subject(s)
Actins/metabolism , Cell Fusion , Embryo, Nonmammalian , Fluorescent Dyes , Myoblasts/cytology , Myoblasts/metabolism , Animals , Drosophila , Image Processing, Computer-Assisted , Microscopy, Fluorescence
15.
Genetics ; 199(3): 777-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25614583

ABSTRACT

Drosophila Midline (Mid) is an ortholog of vertebrate Tbx20, which plays roles in the developing heart, migrating cranial motor neurons, and endothelial cells. Mid functions in cell-fate specification and differentiation of tissues that include the ectoderm, cardioblasts, neuroblasts, and egg chambers; however, a role in the somatic musculature has not been described. We identified mid in genetic and molecular screens for factors contributing to somatic muscle morphogenesis. Mid is expressed in founder cells (FCs) for several muscle fibers, and functions cooperatively with the T-box protein H15 in lateral oblique muscle 1 and the segment border muscle. Mid is particularly important for the specification and development of the lateral transverse (LT) muscles LT3 and LT4, which arise by asymmetric division of a single muscle progenitor. Mid is expressed in this progenitor and its two sibling FCs, but is maintained only in the LT4 FC. Both muscles were frequently missing in mid mutant embryos, and LT4-associated expression of the transcription factor Krüppel (Kr) was lost. When present, LT4 adopted an LT3-like morphology. Coordinately, mid misexpression caused LT3 to adopt an LT4-like morphology and was associated with ectopic Kr expression. From these data, we concluded that mid functions first in the progenitor to direct development of LT3 and LT4, and later in the FCs to influence whichever of these differentiation profiles is selected. Mid is the first T-box factor shown to influence LT3 and LT4 muscle identity and, along with the T-box protein Optomotor-blind-related-gene 1 (Org-1), is representative of a new class of transcription factors in muscle specification.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Muscle Development/genetics , T-Box Domain Proteins/genetics , Animals , Body Patterning/genetics , Drosophila/embryology , Drosophila Proteins/metabolism , Gene Expression , Kruppel-Like Transcription Factors/metabolism , Organ Specificity , Transcription Factors/genetics
16.
Genes Dev ; 28(24): 2750-63, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25512562

ABSTRACT

KAT6 histone acetyltransferases (HATs) are highly conserved in eukaryotes and have been shown to play important roles in transcriptional regulation. Here, we demonstrate that the Drosophila KAT6 Enok acetylates histone H3 Lys 23 (H3K23) in vitro and in vivo. Mutants lacking functional Enok exhibited defects in the localization of Oskar (Osk) to the posterior end of the oocyte, resulting in loss of germline formation and abdominal segments in the embryo. RNA sequencing (RNA-seq) analysis revealed that spire (spir) and maelstrom (mael), both required for the posterior localization of Osk in the oocyte, were down-regulated in enok mutants. Chromatin immunoprecipitation showed that Enok is localized to and acetylates H3K23 at the spir and mael genes. Furthermore, Gal4-driven expression of spir in the germline can largely rescue the defective Osk localization in enok mutant ovaries. Our results suggest that the Enok-mediated H3K23 acetylation (H3K23Ac) promotes the expression of spir, providing a specific mechanism linking oocyte polarization to histone modification.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Gene Expression Regulation, Developmental , Histone Acetyltransferases/metabolism , Microfilament Proteins/genetics , Acetylation , Animals , Chromatin Immunoprecipitation , Drosophila melanogaster/genetics , Embryo, Nonmammalian , Female , Histone Acetyltransferases/genetics , Histones/metabolism , Microfilament Proteins/metabolism , Mutation , Oocytes/cytology , Oocytes/enzymology , Ovary/metabolism , Protein Isoforms
17.
PLoS One ; 9(12): e114126, 2014.
Article in English | MEDLINE | ID: mdl-25474591

ABSTRACT

The process of myogenesis includes the recognition, adhesion, and fusion of committed myoblasts into multinucleate syncytia. In the larval body wall muscles of Drosophila, this elaborate process is initiated by Founder Cells and Fusion-Competent Myoblasts (FCMs), and cell adhesion molecules Kin-of-IrreC (Kirre) and Sticks-and-stones (Sns) on their respective surfaces. The FCMs appear to provide the driving force for fusion, via the assembly of protrusions associated with branched F-actin and the WASp, SCAR and Arp2/3 pathways. In the present study, we utilize the dorsal pharyngeal musculature that forms in the Drosophila embryo as a model to explore myoblast fusion and visualize the fusion process in live embryos. These muscles rely on the same cell types and genes as the body wall muscles, but are amenable to live imaging since they do not undergo extensive morphogenetic movement during formation. Time-lapse imaging with F-actin and membrane markers revealed dynamic FCM-associated actin-enriched protrusions that rapidly extend and retract into the myotube from different sites within the actin focus. Ultrastructural analysis of this actin-enriched area showed that they have two morphologically distinct structures: wider invasions and/or narrow filopodia that contain long linear filaments. Consistent with this, formin Diaphanous (Dia) and branched actin nucleator, Arp3, are found decorating the filopodia or enriched at the actin focus, respectively, indicating that linear actin is present along with branched actin at sites of fusion in the FCM. Gain-of-function Dia and loss-of-function Arp3 both lead to fusion defects, a decrease of F-actin foci and prominent filopodia from the FCMs. We also observed differential endocytosis of cell surface components at sites of fusion, with actin reorganizing factors, WASp and SCAR, and Kirre remaining on the myotube surface and Sns preferentially taken up with other membrane proteins into early endosomes and lysosomes in the myotube.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Endocytosis , Molecular Imaging , Myoblasts/cytology , Pseudopodia/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Fusion , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Drosophila melanogaster/embryology , Drosophila melanogaster/ultrastructure , Formins , Gene Expression Regulation , Muscle Fibers, Skeletal/cytology , Myoblasts/ultrastructure , Pharyngeal Muscles/cytology , Pharyngeal Muscles/embryology , Pseudopodia/ultrastructure
18.
Fly (Austin) ; 8(3): 148-52, 2014.
Article in English | MEDLINE | ID: mdl-25483136

ABSTRACT

The SAGA chromatin modifying complex functions as a transcriptional coactivator for a large number of genes, and SAGA dysfunction has been linked to carcinogenesis and neurodegenerative disease. The protein complex is comprised of approximately 20 subunits, arranged in a modular fashion, and includes 2 enzymatic subunits: the Gcn5 acetyltransferase and the Non-stop deubiquitinase. As we learn more about SAGA, it becomes evident that this complex functions through sophisticated mechanisms that support very precise regulation of gene expression. Here we describe recent findings in which a Drosophila loss-of-function model revealed novel mechanisms for regulation of SAGA-mediated histone H2B deubiquitination. This model also yielded novel and surprising insights into mechanisms that underlie progressive neurodegenerative disease. Lastly, we comment on the utility of Drosophila as a model for neurodegenerative disease through which crucial and conserved mechanisms may be revealed.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Endopeptidases/metabolism , Histones/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/etiology , Animals , Ataxin-7 , Disease Models, Animal , Female , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/metabolism , Retinal Degeneration/etiology , Retinal Degeneration/metabolism , Transcription Factors/metabolism , Ubiquitination
19.
Curr Opin Genet Dev ; 26: 96-104, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25108806

ABSTRACT

Nine genetic diseases arise from expansion of CAG repeats in seemingly unrelated genes. They are referred to as polyglutamine (polyQ) diseases due to the presence of elongated glutamine tracts in the corresponding proteins. The pathologic consequences of polyQ expansion include progressive spinal, cerebellar, and neural degeneration. These pathologies are not identical, however, suggesting that disruption of protein-specific functions is crucial to establish and maintain each disease. A closer examination of protein function reveals that several act as regulators of gene expression. Here we examine the roles these proteins play in regulating gene expression, discuss how polyQ expansion may disrupt these functions to cause disease, and speculate on the neural specificity of perturbing ubiquitous gene regulators.


Subject(s)
Chromatin/genetics , Gene Expression Regulation , Genetic Predisposition to Disease/genetics , Neurodegenerative Diseases/genetics , Trinucleotide Repeat Expansion/genetics , Chromatin/metabolism , Humans , Models, Genetic , Neurodegenerative Diseases/metabolism , Peptides/genetics , Peptides/metabolism , Protein Binding
20.
Rare Dis ; 2: e28859, 2014.
Article in English | MEDLINE | ID: mdl-25054097

ABSTRACT

Spinocerebellar ataxia 7 (SCA7) is an incurable disease caused by expansion of CAG trinucleotide sequences within the Ataxin-7 gene. This elongated CAG tract results in an Ataxin-7 protein bearing an expanded polyglutamine (PolyQ) repeat. SCA7 disease is characterized by progressive neural and retinal degeneration leading to ataxia and blindness. Evidence gathered from investigating SCA7 and other PolyQ diseases strongly suggest that misregulation of gene expression contributes to neurodegeneration. In fact, Ataxin-7 is a subunit of the essential Spt-Ada-Gcn5-Acetltransferase (SAGA) chromatin modifying complex that regulates expression of a large number of genes. Here we discuss recent insights into Ataxin-7 function and, considering these findings, propose a model for how polyglutamine expansion of Ataxin-7 may affect Ataxin-7 function to alter chromatin modifications and gene expression.

SELECTION OF CITATIONS
SEARCH DETAIL
...