Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4461, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37491334

ABSTRACT

Epigenetic regulation of histone H3K27 methylation has recently emerged as a key step during alternative immunoregulatory M2-like macrophage polarization; known to impact cardiac repair after Myocardial Infarction (MI). We hypothesized that EZH2, responsible for H3K27 methylation, could act as an epigenetic checkpoint regulator during this process. We demonstrate for the first time an ectopic EZH2, and putative, cytoplasmic inactive localization of the epigenetic enzyme, during monocyte differentiation into M2 macrophages in vitro as well as in immunomodulatory cardiac macrophages in vivo in the post-MI acute inflammatory phase. Moreover, we show that pharmacological EZH2 inhibition, with GSK-343, resolves H3K27 methylation of bivalent gene promoters, thus enhancing their expression to promote human monocyte repair functions. In line with this protective effect, GSK-343 treatment accelerated cardiac inflammatory resolution preventing infarct expansion and subsequent cardiac dysfunction in female mice post-MI in vivo. In conclusion, our study reveals that pharmacological epigenetic modulation of cardiac-infiltrating immune cells may hold promise to limit adverse cardiac remodeling after MI.


Subject(s)
Monocytes , Myocardial Infarction , Animals , Female , Humans , Mice , Cell Differentiation , Epigenesis, Genetic , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism
2.
Cells ; 11(6)2022 03 11.
Article in English | MEDLINE | ID: mdl-35326417

ABSTRACT

The induction of lineage-specific gene programs are strongly influenced by alterations in local chromatin architecture. However, key players that impact this genome reorganization remain largely unknown. Here, we report that the removal of the special AT-rich binding protein 2 (SATB2), a nuclear protein known to bind matrix attachment regions, is a key event in initiating myogenic differentiation. The deletion of myoblast SATB2 in vitro initiates chromatin remodeling and accelerates differentiation, which is dependent on the caspase 7-mediated cleavage of SATB2. A genome-wide analysis indicates that SATB2 binding within chromatin loops and near anchor points influences both loop and sub-TAD domain formation. Consequently, the chromatin changes that occur with the removal of SATB2 lead to the derepression of differentiation-inducing factors while also limiting the expression of genes that inhibit this cell fate change. Taken together, this study demonstrates that the temporal control of the SATB2 protein is critical in shaping the chromatin environment and coordinating the myogenic differentiation program.


Subject(s)
Matrix Attachment Region Binding Proteins , Caspases , Chromatin , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Myoblasts/metabolism , Transcription Factors/metabolism
3.
Theranostics ; 10(14): 6337-6360, 2020.
Article in English | MEDLINE | ID: mdl-32483456

ABSTRACT

Rationale: Monoacylglycerol lipase (Mgll), a hydrolase that breaks down the endocannabinoid 2-arachidonoyl glycerol (2-AG) to produce arachidonic acid (ARA), is a potential target for neurodegenerative diseases, such as Alzheimer's disease (AD). Increasing evidence shows that impairment of adult neurogenesis by perturbed lipid metabolism predisposes patients to AD. However, it remains unknown what causes aberrant expression of Mgll in AD and how Mgll-regulated lipid metabolism impacts adult neurogenesis, thus predisposing to AD during aging. Here, we identify Mgll as an aging-induced factor that impairs adult neurogenesis and spatial memory in AD, and show that metformin, an FDA-approved anti-diabetic drug, can reduce the expression of Mgll to reverse impaired adult neurogenesis, prevent spatial memory decline and reduce ß-amyloid accumulation. Methods: Mgll expression was assessed in both human AD patient post-mortem hippocampal tissues and 3xTg-AD mouse model. In addition, we used both the 3xTg-AD animal model and the CbpS436A genetic knock-in mouse model to identify that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway, involving atypical protein kinase C (aPKC)-stimulated Ser436 phosphorylation of histone acetyltransferase CBP through biochemical methods. Furthermore, we performed in vivo adult neurogenesis assay with BrdU/EdU labelling and Morris water maze task in both animal models following pharmacological treatments to show the key role of Mgll in metformin-corrected neurogenesis and spatial memory deficits of AD through reactivating the aPKC-CBP pathway. Finally, we performed in vitro adult neurosphere assays using both animal models to study the role of the aPKC-CBP mediated Mgll repression in determining adult neural stem/progenitor cell (NPC) fate. Results: Here, we demonstrate that aging-dependent induction of Mgll is observed in the 3xTg-AD model and human AD patient post-mortem hippocampal tissues. Importantly, we discover that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway. The accumulation of Mgll in the 3xTg-AD mice reduces the genesis of newborn neurons and perturbs spatial memory. However, we find that metformin-stimulated aPKC-CBP pathway decreases Mgll expression to recover these deficits in 3xTg-AD. In addition, we reveal that elevated Mgll levels in cultured adult NPCs from both 3xTg-AD and CbpS436A animal models are responsible for their NPC neuronal differentiation deficits. Conclusion: Our findings set the stage for development of a clinical protocol where Mgll would serve as a biomarker in early stages of AD to identify potential metformin-responsive AD patients to restore their neurogenesis and spatial memory.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Metformin/pharmacology , Monoacylglycerol Lipases/metabolism , Neurogenesis/drug effects , Spatial Memory/drug effects , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , CREB-Binding Protein/metabolism , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Transgenic , Protein Kinase C/metabolism
4.
Stem Cell Reports ; 9(5): 1573-1587, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29033304

ABSTRACT

Human endothelial colony-forming cells (ECFCs) represent a promising source of adult stem cells for vascular repair, yet their regenerative capacity is limited. Here, we set out to understand the molecular mechanism restricting the repair function of ECFCs. We found that key pro-angiogenic pathways are repressed in ECFCs due to the presence of bivalent (H3K27me3/H3K4me3) epigenetic marks, which decreases the cells' regenerative potential. Importantly, ex vivo treatment with a combination of epigenetic drugs that resolves bivalent marks toward the transcriptionally active H3K4me3 state leads to the simultaneous activation of multiple pro-angiogenic signaling pathways (VEGFR, CXCR4, WNT, NOTCH, SHH). This in turn results in improved capacity of ECFCs to form capillary-like networks in vitro and in vivo. Furthermore, restoration of perfusion is accelerated upon transplantation of drug-treated ECFCs in a model of hindlimb ischemia. Thus, ex vivo treatment with epigenetic drugs increases the vascular repair properties of ECFCs through transient activation of pro-angiogenic signaling pathways.


Subject(s)
Endothelial Progenitor Cells/metabolism , Epigenesis, Genetic , Neovascularization, Physiologic , Signal Transduction , Animals , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Female , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Hindlimb/blood supply , Humans , Ischemia/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
5.
Mol Biol Cell ; 28(18): 2347-2359, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28701342

ABSTRACT

Pancreatitis is a debilitating disease of the exocrine pancreas that, under chronic conditions, is a major susceptibility factor for pancreatic ductal adenocarcinoma (PDAC). Although down-regulation of genes that promote the mature acinar cell fate is required to reduce injury associated with pancreatitis, the factors that promote this repression are unknown. Activating transcription factor 3 (ATF3) is a key mediator of the unfolded protein response, a pathway rapidly activated during pancreatic insult. Using chromatin immunoprecipitation followed by next-generation sequencing, we show that ATF3 is bound to the transcriptional regulatory regions of >30% of differentially expressed genes during the initiation of pancreatitis. Of importance, ATF3-dependent regulation of these genes was observed only upon induction of pancreatitis, with pathways involved in inflammation, acinar cell differentiation, and cell junctions being specifically targeted. Characterizing expression of transcription factors that affect acinar cell differentiation suggested that acinar cells lacking ATF3 maintain a mature cell phenotype during pancreatitis, a finding supported by maintenance of junctional proteins and polarity markers. As a result, Atf3-/- pancreatic tissue displayed increased tissue damage and inflammatory cell infiltration at early time points during injury but, at later time points, showed reduced acinar-to-duct cell metaplasia. Thus our results reveal a critical role for ATF3 as a key regulator of the acinar cell transcriptional response during injury and may provide a link between chronic pancreatitis and PDAC.


Subject(s)
Acinar Cells/metabolism , Activating Transcription Factor 3/metabolism , Pancreatitis/metabolism , Pancreatitis/pathology , Acinar Cells/cytology , Activating Transcription Factor 3/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/physiology , Ceruletide , Down-Regulation , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Phenotype , Pancreatic Neoplasms
6.
J Clin Invest ; 126(4): 1555-65, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26999603

ABSTRACT

The X chromosome-encoded histone demethylase UTX (also known as KDM6A) mediates removal of repressive trimethylation of histone H3 lysine 27 (H3K27me3) to establish transcriptionally permissive chromatin. Loss of UTX in female mice is embryonic lethal. Unexpectedly, male UTX-null mice escape embryonic lethality due to expression of UTY, a paralog that lacks H3K27 demethylase activity, suggesting an enzyme-independent role for UTX in development and thereby challenging the need for active H3K27 demethylation in vivo. However, the requirement for active H3K27 demethylation in stem cell-mediated tissue regeneration remains untested. Here, we employed an inducible mouse KO that specifically ablates Utx in satellite cells (SCs) and demonstrated that active H3K27 demethylation is necessary for muscle regeneration. Loss of UTX in SCs blocked myofiber regeneration in both male and female mice. Furthermore, we demonstrated that UTX mediates muscle regeneration through its H3K27 demethylase activity, as loss of demethylase activity either by chemical inhibition or knock-in of demethylase-dead UTX resulted in defective muscle repair. Mechanistically, dissection of the muscle regenerative process revealed that the demethylase activity of UTX is required for expression of the transcription factor myogenin, which in turn drives differentiation of muscle progenitors. Thus, we have identified a critical role for the enzymatic activity of UTX in activating muscle-specific gene expression during myofiber regeneration and have revealed a physiological role for active H3K27 demethylation in vivo.


Subject(s)
Gene Expression Regulation/physiology , Histone Demethylases/biosynthesis , Myofibrils/physiology , Myogenin/metabolism , Regeneration/physiology , Satellite Cells, Skeletal Muscle/enzymology , Animals , Female , Gene Knock-In Techniques , Histone Demethylases/genetics , Histones/genetics , Histones/metabolism , Male , Mice , Mice, Knockout , Myogenin/genetics , Satellite Cells, Skeletal Muscle/cytology
7.
Genes Dev ; 30(5): 508-21, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26944678

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous group of hematological tumors composed of distinct subtypes that vary in their genetic abnormalities, gene expression signatures, and prognoses. However, it remains unclear whether T-ALL subtypes differ at the functional level, and, as such, T-ALL treatments are uniformly applied across subtypes, leading to variable responses between patients. Here we reveal the existence of a subtype-specific epigenetic vulnerability in T-ALL by which a particular subgroup of T-ALL characterized by expression of the oncogenic transcription factor TAL1 is uniquely sensitive to variations in the dosage and activity of the histone 3 Lys27 (H3K27) demethylase UTX/KDM6A. Specifically, we identify UTX as a coactivator of TAL1 and show that it acts as a major regulator of the TAL1 leukemic gene expression program. Furthermore, we demonstrate that UTX, previously described as a tumor suppressor in T-ALL, is in fact a pro-oncogenic cofactor essential for leukemia maintenance in TAL1-positive (but not TAL1-negative) T-ALL. Exploiting this subtype-specific epigenetic vulnerability, we propose a novel therapeutic approach based on UTX inhibition through in vivo administration of an H3K27 demethylase inhibitor that efficiently kills TAL1-positive primary human leukemia. These findings provide the first opportunity to develop personalized epigenetic therapy for T-ALL patients.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy , Histone Demethylases/genetics , Nuclear Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , Histone Demethylases/metabolism , Humans , Nuclear Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1
8.
Cell Stem Cell ; 14(5): 644-57, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24792117

ABSTRACT

A major goal of cell therapy for vascular diseases is to promote revascularization through the injection of endothelial stem/progenitor cells. The gene regulatory mechanisms that underlie endothelial progenitor-mediated vascular repair, however, remain elusive. Here, we identify the transcription factor TAL1/SCL as a key mediator of the vascular repair function of primary human endothelial colony-forming cells (ECFCs). Genome-wide analyses in ECFCs demonstrate that TAL1 activates a transcriptional program that promotes cell adhesion and migration. At the mechanistic level, we show that TAL1 upregulates the expression of migratory and adhesion genes through recruitment of the histone acetyltransferase p300. Based on these findings, we establish a strategy that enhances the revascularization efficiency of ECFCs after ischemia through ex vivo priming with the histone deacetylase inhibitor TSA. Thus, small molecule epigenetics drugs are effective tools for modifying the epigenome of stem/progenitor cells prior to transplantation as a means to enhance their therapeutic potential.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Hydroxamic Acids/pharmacology , Proto-Oncogene Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Endothelial Progenitor Cells/cytology , Epigenesis, Genetic/genetics , Genome-Wide Association Study , Humans , Proto-Oncogene Proteins/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1
9.
PLoS One ; 8(6): e67762, 2013.
Article in English | MEDLINE | ID: mdl-23840772

ABSTRACT

Quiescent satellite cells are myogenic progenitors that enable regeneration of skeletal muscle. One of the early events of satellite cell activation following myotrauma is the induction of the myogenic regulatory factor MyoD, which eventually induces terminal differentiation and muscle function gene expression. The purpose of this study was to elucidate the mechanism by which MyoD is induced during activation of satellite cells in mouse muscle undergoing regeneration. We show that Six1, a transcription factor essential for embryonic myogenesis, also regulates MyoD expression in muscle progenitor cells. Six1 knock-down by RNA interference leads to decreased expression of MyoD in myoblasts. Chromatin immunoprecipitation assays reveal that Six1 binds the Core Enhancer Region of MyoD. Further, transcriptional reporter assays demonstrate that Core Enhancer Region reporter gene activity in myoblasts and in regenerating muscle depends on the expression of Six1 and on Six1 binding sites. Finally, we provide evidence indicating that Six1 is required for the proper chromatin structure at the Core Enhancer Region, as well as for MyoD binding at its own enhancer. Together, our results reveal that MyoD expression in satellite cells depends on Six1, supporting the idea that Six1 plays an important role in adult myogenesis, in addition to its role in embryonic muscle formation.


Subject(s)
Homeodomain Proteins/genetics , Muscle, Skeletal/physiology , MyoD Protein/genetics , Satellite Cells, Skeletal Muscle/physiology , Stem Cells/physiology , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genes, Reporter/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Muscle Development/physiology , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myoblasts/metabolism , Myoblasts/physiology , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Regeneration/genetics , Regeneration/physiology , Satellite Cells, Skeletal Muscle/metabolism , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Genes Dev ; 27(11): 1247-59, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23723416

ABSTRACT

Alternate splicing contributes extensively to cellular complexity by generating protein isoforms with divergent functions. However, the role of alternate isoforms in development remains poorly understood. Mef2 transcription factors are essential transducers of cell signaling that modulate differentiation of many cell types. Among Mef2 family members, Mef2D is unique, as it undergoes tissue-specific splicing to generate a muscle-specific isoform. Since the ubiquitously expressed (Mef2Dα1) and muscle-specific (Mef2Dα2) isoforms of Mef2D are both expressed in muscle, we examined the relative contribution of each Mef2D isoform to differentiation. Using both in vitro and in vivo models, we demonstrate that Mef2D isoforms act antagonistically to modulate differentiation. While chromatin immunoprecipitation (ChIP) sequencing analysis shows that the Mef2D isoforms bind an overlapping set of genes, only Mef2Dα2 activates late muscle transcription. Mechanistically, the differential ability of Mef2D isoforms to activate transcription depends on their susceptibility to phosphorylation by protein kinase A (PKA). Phosphorylation of Mef2Dα1 by PKA provokes its association with corepressors. Conversely, exon switching allows Mef2Dα2 to escape this inhibitory phosphorylation, permitting recruitment of Ash2L for transactivation of muscle genes. Thus, our results reveal a novel mechanism in which a tissue-specific alternate splicing event has evolved that permits a ubiquitously expressed transcription factor to escape inhibitory signaling for temporal regulation of gene expression.


Subject(s)
Alternative Splicing , Cell Differentiation/genetics , Muscles/cytology , Muscles/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Animals , Chromatin Immunoprecipitation , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Exons/genetics , Gene Expression Regulation/genetics , Genome/genetics , MEF2 Transcription Factors , Mice , Muscles/enzymology , Mutation/genetics , Myogenic Regulatory Factors/chemistry , Nuclear Proteins/metabolism , Organ Specificity/genetics , Phosphorylation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Transcription, Genetic/genetics
11.
J Cell Biol ; 196(6): 727-42, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22412020

ABSTRACT

Loss of function of dystonin cytoskeletal linker proteins causes neurodegeneration in dystonia musculorum (dt) mutant mice. Although much investigation has focused on understanding dt pathology, the diverse cellular functions of dystonin isoforms remain poorly characterized. In this paper, we highlight novel functions of the dystonin-a2 isoform in mediating microtubule (MT) stability, Golgi organization, and flux through the secretory pathway. Using dystonin mutant mice combined with isoform-specific loss-of-function analysis, we found dystonin-a2 bound to MT-associated protein 1B (MAP1B) in the centrosomal region, where it maintained MT acetylation. In dt neurons, absence of the MAP1B-dystonin-a2 interaction resulted in altered MAP1B perikaryal localization, leading to MT deacetylation and instability. Deacetylated MT accumulation resulted in Golgi fragmentation and prevented anterograde trafficking via motor proteins. Maintenance of MT acetylation through trichostatin A administration or MAP1B overexpression mitigated the observed defect. These cellular aberrations are apparent in prephenotype dorsal root ganglia and primary sensory neurons from dt mice, suggesting they are causal in the disorder.


Subject(s)
Golgi Apparatus/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Acetylation , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dystonia/genetics , Dystonia/metabolism , Dystonin , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred Strains , Microtubule-Associated Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transfection
12.
Stem Cell Rev Rep ; 8(2): 482-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21735106

ABSTRACT

Human embryonic stem cells (hESCs) are a potential source of material for cell therapy of muscle diseases. To date, it has proven difficult to generate skeletal muscle from hESCs in high yields and within a reasonable timeframe. Further, a hESC-derived Pax3/7-positive skeletal muscle progenitor population has not yet been described. Previous studies have shown that Pax3/7-positive progenitor cells can repopulate the satellite cell niche, indicating the importance of this population for therapy. We sought to optimize the differentiation of hESCs into skeletal muscle in order to characterize myogenesis at a molecular level and shorten the time course. We treated hESCs with retinoic acid (RA) and found an enhancement of skeletal myogenesis, and the expression of the myogenic regulatory factors (MRFs) MyoD and myogenin by day 25. Furthermore, we found that RA treatment expanded the muscle progenitor pool, which occurred as a distinct Pax3(+ve) population prior to MRF expression. Non-skeletal muscle tissue types were not significantly affected. Therefore, we have identified a differentiation pathway in hESCs that provides a skeletal muscle progenitor population which can undergo myogenesis more efficiently. We propose that RA could fit into a directed culture method for deriving skeletal muscle from hESCs.


Subject(s)
Embryonic Stem Cells/cytology , Muscle Development/drug effects , Muscle, Skeletal/cytology , Stem Cells/cytology , Tretinoin/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Proliferation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Muscle Development/genetics , Stem Cells/drug effects , Stem Cells/metabolism , Time Factors
13.
Mol Cell Biol ; 30(24): 5686-97, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20956564

ABSTRACT

The highly related mammalian Sin3A and Sin3B proteins provide a versatile platform for chromatin-modifying activities. Sin3-containing complexes play a role in gene repression through deacetylation of nucleosomes. Here, we explore a role for Sin3 in myogenesis by examining the phenotypes resulting from acute somatic deletion of both isoforms in vivo and from primary myotubes in vitro. Myotubes ablated for Sin3A alone, but not Sin3B, displayed gross defects in sarcomere structure that were considerably enhanced upon simultaneous ablation of both isoforms. Massively parallel sequencing of Sin3A- and Sin3B-bound genomic loci revealed a subset of target genes directly involved in sarcomere function that are positively regulated by Sin3A and Sin3B proteins. Both proteins were coordinately recruited to a substantial number of genes. Interestingly, depletion of Sin3B led to compensatory increases in Sin3A recruitment at certain target loci, but Sin3B was never found to compensate for Sin3A loss. Thus, our analyses describe a novel transcriptional role for Sin3A and Sin3B proteins associated with maintenance of differentiated muscle cells.


Subject(s)
Muscle Development/physiology , Muscle, Skeletal , Protein Isoforms/metabolism , Repressor Proteins/metabolism , Sarcomeres/physiology , Animals , Cell Line , Gene Deletion , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Phenotype , Protein Isoforms/genetics , RNA Interference , Repressor Proteins/genetics , Sarcomeres/ultrastructure , Sin3 Histone Deacetylase and Corepressor Complex , Survival Rate
14.
Nucleic Acids Res ; 38(20): 6857-71, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20601407

ABSTRACT

Precise regulation of gene expression is crucial to myogenesis and is thought to require the cooperation of various transcription factors. On the basis of a bioinformatic analysis of gene regulatory sequences, we hypothesized that myogenic regulatory factors (MRFs), key regulators of skeletal myogenesis, cooperate with members of the SIX family of transcription factors, known to play important roles during embryonic skeletal myogenesis. To this day little is known regarding the exact molecular mechanism by which SIX factors regulate muscle development. We have conducted a functional genomic study of the role played by SIX1 and SIX4 during the differentiation of skeletal myoblasts, a model of adult muscle regeneration. We report that SIX factors cooperate with the members of the MRF family to activate gene expression during myogenic differentiation, and that their function is essential to this process. Our findings also support a model where SIX factors function not only 'upstream' of the MRFs during embryogenesis, but also 'in parallel' to them during myoblast differentiation. We have identified new essential nodes that depend on SIX factor function, in the myogenesis regulatory network, and have uncovered a novel way by which MRF function is modulated during differentiation.


Subject(s)
Homeodomain Proteins/metabolism , Muscle Development/genetics , Myoblasts, Skeletal/metabolism , Myogenic Regulatory Factors/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cell Line , Genome , Homeodomain Proteins/physiology , Humans , Mice , Myoblasts, Skeletal/cytology , Trans-Activators/physiology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...