Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(41): e2311416120, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37782781

ABSTRACT

An evolutionarily conserved region of the TDP-43 low-complexity domain (LCD) twenty residues in length can adopt either an α-helical or ß-strand conformation. When in the latter conformation, TDP-43 self-associates via the formation of a labile, cross-ß structure. Self-association can be monitored via the formation of phase-separated protein droplets. Exposure of droplets to hydrogen peroxide leads to oxidation of conserved methionine residues distributed throughout the LCD. Oxidation disassembles the cross-ß structure, thus eliminating both self-association and phase separation. Here, we demonstrate that this process reciprocally enables formation of α-helical structure in precisely the same region formerly functioning to facilitate ß-strand-mediated self-association. We further observe that the α-helical conformation allows interaction with a lipid-like detergent and that exposure to lipids enhances the ß-to-α conformational switch. We hypothesize that regulation of this oxidative switch will prove to be important to the control of localized translation within vertebrate cells. The experimental observations reported herein were heavily reliant on studies of 1,6-hexanediol, a chemical agent that selectively dissolves labile structures formed via the self-association of protein domains of low sequence complexity. This aliphatic alcohol is shown to exert its dissociative activity primarily via hydrogen-bonding interactions with carbonyl oxygen atoms of the polypeptide backbone. Such observations underscore the central importance of backbone-mediated protein:protein interactions that facilitate the self-association and phase separation of LCDs.


Subject(s)
DNA-Binding Proteins , Peptides , DNA-Binding Proteins/metabolism , Peptides/chemistry , Protein Domains , Methionine/metabolism , Oxidative Stress
2.
bioRxiv ; 2023 Aug 29.
Article in English | MEDLINE | ID: mdl-37693418

ABSTRACT

An evolutionarily conserved region of the TDP-43 low complexity domain twenty residues in length can adopt either an α-helical or ß-strand conformation. When in the latter conformation, TDP-43 self-associates via the formation of a labile, cross-ß structure. Self-association can be monitored via the formation of phase separated protein droplets. Exposure of droplets to hydrogen peroxide leads to oxidation of conserved methionine residues distributed throughout the low complexity domain. Oxidation disassembles the cross-ß structure, thus eliminating both self-association and phase separation. Here we demonstrate that this process reciprocally enables formation of α-helical structure in precisely the same region formerly functioning to facilitate ß-strand mediated self-association. We further observe that the α-helical conformation allows interaction with a lipid-like detergent, and that exposure to lipids enhances the ß-to-α conformational switch. We hypothesize that regulation of this oxidative switch will prove to be important to the control of localized translation within vertebrate cells. The experimental observations reported herein were heavily reliant on studies of 1,6-hexanediol, a chemical agent that selectively dissolves labile structures formed via the self-association of protein domains of low sequence complexity. This aliphatic alcohol is shown to exert its dissociative activity primarily via hydrogen bonding interactions with carbonyl oxygen atoms of the polypeptide backbone. Such observations underscore the central importance of backbone-mediated protein:protein interactions that facilitate the self-association and phase separation of low complexity domains. Significance Statement: The TDP-43 protein is a constituent of RNA granules involved in regulated translation. TDP-43 contains a C-terminal domain of 150 amino acids of low sequence complexity conspicuously decorated with ten methionine residues. An evolutionarily conserved region (ECR) of 20 residues within this domain can adopt either of two forms of labile secondary structure. Under normal conditions wherein methionine residues are reduced, the ECR forms a labile cross-ß structure that enables RNA granule condensation. Upon methionine oxidation, the ECR undergoes a conformational switch to become an α-helix incompatible with self-association and granule integrity. Oxidation of the TDP-43 low complexity domain is hypothesized to occur proximal to mitochondria, thus facilitating dissolution of RNA granules and activation of localized translation.

3.
Science ; 377(6601): eabn5582, 2022 07.
Article in English | MEDLINE | ID: mdl-35771920

ABSTRACT

Protein domains of low sequence complexity do not fold into stable, three-dimensional structures. Nevertheless, proteins with these sequences assist in many aspects of cell organization, including assembly of nuclear and cytoplasmic structures not surrounded by membranes. The dynamic nature of these cellular assemblies is caused by the ability of low-complexity domains (LCDs) to transiently self-associate through labile, cross-ß structures. Mechanistic studies useful for the study of LCD self-association have evolved over the past decade in the form of simple assays of phase separation. Here, we have used such assays to demonstrate that the interactions responsible for LCD self-association can be dictated by labile protein structures poised close to equilibrium between the folded and unfolded states. Furthermore, missense mutations causing Charcot-Marie-Tooth disease, frontotemporal dementia, and Alzheimer's disease manifest their pathophysiology in vitro and in cultured cell systems by enhancing the stability of otherwise labile molecular structures formed upon LCD self-association.


Subject(s)
Alzheimer Disease , Charcot-Marie-Tooth Disease , DNA-Binding Proteins , Frontotemporal Dementia , Alzheimer Disease/genetics , Cells, Cultured , Charcot-Marie-Tooth Disease/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Frontotemporal Dementia/genetics , Humans , Mutation, Missense , Protein Domains , Protein Folding , Protein Stability
4.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Article in English | MEDLINE | ID: mdl-33593918

ABSTRACT

Low complexity (LC) head domains 92 and 108 residues in length are, respectively, required for assembly of neurofilament light (NFL) and desmin intermediate filaments (IFs). As studied in isolation, these IF head domains interconvert between states of conformational disorder and labile, ß-strand-enriched polymers. Solid-state NMR (ss-NMR) spectroscopic studies of NFL and desmin head domain polymers reveal spectral patterns consistent with structural order. A combination of intein chemistry and segmental isotope labeling allowed preparation of fully assembled NFL and desmin IFs that could also be studied by ss-NMR. Assembled IFs revealed spectra overlapping with those observed for ß-strand-enriched polymers formed from the isolated NFL and desmin head domains. Phosphorylation and disease-causing mutations reciprocally alter NFL and desmin head domain self-association yet commonly impede IF assembly. These observations show how facultative structural assembly of LC domains via labile, ß-strand-enriched self-interactions may broadly influence cell morphology.


Subject(s)
Desmin/chemistry , Desmin/metabolism , Intermediate Filaments/chemistry , Intermediate Filaments/metabolism , Humans , Phosphorylation , Protein Conformation , Protein Domains
5.
Proc Natl Acad Sci U S A ; 117(38): 23510-23518, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32907935

ABSTRACT

The coiled-coil domains of intermediate filament (IF) proteins are flanked by regions of low sequence complexity. Whereas IF coiled-coil domains assume dimeric and tetrameric conformations on their own, maturation of eight tetramers into cylindrical IFs is dependent on either "head" or "tail" domains of low sequence complexity. Here we confirm that the tail domain required for assembly of Drosophila Tm1-I/C IFs functions by forming labile cross-ß interactions. These interactions are seen in polymers made from the tail domain alone, as well as in assembled IFs formed by the intact Tm1-I/C protein. The ability to visualize such interactions in situ within the context of a discrete cellular assembly lends support to the concept that equivalent interactions may be used in organizing other dynamic aspects of cell morphology.


Subject(s)
Intermediate Filament Proteins , Intermediate Filaments , Animals , Drosophila/chemistry , Drosophila/metabolism , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/ultrastructure , Intermediate Filaments/chemistry , Intermediate Filaments/metabolism , Intermediate Filaments/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Polymerization , Protein Conformation
6.
Genes Dev ; 28(15): 1641-6, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25085416

ABSTRACT

Regeneration of injured adult skeletal muscle involves fusion of activated satellite cells to form new myofibers. Myomaker is a muscle-specific membrane protein required for fusion of embryonic myoblasts, but its potential involvement in adult muscle regeneration has not been explored. We show that myogenic basic helix-loop-helix (bHLH) transcription factors induce myomaker expression in satellite cells during acute and chronic muscle regeneration. Moreover, genetic deletion of myomaker in adult satellite cells completely abolishes muscle regeneration, resulting in severe muscle destruction after injury. Myomaker is the only muscle-specific protein known to be absolutely essential for fusion of embryonic and adult myoblasts.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Regeneration/genetics , Animals , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , MyoD Protein/metabolism , Myogenin/metabolism , Promoter Regions, Genetic/genetics , Satellite Cells, Skeletal Muscle/metabolism
7.
Proc Natl Acad Sci U S A ; 110(42): 16850-5, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24082095

ABSTRACT

Myocardin-related transcription factors (MRTFs) regulate cellular contractility and motility by associating with serum response factor (SRF) and activating genes involved in cytoskeletal dynamics. We reported previously that MRTF-A contributes to pathological cardiac remodeling by promoting differentiation of fibroblasts to myofibroblasts following myocardial infarction. Here, we show that forced expression of MRTF-A in dermal fibroblasts stimulates contraction of a collagen matrix, whereas contractility of MRTF-A null fibroblasts is impaired under basal conditions and in response to TGF-ß1 stimulation. We also identify an isoxazole ring-containing small molecule, previously shown to induce smooth muscle α-actin gene expression in cardiac progenitor cells, as an agonist of myofibroblast differentiation. Isoxazole stimulates myofibroblast differentiation via induction of MRTF-A-dependent gene expression. The MRTF-SRF signaling axis is activated in response to skin injury, and treatment of dermal wounds with isoxazole accelerates wound closure and suppresses the inflammatory response. These results reveal an important role for MRTF-SRF signaling in dermal myofibroblast differentiation and wound healing and suggest that targeting MRTFs pharmacologically may prove useful in treating diseases associated with inappropriate myofibroblast activity.


Subject(s)
Cell Differentiation , Dermis/injuries , Dermis/metabolism , Gene Expression Regulation , Myofibroblasts/metabolism , Trans-Activators/metabolism , Wound Healing , Androstenols/pharmacology , Animals , Dermis/pathology , Mice , Myofibroblasts/pathology , Transforming Growth Factor beta1/pharmacology
8.
Proc Natl Acad Sci U S A ; 110(34): 13839-44, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918388

ABSTRACT

The adult mammalian heart has limited potential for regeneration. Thus, after injury, cardiomyocytes are permanently lost, and contractility is diminished. In contrast, the neonatal heart can regenerate owing to sustained cardiomyocyte proliferation. Identification of critical regulators of cardiomyocyte proliferation and quiescence represents an important step toward potential regenerative therapies. Yes-associated protein (Yap), a transcriptional cofactor in the Hippo signaling pathway, promotes proliferation of embryonic cardiomyocytes by activating the insulin-like growth factor and Wnt signaling pathways. Here we report that mice bearing mutant alleles of Yap and its paralog WW domain containing transcription regulator 1 (Taz) exhibit gene dosage-dependent cardiac phenotypes, suggesting redundant roles of these Hippo pathway effectors in establishing proper myocyte number and maintaining cardiac function. Cardiac-specific deletion of Yap impedes neonatal heart regeneration, resulting in a default fibrotic response. Conversely, forced expression of a constitutively active form of Yap in the adult heart stimulates cardiac regeneration and improves contractility after myocardial infarction. The regenerative activity of Yap is correlated with its activation of embryonic and proliferative gene programs in cardiomyocytes. These findings identify Yap as an important regulator of cardiac regeneration and provide an experimental entry point to enhance this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Heart/physiology , Myocytes, Cardiac/physiology , Phosphoproteins/metabolism , Regeneration/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Cell Cycle Proteins , DNA Primers/genetics , Echocardiography , Hippo Signaling Pathway , Histological Techniques , Mice , Mice, Transgenic , Mutation, Missense/genetics , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Tetrazolium Salts , Transcription Factors/genetics , YAP-Signaling Proteins
9.
Nature ; 499(7458): 301-5, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868259

ABSTRACT

Fusion of myoblasts is essential for the formation of multi-nucleated muscle fibres. However, the identity of muscle-specific proteins that directly govern this fusion process in mammals has remained elusive. Here we identify a muscle-specific membrane protein, named myomaker, that controls myoblast fusion. Myomaker is expressed on the cell surface of myoblasts during fusion and is downregulated thereafter. Overexpression of myomaker in myoblasts markedly enhances fusion, and genetic disruption of myomaker in mice causes perinatal death due to an absence of multi-nucleated muscle fibres. Remarkably, forced expression of myomaker in fibroblasts promotes fusion with myoblasts, demonstrating the direct participation of this protein in the fusion process. Pharmacological perturbation of the actin cytoskeleton abolishes the activity of myomaker, consistent with previous studies implicating actin dynamics in myoblast fusion. These findings reveal a long-sought myogenic fusion protein that controls mammalian myoblast fusion and provide new insights into the molecular underpinnings of muscle formation.


Subject(s)
Membrane Proteins/physiology , Muscle Development , Muscle Proteins/physiology , Muscle, Skeletal/embryology , Myoblasts/cytology , Animals , Cell Fusion , Cell Line , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/metabolism
10.
Cell ; 149(3): 671-83, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541436

ABSTRACT

Obesity, type 2 diabetes, and heart failure are associated with aberrant cardiac metabolism. We show that the heart regulates systemic energy homeostasis via MED13, a subunit of the Mediator complex, which controls transcription by thyroid hormone and other nuclear hormone receptors. MED13, in turn, is negatively regulated by a heart-specific microRNA, miR-208a. Cardiac-specific overexpression of MED13 or pharmacologic inhibition of miR-208a in mice confers resistance to high-fat diet-induced obesity and improves systemic insulin sensitivity and glucose tolerance. Conversely, genetic deletion of MED13 specifically in cardiomyocytes enhances obesity in response to high-fat diet and exacerbates metabolic syndrome. The metabolic actions of MED13 result from increased energy expenditure and regulation of numerous genes involved in energy balance in the heart. These findings reveal a role of the heart in systemic metabolic control and point to MED13 and miR-208a as potential therapeutic targets for metabolic disorders.


Subject(s)
Energy Metabolism , Insulin Resistance , MicroRNAs/metabolism , Myocardium/metabolism , Obesity/genetics , Animals , Diabetes Mellitus, Type 2 , Female , Glucose/metabolism , Heart/physiology , Homeostasis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Obesity/prevention & control
11.
Sci Signal ; 4(196): ra70, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22028467

ABSTRACT

The Hippo signaling pathway regulates growth of the heart and other tissues. Hippo pathway kinases influence the activity of various targets, including the transcriptional coactivator Yap, but the specific role of Yap in heart growth has not been investigated. We show that Yap is necessary and sufficient for embryonic cardiac growth in mice. Deletion of Yap in the embryonic mouse heart impeded cardiomyocyte proliferation, causing myocardial hypoplasia and lethality at embryonic stage 10.5. Conversely, forced expression of a constitutively active form of Yap in the embryonic heart increased cardiomyocyte number and heart size. Yap activated the insulin-like growth factor (IGF) signaling pathway in cardiomyocytes, resulting in inactivation of glycogen synthase kinase 3ß, which led to increased abundance of ß-catenin, a positive regulator of cardiac growth. Our results point to Yap as a critical downstream effector of the Hippo pathway in the control of cardiomyocyte proliferation and a nexus for coupling the IGF, Wnt, and Hippo signaling pathways with the developmental program for heart growth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Signal Transduction , Somatomedins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Blotting, Western , Cell Cycle Proteins , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Fetal Heart/growth & development , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred ICR , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Organ Size , Phosphoproteins/genetics , Rats , Rats, Sprague-Dawley , Somatomedins/genetics , YAP-Signaling Proteins , beta Catenin/genetics , beta Catenin/metabolism
12.
Circ Res ; 107(11): 1336-44, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20947829

ABSTRACT

RATIONALE: Establishment of a functional vasculature requires the interconnection and remodeling of nascent blood vessels. Precise regulation of factors that influence endothelial cell migration and function is essential for these stereotypical vascular patterning events. The secreted Slit ligands and their Robo receptors constitute a critical signaling pathway controlling the directed migration of both neurons and vascular endothelial cells during embryonic development, but the mechanisms of their regulation are incompletely understood. OBJECTIVE: To identify microRNAs regulating aspects of the Slit-Robo pathway and vascular patterning. METHODS AND RESULTS: Here, we provide evidence that microRNA (miR)-218, which is encoded by an intron of the Slit genes, inhibits the expression of Robo1 and Robo2 and multiple components of the heparan sulfate biosynthetic pathway. Using in vitro and in vivo approaches, we demonstrate that miR-218 directly represses the expression of Robo1, Robo2, and glucuronyl C5-epimerase (GLCE), and that an intact miR-218-Slit-Robo regulatory network is essential for normal vascularization of the retina. Knockdown of miR-218 results in aberrant regulation of this signaling axis, abnormal endothelial cell migration, and reduced complexity of the retinal vasculature. CONCLUSIONS: Our findings link Slit gene expression to the posttranscriptional regulation of Robo receptors and heparan sulfate biosynthetic enzymes, allowing for precise control over vascular guidance cues influencing the organization of blood vessels during development.


Subject(s)
Glycoproteins/antagonists & inhibitors , MicroRNAs/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Receptors, Immunologic/antagonists & inhibitors , Retinal Vessels/embryology , Signal Transduction/genetics , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Expression Regulation, Developmental/physiology , Glycoproteins/physiology , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/biosynthesis , Heparitin Sulfate/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Molecular Sequence Data , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Receptors, Immunologic/genetics , Retinal Vessels/physiology , Transcription, Genetic , Roundabout Proteins
13.
Circ Res ; 107(2): 294-304, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20558820

ABSTRACT

RATIONALE: Myocardial infarction (MI) results in loss of cardiac myocytes in the ischemic zone of the heart, followed by fibrosis and scar formation, which diminish cardiac contractility and impede angiogenesis and repair. Myofibroblasts, a specialized cell type that switches from a fibroblast-like state to a contractile, smooth muscle-like state, are believed to be primarily responsible for fibrosis of the injured heart and other tissues, although the transcriptional mediators of fibrosis and myofibroblast activation remain poorly defined. Myocardin-related transcription factors (MRTFs) are serum response factor (SRF) cofactors that promote a smooth muscle phenotype and are emerging as components of stress-responsive signaling. OBJECTIVE: We aimed to examine the effect of MRTF-A on cardiac remodeling and fibrosis. METHODS AND RESULTS: Here, we show that MRTF-A controls the expression of a fibrotic gene program that includes genes involved in extracellular matrix production and smooth muscle cell differentiation in the heart. In MRTF-A-null mice, fibrosis and scar formation following MI or angiotensin II treatment are dramatically diminished compared with wild-type littermates. This protective effect of MRTF-A deletion is associated with a reduction in expression of fibrosis-associated genes, including collagen 1a2, a direct transcriptional target of SRF/MRTF-A. CONCLUSIONS: We conclude that MRTF-A regulates myofibroblast activation and fibrosis in response to the renin-angiotensin system and post-MI remodeling.


Subject(s)
Cell Transdifferentiation , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Smooth Muscle/metabolism , Trans-Activators/metabolism , Ventricular Remodeling , Amides/pharmacology , Angiotensin II/administration & dosage , Animals , Base Sequence , COS Cells , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Chlorocebus aethiops , Collagen/genetics , Collagen Type I , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Male , Mice , Mice, Knockout , Molecular Sequence Data , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phenotype , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Time Factors , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription, Genetic , Transfection , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics , rho-Associated Kinases/metabolism
14.
Proc Natl Acad Sci U S A ; 107(9): 4218-23, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20142475

ABSTRACT

microRNAs (miRNAs) play key roles in modulating a variety of cellular processes through repression of mRNA targets. In a screen for miRNAs regulated by myocardin-related transcription factor-A (MRTF-A), a coactivator of serum response factor (SRF), we discovered a muscle-enriched miRNA, miR-486, controlled by an alternative promoter within intron 40 of the Ankyrin-1 gene. Transcription of miR-486 is directly controlled by SRF and MRTF-A, as well as by MyoD. Among the most strongly predicted targets of miR-486 are phosphatase and tensin homolog (PTEN) and Foxo1a, which negatively affect phosphoinositide-3-kinase (PI3K)/Akt signaling. Accordingly, PTEN and Foxo1a protein levels are reduced by miR-486 overexpression, which, in turn, enhances PI3K/Akt signaling. Similarly, we show that MRTF-A promotes PI3K/Akt signaling by up-regulating miR-486 expression. Conversely, inhibition of miR-486 expression enhances the expression of PTEN and Foxo1a and dampens signaling through the PI3K/Akt-signaling pathway. Our findings implicate miR-486 as a downstream mediator of the actions of SRF/MRTF-A and MyoD in muscle cells and as a potential modulator of PI3K/Akt signaling.


Subject(s)
MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Blotting, Northern , Electrophoretic Mobility Shift Assay , In Situ Hybridization , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Rats , Signal Transduction , Trans-Activators/metabolism
15.
Circ Res ; 106(1): 155-65, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19893013

ABSTRACT

RATIONALE: Gender differences in cardiovascular disease have long been recognized and attributed to beneficial cardiovascular actions of estrogen. Class II histone deacetylases (HDACs) act as key modulators of heart disease by repressing the activity of the myocyte enhancer factor (MEF)2 transcription factor, which promotes pathological cardiac remodeling in response to stress. Although it is proposed that HDACs additionally influence nuclear receptor signaling, the effect of class II HDACs on gender differences in cardiovascular disease remains unstudied. OBJECTIVE: We aimed to examine the effect of class II HDACs on post-myocardial infarction remodeling in male and female mice. METHODS AND RESULTS: Here we show that the absence of HDAC5 or -9 in female mice protects against maladaptive remodeling following myocardial infarction, during which there is an upregulation of estrogen-responsive genes in the heart. This genetic reprogramming coincides with a pronounced increase in expression of the estrogen receptor (ER)alpha gene, which we show to be a direct MEF2 target gene. ERalpha also directly interacts with class II HDACs. Cardioprotection resulting from the absence of HDAC5 or -9 in female mice can be attributed, at least in part, to enhanced neoangiogenesis in the infarcted region via upregulation of the ER target gene vascular endothelial growth factor-a. CONCLUSIONS: Our results reveal a novel gender-specific pathway of cardioprotection mediated by ERalpha and its regulation by MEF2 and class II HDACs.


Subject(s)
Estrogen Receptor alpha/metabolism , Histone Deacetylases/metabolism , Myocardial Infarction/metabolism , Myogenic Regulatory Factors/metabolism , Repressor Proteins/metabolism , Sex Characteristics , Animals , Estrogen Receptor alpha/genetics , Female , Histone Deacetylases/genetics , MEF2 Transcription Factors , Male , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myogenic Regulatory Factors/genetics , Neovascularization, Physiologic/genetics , Repressor Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Dev Cell ; 17(5): 662-73, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19922871

ABSTRACT

Myosin is the primary regulator of muscle strength and contractility. Here we show that three myosin genes, Myh6, Myh7, and Myh7b, encode related intronic microRNAs (miRNAs), which, in turn, control muscle myosin content, myofiber identity, and muscle performance. Within the adult heart, the Myh6 gene, encoding a fast myosin, coexpresses miR-208a, which regulates the expression of two slow myosins and their intronic miRNAs, Myh7/miR-208b and Myh7b/miR-499, respectively. miR-208b and miR-499 play redundant roles in the specification of muscle fiber identity by activating slow and repressing fast myofiber gene programs. The actions of these miRNAs are mediated in part by a collection of transcriptional repressors of slow myofiber genes. These findings reveal that myosin genes not only encode the major contractile proteins of muscle, but act more broadly to influence muscle function by encoding a network of intronic miRNAs that control muscle gene expression and performance.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Myosin Heavy Chains/genetics , Animals , Base Sequence , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line , Chlorocebus aethiops , Mice , Myosin Heavy Chains/metabolism , Myosin Type II/genetics , Myosin Type II/metabolism
17.
Genes Dev ; 23(18): 2166-78, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19720868

ABSTRACT

Vascular injury triggers dedifferentiation and cytoskeletal remodeling of smooth muscle cells (SMCs), culminating in vessel occlusion. Serum response factor (SRF) and its coactivator, myocardin, play a central role in the control of smooth muscle phenotypes by regulating the expression of cytoskeletal genes. We show that SRF and myocardin regulate a cardiovascular-specific microRNA (miRNA) cluster encoding miR-143 and miR-145. To assess the functions of these miRNAs in vivo, we systematically deleted them singly and in combination in mice. Mice lacking both miR-143 and miR-145 are viable and do not display overt abnormalities in smooth muscle differentiation, although they show a significant reduction in blood pressure due to reduced vascular tone. Remarkably, however, neointima formation in response to vascular injury is profoundly impeded in mice lacking these miRNAs, due to disarray of actin stress fibers and diminished migratory activity of SMCs. These abnormalities reflect the regulation of a cadre of modulators of SRF activity and actin dynamics by miR-143 and miR-145. Thus, miR-143 and miR-145 act as integral components of the regulatory network whereby SRF controls cytoskeletal remodeling and phenotypic switching of SMCs during vascular disease.


Subject(s)
Cytoskeleton/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Myocytes, Smooth Muscle/metabolism , Actins/metabolism , Animals , Base Sequence , Carotid Artery Injuries/metabolism , Cells, Cultured , Enhancer Elements, Genetic/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Mutation , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Rats , Sequence Alignment , Trans-Activators/metabolism
18.
Proc Natl Acad Sci U S A ; 105(35): 13027-32, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18723672

ABSTRACT

Acute myocardial infarction (MI) due to coronary artery occlusion is accompanied by a pathological remodeling response that includes hypertrophic cardiac growth and fibrosis, which impair cardiac contractility. Previously, we showed that cardiac hypertrophy and heart failure are accompanied by characteristic changes in the expression of a collection of specific microRNAs (miRNAs), which act as negative regulators of gene expression. Here, we show that MI in mice and humans also results in the dysregulation of specific miRNAs, which are similar to but distinct from those involved in hypertrophy and heart failure. Among the MI-regulated miRNAs are members of the miR-29 family, which are down-regulated in the region of the heart adjacent to the infarct. The miR-29 family targets a cadre of mRNAs that encode proteins involved in fibrosis, including multiple collagens, fibrillins, and elastin. Thus, down-regulation of miR-29 would be predicted to derepress the expression of these mRNAs and enhance the fibrotic response. Indeed, down-regulation of miR-29 with anti-miRs in vitro and in vivo induces the expression of collagens, whereas over-expression of miR-29 in fibroblasts reduces collagen expression. We conclude that miR-29 acts as a regulator of cardiac fibrosis and represents a potential therapeutic target for tissue fibrosis in general.


Subject(s)
Endomyocardial Fibrosis/genetics , Gene Expression Regulation , MicroRNAs/genetics , Myocardial Infarction/genetics , Animals , COS Cells , Chlorocebus aethiops , Collagen/genetics , Collagen/metabolism , Down-Regulation , Endomyocardial Fibrosis/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Myocardial Infarction/pathology , RNA, Messenger
19.
Proc Natl Acad Sci U S A ; 104(52): 20844-9, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18093911

ABSTRACT

The muscle-specific microRNAs, miR-1 and miR-133, play important roles in muscle growth and differentiation. Here, we show that the MEF2 transcription factor, an essential regulator of muscle development, directly activates transcription of a bicistronic primary transcript encoding miR-1-2 and 133a-1 via an intragenic muscle-specific enhancer located between the miR-1-2 and 133a-1 coding regions. This MEF2-dependent enhancer is activated in the linear heart tube during mouse embryogenesis and thereafter controls transcription throughout the atrial and ventricular chambers of the heart. MEF2 together with MyoD also regulates the miR-1-2/-133a-1 intragenic enhancer in the somite myotomes and in all skeletal muscle fibers during embryogenesis and adulthood. A similar muscle-specific intragenic enhancer controls transcription of the miR-1-1/-133a-2 locus. These findings reveal a common architecture of regulatory elements associated with the miR-1/-133 genes and underscore the central role of MEF2 as a regulator of the transcriptional and posttranscriptional pathways that control cardiac and skeletal muscle development.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/physiology , Transcription, Genetic , Animals , Gene Deletion , Heart/embryology , MEF2 Transcription Factors , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic , Myocardium/metabolism
20.
Dev Dyn ; 236(9): 2475-84, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17685485

ABSTRACT

Nkx2-5 is a homeobox containing transcription factor that is conserved and expressed in organisms that form hearts. Fruit flies lacking the gene (tinman) fail to form a dorsal vessel, mice that are homozygous null for Nkx2-5 form small, deformed hearts, and several human cardiac defects have been linked to dominant mutations in the Nkx2-5 gene. The Xenopus homologs (XNkx2-5) of two truncated forms of Nkx2-5 that have been identified in humans with congenital heart defects were used in the studies reported here. mRNAs encoding these mutations were injected into single cell Xenopus embryos, and heart development was monitored. Our results indicate that the introduction of truncated XNkx2-5 variants leads to three principle developmental defects. The atrial septum and the valve of the atrioventricular canal were both abnormal. In addition, video microscopic timing of heart contraction indicated that embryos injected with either mutant form of XNkx2-5 have conduction defects.


Subject(s)
Gene Expression Regulation, Developmental , Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Mutation , Point Mutation , Transcription Factors/genetics , Transcription Factors/physiology , Xenopus Proteins/genetics , Xenopus Proteins/physiology , Animals , Gene Deletion , Homeobox Protein Nkx-2.5 , Homozygote , Humans , Microscopy, Video , Models, Genetic , Myocardial Contraction , Protein Structure, Tertiary , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...