Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
2.
Circ Res ; 110(7): 958-67, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22374132

ABSTRACT

RATIONALE: MyomiRs miR-499, miR-208a and miR-208b direct cardiac myosin gene expression. Sequence complementarity between miRs and their mRNA targets determines miR effects, but the functional consequences of human myomiR sequence variants are unknown. OBJECTIVE: To identify and investigate mutations in human myomiRs in order to better understand how and to what extent naturally-occurring sequence variation can impact miR-mRNA targeting and end-organ function. METHODS AND RESULTS: Screening of ≈2,600 individual DNAs for myomiR sequence variants identified a rare mutation of miR-499, u17c in the 3' end, well outside the seed region thought to determine target recognition. In vitro luciferase reporter analysis showed that the 3' miR-499 mutation altered suppression of a subset of artificial and natural mRNA targets. Cardiac-specific transgenic expression was used to compare consequences of wild-type and mutant miR-499. Both wild-type and mutant miR-499 induced heart failure in mice, but miR-499 c17 misdirected recruitment of a subset of miR-499 target mRNAs to cardiomyocyte RNA-induced silencing complexes, altering steady-state cardiac mRNA and protein make-up and favorably impacting cardiac function. In vitro analysis of miR-499 target site mutations and modeling of binding energies revealed abnormal miR-mRNA duplex configurations induced by the c17 mutation. CONCLUSIONS: A naturally occurring miR-499 mutation outside the critical seed sequence modifies mRNA targeting and end-organ function. This first description of in vivo effects from a natural human miR mutation outside the seed sequence supports comprehensive studies of individual phenotypes or disease-modification conferred by miR mutations.


Subject(s)
Heart/physiopathology , MicroRNAs/genetics , Mutation/genetics , Myocardium/metabolism , RNA, Messenger/metabolism , Animals , Base Sequence , Disease Models, Animal , Gene Expression Profiling , Heart Failure/genetics , Heart Failure/metabolism , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Molecular Sequence Data , Myosins/genetics , Myosins/metabolism , Proteomics
3.
Circ Res ; 111(5): 521-31, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22752967

ABSTRACT

RATIONALE: MicroRNA-499 and other members of the myomiR family regulate myosin isoforms in pressure-overload hypertrophy. miR-499 expression varies in human disease, but results of mouse cardiac miR-499 overexpression are inconsistent, either protecting against ischemic damage or aggravating cardiomyopathy after pressure overload. Likewise, there is disagreement over direct and indirect cardiac mRNAs targeted in vivo by miR-499. OBJECTIVE: To define the associations between regulated miR-499 level in clinical and experimental heart disease and modulation of its predicted mRNA targets and to determine the consequences of increased cardiac miR-499 on direct mRNA targeting, indirect mRNA modulation, and on myocardial protein content and posttranslational modification. METHODS AND RESULTS: miR-499 levels were increased in failing and hypertrophied human hearts and associated with decreased levels of predicted target mRNAs. Likewise, miR-499 is increased in Gq-mediated murine cardiomyopathy. Forced cardiomyocyte expression of miR-499 at levels comparable to human cardiomyopathy induced progressive murine heart failure and exacerbated cardiac remodeling after pressure overloading. Genome-wide RNA-induced silencing complex and RNA sequencing identified 67 direct, and numerous indirect, cardiac mRNA targets, including Akt and MAPKs. Myocardial proteomics identified alterations in protein phosphorylation linked to the miR-499 cardiomyopathy phenotype, including of heat shock protein 90 and protein serine/threonine phosphatase 1-α. CONCLUSIONS: miR-499 is increased in human and murine cardiac hypertrophy and cardiomyopathy, is sufficient to cause murine heart failure, and accelerates maladaptation to pressure overloading. The deleterious effects of miR-499 reflect the cumulative consequences of direct and indirect mRNA regulation, modulation of cardiac kinase and phosphatase pathways, and higher-order effects on posttranslational modification of myocardial proteins.


Subject(s)
Cardiomyopathies , Heart Failure , MAP Kinase Signaling System/genetics , MicroRNAs/physiology , Aging/physiology , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Disease Models, Animal , Down-Regulation/physiology , Gene Expression Profiling , HSP90 Heat-Shock Proteins/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , MAP Kinase Signaling System/physiology , Mice , Mice, Transgenic , Phosphorylation/physiology , Protein Phosphatase 1/metabolism , Protein Processing, Post-Translational/physiology , Proteomics , Transgenes/physiology
5.
Circ Res ; 108(1): 18-26, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21030712

ABSTRACT

RATIONALE: MicroRNAs (miRs) are expanding our understanding of cardiac disease and have the potential to transform cardiovascular therapeutics. One miR can target hundreds of individual mRNAs, but existing methodologies are not sufficient to accurately and comprehensively identify these mRNA targets in vivo. OBJECTIVE: To develop methods permitting identification of in vivo miR targets in an unbiased manner, using massively parallel sequencing of mouse cardiac transcriptomes in combination with sequencing of mRNA associated with mouse cardiac RNA-induced silencing complexes (RISCs). METHODS AND RESULTS: We optimized techniques for expression profiling small amounts of RNA without introducing amplification bias and applied this to anti-Argonaute 2 immunoprecipitated RISCs (RISC-Seq) from mouse hearts. By comparing RNA-sequencing results of cardiac RISC and transcriptome from the same individual hearts, we defined 1645 mRNAs consistently targeted to mouse cardiac RISCs. We used this approach in hearts overexpressing miRs from Myh6 promoter-driven precursors (programmed RISC-Seq) to identify 209 in vivo targets of miR-133a and 81 in vivo targets of miR-499. Consistent with the fact that miR-133a and miR-499 have widely differing "seed" sequences and belong to different miR families, only 6 targets were common to miR-133a- and miR-499-programmed hearts. CONCLUSIONS: RISC-sequencing is a highly sensitive method for general RISC profiling and individual miR target identification in biological context and is applicable to any tissue and any disease state.


Subject(s)
Gene Expression Profiling/methods , MicroRNAs , Myocardium/metabolism , RNA, Messenger/biosynthesis , RNA-Induced Silencing Complex , Sequence Analysis, RNA/methods , Animals , Gene Expression Regulation/physiology , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA, Messenger/genetics , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism
6.
Circ Res ; 108(1): 12-7, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21148429

ABSTRACT

RATIONALE: Mitochondria interact via actions of outer and inner membrane fusion proteins. The role of mitochondrial fusion in functioning of the heart, where mitochondria comprise ≈30% of cardiomyocyte volume and their intermyofilament spatial arrangement with other mitochondria is highly ordered, is unknown. OBJECTIVE: Model and analyze mitochondrial fusion defects in Drosophila melanogaster heart tubes with tincΔ4Gal4-directed expression of RNA interference (RNAi) for mitochondrial assembly regulatory factor (MARF) and optic atrophy (Opa)1. METHODS AND RESULTS: Live imaging analysis revealed that heart tube-specific knockdown of MARF or Opa1 increases mitochondrial morphometric heterogeneity and induces heart tube dilation with profound contractile impairment. Sarcoplasmic reticular structure was unaffected. Cardiomyocyte expression of human mitofusin (mfn)1 or -2 rescued MARF RNAi cardiomyopathy, demonstrating functional homology between Drosophila MARF and human mitofusins. Suppressing mitochondrial fusion increased compensatory expression of nuclear-encoded mitochondrial genes, indicating mitochondrial biogenesis. The MARF RNAi cardiomyopathy was prevented by transgenic expression of superoxide dismutase 1. CONCLUSIONS: Mitochondrial fusion is essential to cardiomyocyte mitochondrial function and regeneration. Reactive oxygen species are key mediators of cardiomyopathy in mitochondrial fusion-defective cardiomyocytes. Postulated mitochondrial-endoplasmic reticulum interactions mediated uniquely by mfn2 appear dispensable to functioning of the fly heart.


Subject(s)
Drosophila Proteins/metabolism , Membrane Fusion/physiology , Membrane Proteins/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Genetically Modified , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/biosynthesis , GTP Phosphohydrolases/genetics , Gene Knockdown Techniques , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mitochondria, Heart/genetics , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , RNA Interference
7.
Circ Res ; 106(1): 166-75, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19893015

ABSTRACT

RATIONALE: MicroRNA (miR)-133a regulates cardiac and skeletal muscle differentiation and plays an important role in cardiac development. Because miR-133a levels decrease during reactive cardiac hypertrophy, some have considered that restoring miR-133a levels could suppress hypertrophic remodeling. OBJECTIVE: To prevent the "normal" downregulation of miR-133a induced by an acute hypertrophic stimulus in the adult heart. METHODS AND RESULTS: miR-133a is downregulated in transverse aortic constriction (TAC) and isoproterenol-induced hypertrophy, but not in 2 genetic hypertrophy models. Using MYH6 promoter-directed expression of a miR-133a genomic precursor, increased cardiomyocyte miR-133a had no effect on postnatal cardiac development assessed by measures of structure, function, and mRNA profile. However, increased miR-133a levels increased QT intervals in surface electrocardiographic recordings and action potential durations in isolated ventricular myocytes, with a decrease in the fast component of the transient outward K+ current, I(to,f), at baseline. Transgenic expression of miR-133a prevented TAC-associated miR-133a downregulation and improved myocardial fibrosis and diastolic function without affecting the extent of hypertrophy. I(to,f) downregulation normally observed post-TAC was prevented in miR-133a transgenic mice, although action potential duration and QT intervals did not reflect this benefit. miR-133a transgenic hearts had no significant alterations of basal or post-TAC mRNA expression profiles, although decreased mRNA and protein levels were observed for the I(to,f) auxiliary KChIP2 subunit, which is not a predicted target. CONCLUSIONS: These results reveal striking differences between in vitro and in vivo phenotypes of miR expression, and further suggest that mRNA signatures do not reliably predict either direct miR targets or major miR effects.


Subject(s)
Cardiomegaly/metabolism , MicroRNAs/biosynthesis , Myocytes, Cardiac/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacology , Diastole , Electrocardiography , Fibrosis , Gene Expression Regulation/genetics , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Kv Channel-Interacting Proteins/biosynthesis , Kv Channel-Interacting Proteins/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/genetics
8.
Circulation ; 119(9): 1263-71, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19237659

ABSTRACT

BACKGROUND: Much has been learned about transcriptional control of cardiac gene expression in clinical and experimental congestive heart failure (CHF), but less is known about dynamic regulation of microRNAs (miRs) in CHF and during CHF treatment. We performed comprehensive microarray profiling of miRs and messenger RNAs (mRNAs) in myocardial specimens from human CHF with (n=10) or without (n=17) biomechanical support from left ventricular assist devices in comparison to nonfailing hearts (n=11). METHODS AND RESULTS: Twenty-eight miRs were upregulated >2.0-fold (P<0.001) in CHF, with nearly complete normalization of the heart failure miR signature by left ventricular assist device treatment. In contrast, of 444 mRNAs that were altered by >1.3-fold in failing hearts, only 29 mRNAs normalized by as much as 25% in post-left ventricular assist device hearts. Unsupervised hierarchical clustering of upregulated miRs and mRNAs with nearest centroid analysis and leave-1-out cross-validation revealed that combining the miR and mRNA signatures increased the ability of RNA profiling to serve as a clinical biomarker of diagnostic group and functional class. CONCLUSIONS: These results show that miRs are more sensitive than mRNAs to the acute functional status of end-stage heart failure, consistent with important functions for regulated miRs in the myocardial response to stress. Combined miR and mRNA profiling may have superior potential as a diagnostic and prognostic test in end-stage cardiomyopathy.


Subject(s)
Heart Failure/genetics , Heart Failure/therapy , Heart-Assist Devices , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Gene Expression Profiling , Genetic Markers , Heart Failure/diagnosis , Humans , Myocardium , Prognosis , Recovery of Function/genetics
9.
PLoS One ; 7(9): e44296, 2012.
Article in English | MEDLINE | ID: mdl-22957060

ABSTRACT

Mitochondrial fusion is essential to organelle homeostasis and organ health. Inexplicably, loss of function mutations of mitofusin 2 (Mfn2) specifically affect neurological tissue, causing Charcot Marie Tooth syndrome (CMT) and atypical optic atrophy. As CMT-linked Mfn2 mutations are predominantly within the GTPase domain, we postulated that Mfn2 mutations in other functional domains might affect non-neurological tissues. Here, we defined in vitro and in vivo consequences of rare human mutations in the poorly characterized Mfn2 HR1 domain. Human exome sequencing data identified 4 rare non-synonymous Mfn2 HR1 domain mutations, two bioinformatically predicted as damaging. Recombinant expression of these (Mfn2 M393I and R400Q) in Mfn2-null murine embryonic fibroblasts (MEFs) revealed incomplete rescue of characteristic mitochondrial fragmentation, compared to wild-type human Mfn2 (hMfn2); Mfn2 400Q uniquely induced mitochondrial fragmentation in normal MEFs. To compare Mfn2 mutation effects in neurological and non-neurological tissues in vivo, hMfn2 and the two mutants were expressed in Drosophila eyes or heart tubes made deficient in endogenous fly mitofusin (dMfn) through organ-specific RNAi expression. The two mutants induced similar Drosophila eye phenotypes: small eyes and an inability to rescue the eye pathology induced by suppression of dMfn. In contrast, Mfn2 400Q induced more severe cardiomyocyte mitochondrial fragmentation and cardiac phenotypes than Mfn2 393I, including heart tube dilation, depressed fractional shortening, and progressively impaired negative geotaxis. These data reveal a central functional role for Mfn2 HR1 domains, describe organ-specific effects of two Mfn2 HR1 mutations, and strongly support prospective studies of Mfn2 400Q in heritable human heart disease of unknown genetic etiology.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mutation , Myocardium/pathology , Retina/pathology , Animals , Base Sequence , Computational Biology/methods , Drosophila Proteins/metabolism , Fibroblasts/metabolism , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mitochondrial Proteins/metabolism , Models, Genetic , Molecular Sequence Data , Phenotype , Recombinant Proteins/metabolism , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL