Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Heart Circ Physiol ; 325(2): H203-H231, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37204871

ABSTRACT

Heart failure (HF) is a leading cause of morbidity and mortality particularly in older adults and patients with multiple metabolic comorbidities. Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome with multisystem organ dysfunction in which patients develop symptoms of HF as a result of high left ventricular (LV) diastolic pressure in the context of normal or near normal LV ejection fraction (LVEF; ≥50%). Challenges to create and reproduce a robust rodent phenotype that recapitulates the multiple comorbidities that exist in this syndrome explain the presence of various animal models that fail to satisfy all the criteria of HFpEF. Using a continuous infusion of angiotensin II and phenylephrine (ANG II/PE), we demonstrate a strong HFpEF phenotype satisfying major clinically relevant manifestations and criteria of this pathology, including exercise intolerance, pulmonary edema, concentric myocardial hypertrophy, diastolic dysfunction, histological signs of microvascular impairment, and fibrosis. Conventional echocardiographic analysis of diastolic dysfunction identified early stages of HFpEF development and speckle tracking echocardiography analysis including the left atrium (LA) identified strain abnormalities indicative of contraction-relaxation cycle impairment. Diastolic dysfunction was validated by retrograde cardiac catheterization and analysis of LV end-diastolic pressure (LVEDP). Among mice that developed HFpEF, two major subgroups were identified with predominantly perivascular fibrosis and interstitial myocardial fibrosis. In addition to major phenotypic criteria of HFpEF that were evident at early stages of this model (3 and 10 days), accompanying RNAseq data demonstrate activation of pathways associated with myocardial metabolic changes, inflammation, activation of extracellular matrix (ECM) deposition, microvascular rarefaction, and pressure- and volume-related myocardial stress.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is an emerging epidemic affecting up to half of patients with heart failure. Here we used a chronic angiotensin II/phenylephrine (ANG II/PE) infusion model and instituted an updated algorithm for HFpEF assessment. Given the simplicity in generating this model, it may become a useful tool for investigating pathogenic mechanisms, identification of diagnostic markers, and for drug discovery aimed at both prevention and treatment of HFpEF.


Subject(s)
Cardiomyopathies , Heart Failure , Animals , Mice , Heart Failure/drug therapy , Stroke Volume/physiology , Angiotensin II , Ventricular Function, Left/physiology , Disease Models, Animal , Fibrosis , Phenylephrine
2.
PLoS Biol ; 17(5): e3000245, 2019 05.
Article in English | MEDLINE | ID: mdl-31086360

ABSTRACT

Lysosomes are ubiquitous acidified organelles that degrade intracellular and extracellular material trafficked via multiple pathways. Lysosomes also sense cellular nutrient levels to regulate target of rapamycin (TOR) kinase, a signaling enzyme that drives growth and suppresses activity of the MiT/TFE family of transcription factors that control biogenesis of lysosomes. In this study, we subjected worms lacking basic helix-loop-helix transcription factor 30 (hlh-30), the Caenorhabditis elegans MiT/TFE ortholog, to starvation followed by refeeding to understand how this pathway regulates survival with variable nutrient supply. Loss of HLH-30 markedly impaired survival in starved larval worms and recovery upon refeeding bacteria. Remarkably, provision of simple nutrients in a completely defined medium (C. elegans maintenance medium [CeMM]), specifically glucose and linoleic acid, restored lysosomal acidification, TOR activation, and survival with refeeding despite the absence of HLH-30. Worms deficient in lysosomal lipase 2 (lipl-2), a lysosomal enzyme that is transcriptionally up-regulated in starvation in an HLH-30-dependent manner, also demonstrated increased mortality with starvation-refeeding that was partially rescued with glucose, suggesting a critical role for LIPL-2 in lipid metabolism under starvation. CeMM induced transcription of vacuolar proton pump subunits in hlh-30 mutant worms, and knockdown of vacuolar H+-ATPase 12 (vha-12) and its upstream regulator, nuclear hormone receptor 31 (nhr-31), abolished the rescue with CeMM. Loss of Ras-related GTP binding protein C homolog 1 RAGC-1, the ortholog for mammalian RagC/D GTPases, conferred starvation-refeeding lethality, and RAGC-1 overexpression was sufficient to rescue starved hlh-30 mutant worms, demonstrating a critical need for TOR activation with refeeding. These results show that HLH-30 activation is critical for sustaining survival during starvation-refeeding stress via regulating TOR. Glucose and linoleic acid bypass the requirement for HLH-30 in coupling lysosome nutrient sensing to survival.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Lysosomes/metabolism , Nutrients , Animals , Cell Nucleus/metabolism , Citric Acid Cycle , Culture Media , Energy Metabolism/genetics , Feeding Behavior , Linoleic Acid/metabolism , Lipase/metabolism , Metabolome , Mutation/genetics , Phenotype , Proton Pumps/metabolism , Starvation/metabolism , Stress, Physiological/genetics , Survival Analysis , Transcriptional Activation/genetics
3.
Circ Res ; 124(8): 1198-1213, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30739589

ABSTRACT

RATIONALE: LMNA (Lamin A/C), a nuclear membrane protein, interacts with genome through lamin-associated domains (LADs) and regulates gene expression. Mutations in the LMNA gene cause a diverse array of diseases, including dilated cardiomyopathy (DCM). DCM is the leading cause of death in laminopathies. OBJECTIVE: To identify LADs and characterize their associations with CpG methylation and gene expression in human cardiac myocytes in DCM. METHODS AND RESULTS: LMNA chromatin immunoprecipitation-sequencing, reduced representative bisulfite sequencing, and RNA-sequencing were performed in 5 control and 5 LMNA-associated DCM hearts. LADs were identified using enriched domain detector program. Genome-wide 331±77 LADs with an average size of 2.1±1.5 Mbp were identified in control human cardiac myocytes. LADs encompassed ≈20% of the genome and were predominantly located in the heterochromatin and less so in the promoter and actively transcribed regions. LADs were redistributed in DCM as evidenced by a gain of 520 and loss of 149 genomic regions. Approximately, 4500 coding genes and 800 long noncoding RNAs, whose levels correlated with the transcript levels of coding genes in cis, were differentially expressed in DCM. TP53 (tumor protein 53) was the most prominent among the dysregulated pathways. CpG sites were predominantly hypomethylated genome-wide in controls and DCM hearts, but overall CpG methylation was increased in DCM. LADs were associated with increased CpG methylation and suppressed gene expression. Integrated analysis identified genes whose expressions were regulated by LADs or CpG methylation, or by both, the latter pertained to genes involved in cell death, cell cycle, and metabolic regulation. CONCLUSIONS: LADs encompass ≈20% of the genome in human cardiac myocytes comprised several hundred coding and noncoding genes. LADs are redistributed in LMNA-associated DCM in association with markedly altered CpG methylation and gene expression. Thus, LADs through genomic alterations contribute to the pathogenesis of DCM in laminopathies.


Subject(s)
Cardiomyopathy, Dilated/genetics , DNA Methylation , Gene Expression Regulation , Lamin Type A/genetics , Myocytes, Cardiac , Adult , Cell Nucleus , Chromatin Immunoprecipitation Sequencing/methods , CpG Islands/genetics , Female , Heterochromatin/genetics , Humans , Male , Nucleic Acid Amplification Techniques , RNA/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
FASEB J ; 33(1): 652-667, 2019 01.
Article in English | MEDLINE | ID: mdl-30028636

ABSTRACT

Lipin 1 regulates glycerolipid homeostasis by acting as a phosphatidic acid phosphohydrolase (PAP) enzyme in the triglyceride-synthesis pathway and by regulating transcription factor activity. Mutations in human lipin 1 are a common cause of recurrent rhabdomyolysis in children. Mice with constitutive whole-body lipin 1 deficiency have been used to examine mechanisms connecting lipin 1 deficiency to myocyte injury. However, that mouse model is confounded by lipodystrophy not phenocopied in people. Herein, 2 muscle-specific mouse models were studied: 1) Lpin1 exon 3 and 4 deletion, resulting in a hypomorphic protein without PAP activity, but which preserved transcriptional coregulatory function; and 2) Lpin1 exon 7 deletion, resulting in total protein loss. In both models, skeletal muscles exhibited a chronic myopathy with ongoing muscle fiber necrosis and regeneration and accumulation of phosphatidic acid and, paradoxically, diacylglycerol. Additionally, lipin 1-deficient mice had abundant, but abnormal, mitochondria likely because of impaired autophagy. Finally, these mice exhibited increased plasma creatine kinase following exhaustive exercise when unfed. These data suggest that mice lacking lipin 1-mediated PAP activity in skeletal muscle may serve as a model for determining the mechanisms by which lipin 1 deficiency leads to myocyte injury and for testing potential therapeutic approaches.-Schweitzer, G. G., Collier, S. L., Chen, Z., McCommis, K. S., Pittman, S. K., Yoshino, J., Matkovich, S. J., Hsu, F.-F., Chrast, R., Eaton, J. M., Harris, T. E., Weihl, C. C., Finck, B. N. Loss of lipin 1-mediated phosphatidic acid phosphohydrolase activity in muscle leads to skeletal myopathy in mice.


Subject(s)
Disease Models, Animal , Gene Expression Regulation , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Nuclear Proteins/physiology , Phosphatidate Phosphatase/metabolism , Phosphatidic Acids/metabolism , Animals , Autophagy , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Diseases/etiology , Muscular Diseases/metabolism , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/physiology
5.
Curr Opin Cardiol ; 34(3): 246-253, 2019 05.
Article in English | MEDLINE | ID: mdl-30973833

ABSTRACT

PURPOSE OF REVIEW: Comprehensive analyses of the genome, transcriptome, proteome and metabolome are instrumental in identifying biomarkers of disease, to gain insight into mechanisms underlying the development of cardiovascular disease, and show promise for better stratifying patients according to disease subtypes. This review highlights recent 'omics' studies, including integration of multiple 'omics' that have advanced mechanistic understanding and diagnosis in humans and animal models. RECENT FINDINGS: Transcriptome-based discovery continues to be a primary method to obtain data for hypothesis generation and the understanding of disease pathogenesis has been enhanced by single cell-based methods capable of revealing heterogeneity in cellular responses. Advances in proteome coverage and quantitation of individual protein species, together with enhanced methods for detecting posttranslational modifications, have improved discovery of protein-based biomarkers. SUMMARY: High-throughput assays capable of quantitating the vast majority of any particular type of biomolecule within a tissue sample, isolated cells or plasma are now available. In order to make best use of the large amount of data that can be generated on given molecule types, as well as their interrelationships in disease, continued development of pattern-recognition algorithms ('machine learning') will be required and the subclassification of disease that is made possible by such algorithms will be likely to inform clinical practice, and vice versa.


Subject(s)
Heart Diseases , Metabolome , Animals , Biomarkers , Heart Diseases/diagnosis , Heart Diseases/genetics , Humans , Proteome , Transcriptome
6.
J Mol Cell Cardiol ; 121: 60-68, 2018 08.
Article in English | MEDLINE | ID: mdl-29969579

ABSTRACT

G-protein receptor kinases (GRKs) regulate adult hearts by modulating inotropic, chronotropic and hypertrophic signaling of 7-transmembrane spanning neurohormone receptors. GRK-mediated desensitization and downregulation of ß-adrenergic receptors has been implicated in adult heart failure; GRKs are therefore a promising therapeutic target. However, germ-line (but not cardiomyocyte-specific) GRK2 deletion provoked lethal fetal heart defects, suggesting an unexplained role for GRKs in heart development. Here we undertook to better understand the consequences of GRK deficiency on fetal heart development by creating mice and cultured murine embryonic fibroblasts (MEFs) having floxed GRK2 and GRK5 alleles on the GRK6 null background; simultaneous conditional deletion of these 3 GRK genes was achieved using Nkx2-5 Cre or adenoviral Cre, respectively. Phenotypes were related to GRK-modulated gene expression using whole-transcriptome RNA sequencing, RT-qPCR, and luciferase reporter assays. In cultured MEFs the atypical 7-transmembrane spanning protein and GRK2 substrate Smoothened (Smo) stimulated Gli-mediated transcriptional activity, which was interrupted by deleting GRK2/5/6. Mice with Nkx2-5 Cre mediated GRK2/5/6 ablation died between E15.5 and E16.5, whereas mice expressing any one of these 3 GRKs (i.e. GRK2/5, GRK2/6 or GRK5/6 deleted) were developmentally normal. GRK2/5/6 triple null mice at E14.5 exhibited left and right heart blood intermixing through single atrioventricular valves or large membranous ventricular septal defects. Hedgehog and GATA pathway gene expression promoted by Smo/Gli was suppressed in GRK2/5/6 deficient fetal hearts and MEFs. These data indicate that GRK2, GRK5 and GRK6 redundantly modulate Smo-GATA crosstalk in fetal mouse hearts, orchestrating transcriptional pathways previously linked to clinical and experimental atrioventricular canal defects. GRK modulation of Smo reflects convergence of conventional neurohormonal signaling and transcriptional regulation pathways, comprising an unanticipated mechanism for spatiotemporal orchestration of developmental gene expression in the heart.


Subject(s)
Fetal Heart/growth & development , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinases/genetics , Smoothened Receptor/genetics , Animals , Embryo, Mammalian , Embryonic Development/genetics , Fetal Heart/physiopathology , Fibroblasts/metabolism , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sequence Analysis, RNA , Transcriptome/genetics
7.
J Biol Chem ; 292(49): 20228-20239, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29021253

ABSTRACT

Small nucleolar RNAs (snoRNAs) guide chemical modifications of ribosomal and small nuclear RNAs, functions that are carried out in the nucleus. Although most snoRNAs reside in the nucleolus, a growing body of evidence indicates that snoRNAs are also present in the cytoplasm and that snoRNAs move between the nucleus and cytoplasm by a mechanism that is regulated by lipotoxic and oxidative stress. Here, in a genome-wide shRNA-based screen, we identified nuclear export factor 3 (NXF3) as a transporter that alters the nucleocytoplasmic distribution of box C/D snoRNAs from the ribosomal protein L13a (Rpl13a) locus. Using RNA-sequencing analysis, we show that NXF3 associates not only with Rpl13a snoRNAs, but also with a broad range of box C/D and box H/ACA snoRNAs. Under homeostatic conditions, gain- or loss-of-function of NXF3, but not related family member NXF1, decreases or increases cytosolic Rpl13a snoRNAs, respectively. Furthermore, treatment with the adenylyl cyclase activator forskolin diminishes cytosolic localization of the Rpl13a snoRNAs through a mechanism that is dependent on NXF3 but not NXF1. Our results provide evidence of a new role for NXF3 in regulating the distribution of snoRNAs between the nuclear and cytoplasmic compartments.


Subject(s)
Active Transport, Cell Nucleus , Nucleocytoplasmic Transport Proteins/physiology , RNA, Small Nucleolar/metabolism , RNA-Binding Proteins/physiology , Animals , Base Sequence , Cell Nucleus/metabolism , Cytoplasm/metabolism , Mice , Nucleocytoplasmic Transport Proteins/metabolism , Ribosomal Proteins
8.
Crit Care Med ; 45(3): 407-414, 2017 03.
Article in English | MEDLINE | ID: mdl-28067713

ABSTRACT

OBJECTIVES: The mechanism(s) for septic cardiomyopathy in humans is not known. To address this, we measured messenger RNA alterations in hearts from patients who died from systemic sepsis, in comparison to changed messenger RNA expression in nonfailing and failing human hearts. DESIGN: Identification of genes with altered abundance in septic cardiomyopathy, ischemic heart disease, or dilated cardiomyopathy, in comparison to nonfailing hearts. SETTING: ICUs at Barnes-Jewish Hospital, St. Louis, MO. PATIENTS: Twenty sepsis patients, 11 ischemic heart disease, nine dilated cardiomyopathy, and 11 nonfailing donors. INTERVENTIONS: None other than those performed as part of patient care. MEASUREMENTS AND MAIN RESULTS: Messenger RNA expression levels for 198 mitochondrially localized energy production components, including Krebs cycle and electron transport genes, decreased by 43% ± 5% (mean ± SD). Messenger RNAs for nine genes responsible for sarcomere contraction and excitation-contraction coupling decreased by 43% ± 4% in septic hearts. Surprisingly, the alterations in messenger RNA levels in septic cardiomyopathy were both distinct from and more profound than changes in messenger RNA levels in the hearts of patients with end-stage heart failure. CONCLUSIONS: The expression profile of messenger RNAs in the heart of septic patients reveals striking decreases in expression levels of messenger RNAs that encode proteins involved in cardiac energy production and cardiac contractility and is distinct from that observed in patients with heart failure. Although speculative, the global nature of the decreases in messenger RNA expression for genes involved in cardiac energy production and contractility suggests that these changes may represent a short-term adaptive response of the heart in response to acute change in cardiovascular homeostasis.


Subject(s)
Cardiomyopathies/genetics , Down-Regulation , RNA, Messenger/metabolism , Sepsis/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Cardiomyopathies/microbiology , Cardiomyopathy, Dilated/genetics , Citric Acid Cycle/genetics , Electron Transport/genetics , Female , Humans , Male , Middle Aged , Mitochondria, Heart/genetics , Mitochondria, Heart/physiology , Myocardial Ischemia/genetics , Sarcomeres/genetics , Sarcomeres/physiology , Sepsis/complications , Sepsis/physiopathology
9.
Circ Res ; 117(4): 346-51, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26038571

ABSTRACT

RATIONALE: The role of Parkin in hearts is unclear. Germ-line Parkin knockout mice have normal hearts, but Parkin is protective in cardiac ischemia. Parkin-mediated mitophagy is reportedly either irrelevant, or a major factor, in the lethal cardiomyopathy evoked by cardiac myocyte-specific interruption of dynamin-related protein 1 (Drp1)-mediated mitochondrial fission. OBJECTIVE: To understand the role of Parkin-mediated mitophagy in normal and mitochondrial fission-defective adult mouse hearts. METHODS AND RESULTS: Parkin mRNA and protein were present at low levels in normal mouse hearts, but were upregulated after cardiac myocyte-directed Drp1 gene deletion in adult mice. Alone, forced cardiac myocyte Parkin overexpression activated mitophagy without adverse effects. Likewise, cardiac myocyte-specific Parkin deletion evoked no adult cardiac phenotype, revealing no essential function for, and tolerance of, Parkin-mediated mitophagy in normal hearts. Concomitant conditional Parkin deletion with Drp1 ablation in adult mouse hearts prevented Parkin upregulation in mitochondria of fission-defective hearts, also increasing 6-week survival, improving ventricular ejection performance, mitigating adverse cardiac remodeling, and decreasing cardiac myocyte necrosis and replacement fibrosis. Underlying the Parkin knockout rescue was suppression of Drp1-induced hyper-mitophagy, assessed as ubiquitination of mitochondrial proteins and mitochondrial association of autophagosomal p62/sequestosome 1 (SQSTM1) and processed microtubule-associated protein 1 light chain 3 (LC3-II). Consequently, mitochondrial content of Drp1-deficient hearts was preserved. Parkin deletion did not alter characteristic mitochondrial enlargement of Drp1-deficient cardiac myocytes. CONCLUSIONS: Parkin is rare in normal hearts and dispensable for constitutive mitophagic quality control. Ablating Drp1 in adult mouse cardiac myocytes not only interrupts mitochondrial fission, but also markedly upregulates Parkin, thus provoking mitophagic mitochondrial depletion that contributes to the lethal cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Mitophagy , Myocardium/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Dynamins/genetics , Dynamins/metabolism , Fibrosis , Gene Expression Regulation , Genetic Predisposition to Disease , Mice, Knockout , Mitochondria, Heart/ultrastructure , Myocardium/ultrastructure , Necrosis , Phenotype , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ventricular Function, Left , Ventricular Remodeling
10.
Circ Res ; 116(5): e28-39, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25605649

ABSTRACT

RATIONALE: Sustained activation of Gαq transgenic (Gq) signaling during pressure overload causes cardiac hypertrophy that ultimately progresses to dilated cardiomyopathy. The molecular events that drive hypertrophy decompensation are incompletely understood. Ca(2+)/calmodulin-dependent protein kinase II δ (CaMKIIδ) is activated downstream of Gq, and overexpression of Gq and CaMKIIδ recapitulates hypertrophy decompensation. OBJECTIVE: To determine whether CaMKIIδ contributes to hypertrophy decompensation provoked by Gq. METHODS AND RESULTS: Compared with Gq mice, compound Gq/CaMKIIδ knockout mice developed a similar degree of cardiac hypertrophy but exhibited significantly improved left ventricular function, less cardiac fibrosis and cardiomyocyte apoptosis, and fewer ventricular arrhythmias. Markers of oxidative stress were elevated in mitochondria from Gq versus wild-type mice and respiratory rates were lower; these changes in mitochondrial function were restored by CaMKIIδ deletion. Gq-mediated increases in mitochondrial oxidative stress, compromised membrane potential, and cell death were recapitulated in neonatal rat ventricular myocytes infected with constitutively active Gq and attenuated by CaMKII inhibition. Deep RNA sequencing revealed altered expression of 41 mitochondrial genes in Gq hearts, with normalization of ≈40% of these genes by CaMKIIδ deletion. Uncoupling protein 3 was markedly downregulated in Gq or by Gq expression in neonatal rat ventricular myocytes and reversed by CaMKIIδ deletion or inhibition, as was peroxisome proliferator-activated receptor α. The protective effects of CaMKIIδ inhibition on reactive oxygen species generation and cell death were abrogated by knock down of uncoupling protein 3. Conversely, restoration of uncoupling protein 3 expression attenuated reactive oxygen species generation and cell death induced by CaMKIIδ. Our in vivo studies further demonstrated that pressure overload induced decreases in peroxisome proliferator-activated receptor α and uncoupling protein 3, increases in mitochondrial protein oxidation, and hypertrophy decompensation, which were attenuated by CaMKIIδ deletion. CONCLUSIONS: Mitochondrial gene reprogramming induced by CaMKIIδ emerges as an important mechanism contributing to mitotoxicity in decompensating hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cardiomegaly/enzymology , Cardiomyopathy, Dilated/etiology , Heart Failure/etiology , Mitochondria, Heart/physiology , Acetylcysteine/pharmacology , Animals , Apoptosis , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/deficiency , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cardiomegaly/physiopathology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Disease Progression , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Gene Expression Profiling , Heart Failure/physiopathology , Ion Channels/biosynthesis , Ion Channels/genetics , Ion Channels/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Mitochondrial Proteins/physiology , Myocytes, Cardiac/metabolism , Oxidative Stress , PPAR alpha/biosynthesis , PPAR alpha/genetics , Point Mutation , Pressure , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Reactive Oxygen Species , Sequence Analysis, RNA , Sulfonamides/pharmacology , Transfection , Uncoupling Protein 3
11.
Proc Natl Acad Sci U S A ; 111(33): 12264-9, 2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25071214

ABSTRACT

The vast majority of mammalian DNA does not encode for proteins but instead is transcribed into noncoding (nc)RNAs having diverse regulatory functions. The poorly characterized subclass of long ncRNAs (lncRNAs) can epigenetically regulate protein-coding genes by interacting locally in cis or distally in trans. A few reports have implicated specific lncRNAs in cardiac development or failure, but precise details of lncRNAs expressed in hearts and how their expression may be altered during embryonic heart development or by adult heart disease is unknown. Using comprehensive quantitative RNA sequencing data from mouse hearts, livers, and skin cells, we identified 321 lncRNAs present in the heart, 117 of which exhibit a cardiac-enriched pattern of expression. By comparing lncRNA profiles of normal embryonic (∼E14), normal adult, and hypertrophied adult hearts, we defined a distinct fetal lncRNA abundance signature that includes 157 lncRNAs differentially expressed compared with adults (fold-change ≥ 50%, false discovery rate = 0.02) and that was only poorly recapitulated in hypertrophied hearts (17 differentially expressed lncRNAs; 13 of these observed in embryonic hearts). Analysis of protein-coding mRNAs from the same samples identified 22 concordantly and 11 reciprocally regulated mRNAs within 10 kb of dynamically expressed lncRNAs, and reciprocal relationships of lncRNA and mRNA levels were validated for the Mccc1 and Relb genes using in vitro lncRNA knockdown in C2C12 cells. Network analysis suggested a central role for lncRNAs in modulating NFκB- and CREB1-regulated genes during embryonic heart growth and identified multiple mRNAs within these pathways that are also regulated, but independently of lncRNAs.


Subject(s)
Epigenesis, Genetic , Heart/embryology , RNA, Long Noncoding/genetics , Transcription, Genetic/genetics , Animals , Mice
12.
J Biol Chem ; 290(18): 11741-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25792744

ABSTRACT

Small nucleolar RNAs (snoRNAs) guide nucleotide modifications of cellular RNAs in the nucleus. We previously showed that box C/D snoRNAs from the Rpl13a locus are unexpected mediators of physiologic oxidative stress, independent of their predicted ribosomal RNA modifications. Here we demonstrate that oxidative stress induced by doxorubicin causes rapid cytoplasmic accumulation of the Rpl13a snoRNAs through a mechanism that requires superoxide and a nuclear splice variant of NADPH oxidase. RNA-sequencing analysis reveals that box C/D snoRNAs as a class are present in the cytoplasm, where their levels are dynamically regulated by NADPH oxidase. These findings suggest that snoRNAs may orchestrate the response to environmental stress through molecular interactions outside of the nucleus.


Subject(s)
Cytosol/metabolism , NADPH Oxidases/metabolism , RNA, Small Nucleolar/metabolism , Animals , Biocatalysis , Biological Transport/drug effects , Cytosol/drug effects , Doxorubicin/pharmacology , Oxidative Stress/drug effects , RNA, Small Nucleolar/genetics , Rats , Ribosomal Proteins/genetics , Superoxides/metabolism
13.
Curr Opin Cardiol ; 31(3): 242-8, 2016 May.
Article in English | MEDLINE | ID: mdl-27054504

ABSTRACT

PURPOSE OF REVIEW: Genome-wide analysis of RNA abundances (transcriptome analysis) offers great potential to identify biomarkers of disease and to gain insight into mechanisms underlying the development of heart failure. I will discuss key factors in generating and evaluating transcriptome data, and recent studies that have contributed to the understanding of cardiac disease in humans and animal models. RECENT FINDINGS: Thorough assessments of RNA-sequencing performance performed across multiple laboratories have demonstrated its primacy in measuring differential RNA abundances, due in part to its wide dynamic range and accuracy. In combination with network and pathway analysis tools, this enables greater understanding of heart failure mechanisms than previously possible, and renders possible current and future investigations into the role of RNA alternative splicing. SUMMARY: Current and future acquisition of accurate, unbiased, and comprehensive transcriptome data will continue to inform the understanding of heart failure, enabling hypothesis testing as well as hypothesis generation. Transcriptome data represent a vital bridge between genomic and epigenomic variation and proteomic output; progressive integration of data from these and other domains will fully realize the potential inherent in transcriptome analyses.


Subject(s)
Gene Expression Profiling/methods , Heart Failure/metabolism , Transcriptome , Animals , Humans , Myocardium/cytology , Myocardium/metabolism , Sequence Analysis, RNA
14.
J Physiol ; 593(8): 1799-808, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25433070

ABSTRACT

Development, homeostasis and responses to stress in the heart all depend on appropriate control of mRNA expression programmes, which may be enacted at the level of DNA sequence, DNA accessibility and RNA-mediated control of mRNA output. Diverse mechanisms underlie promoter-driven transcription of coding mRNAs and their translation into protein, and the ways in which sequence alteration of DNA can make an impact on these processes have been studied for some time. The field of epigenetics explores changes in DNA structure that influence its accessibility by transcriptional machinery, and we are continuing to develop our understanding of how these processes modify cardiac RNA production. In this topical review, we do not focus on how DNA sequence and methylation, and histone interactions, may alter its accessibility, but rather on newly described mechanisms by which some transcribed RNAs may alter initial transcription or downstream processing of other RNAs, involving both short non-coding RNAs (microRNAs) and long non-coding RNAs (lncRNAs). Here we present examples of how these two classes of non-coding RNAs mediate widespread effects on cardiac transcription and protein output in processes for which we use the broad term 'epitranscriptional regulation' and that are complementary to the DNA methylation and histone modification events studied by classical epigenetics.


Subject(s)
Cardiovascular Diseases/genetics , Cardiovascular System/metabolism , Epigenesis, Genetic , Transcription, Genetic , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular System/growth & development , Cardiovascular System/pathology , DNA Methylation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism
16.
Circ Res ; 113(1): 62-71, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23625950

ABSTRACT

RATIONALE: MicroRNAs modestly suppress their direct mRNA targets, and these direct effects are amplified by modulation of gene transcription pathways. Consequently, indirect mRNA modulatory effects of microRNAs to increase or decrease mRNAs greatly outnumber direct target suppressions. Because microRNAs are products of transcription, the potential exists for microRNAs that regulate transcription to regulate other microRNAs. OBJECTIVE: Determine whether cardiac-expressed microRNAs regulate expression of other cardiac microRNAs, and measure the impact of microRNA-mediated microRNA regulation on indirect regulation of nontarget mRNAs. METHODS AND RESULTS: Transgenic expression of pre-microRNAs was used to generate mouse hearts expressing 6- to 16-fold normal levels of microRNA (miR)-143, miR-378, and miR-499. Genome-wide mRNA and microRNA signatures were established using deep sequencing; expression profiles provoked by each microRNA were defined. miR-143 suppressed its direct cardiac mRNA target hexokinase 2, but exhibited little indirect target regulation and did not regulate other cardiac microRNAs. Both miR-378 and miR-499 indirectly regulated hundreds of cardiac mRNAs and 15 to 30 cardiac microRNAs. MicroRNA overexpression did not alter normal processing of either transgenic or endogenous cardiac microRNAs, and microRNA-mediated regulation of other microRNAs encoded within parent genes occurred in tandem with parent mRNAs. MicroRNA regulation by miR-378 and miR-499 was stimulus specific, and contributed to observed mRNA downregulation. CONCLUSIONS: MicroRNAs that modulate cardiac transcription can indirectly regulate other microRNAs. Transcriptional modulation by microRNAs, and microRNA-mediated microRNA regulation, help explain how small direct effects of microRNAs are amplified to generate striking phenotypes.


Subject(s)
Gene Expression Regulation/genetics , MicroRNAs/physiology , Myocytes, Cardiac/metabolism , Animals , Gene Expression Profiling , Mice , Mice, Transgenic , MicroRNAs/biosynthesis , MicroRNAs/genetics , Phenotype , RNA, Messenger/genetics , Sequence Analysis, RNA , Transcription, Genetic
17.
J Immunol ; 191(12): 5904-13, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227772

ABSTRACT

NK cells are innate lymphocytes important for host defense against viral infections and malignancy. However, the molecular programs orchestrating NK cell activation are incompletely understood. MicroRNA-155 (miR-155) is markedly upregulated following cytokine activation of human and mouse NK cells. Surprisingly, mature human and mouse NK cells transduced to overexpress miR-155, NK cells from mice with NK cell-specific miR-155 overexpression, and miR-155(-/-) NK cells all secreted more IFN-γ compared with controls. Investigating further, we found that activated NK cells with miR-155 overexpression had increased per-cell IFN-γ with normal IFN-γ(+) percentages, whereas greater percentages of miR-155(-/-) NK cells were IFN-γ(+). In vivo murine CMV-induced IFN-γ expression by NK cells in these miR-155 models recapitulated the in vitro phenotypes. We performed unbiased RNA-induced silencing complex sequencing on wild-type and miR-155(-/-) NK cells and found that mRNAs targeted by miR-155 were enriched in NK cell activation signaling pathways. Using specific inhibitors, we confirmed these pathways were mechanistically involved in regulating IFN-γ production by miR-155(-/-) NK cells. These data indicate that miR-155 regulation of NK cell activation is complex and that miR-155 functions as a dynamic tuner for NK cell activation via both setting the activation threshold as well as controlling the extent of activation in mature NK cells. In summary, miR-155(-/-) NK cells are more easily activated, through increased expression of proteins in the PI3K, NF-κB, and calcineurin pathways, and miR-155(-/-) and 155-overexpressing NK cells exhibit increased IFN-γ production through distinct cellular mechanisms.


Subject(s)
Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/physiology , MicroRNAs/physiology , Signal Transduction/physiology , Animals , Calcineurin/physiology , Cells, Cultured , Cytomegalovirus Infections/immunology , Gene Expression Regulation/drug effects , Genes, Reporter , Genetic Vectors/genetics , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , Models, Immunological , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , RNA Interference , Recombinant Fusion Proteins/metabolism , Sequence Analysis, RNA , Specific Pathogen-Free Organisms , Transduction, Genetic , Up-Regulation
18.
Proc Natl Acad Sci U S A ; 109(48): 19864-9, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23150554

ABSTRACT

Cardiac stress responses are driven by an evolutionarily conserved gene expression program comprising dozens of microRNAs and hundreds of mRNAs. Functionalities of different individual microRNAs are being studied, but the overall purpose of interactions between stress-regulated microRNAs and mRNAs and potentially distinct roles for microRNA-mediated epigenetic and conventional transcriptional genetic reprogramming of the stressed heart are unknown. Here we used deep sequencing to interrogate microRNA and mRNA regulation in pressure-overloaded mouse hearts, and performed a genome-wide examination of microRNA-mRNA interactions during early cardiac hypertrophy. Based on abundance and regulatory patterns, cardiac microRNAs were categorized as constitutively expressed housekeeping, regulated homeostatic, or dynamic early stress-responsive microRNAs. Regulation of 62 stress-responsive cardiac microRNAs directly affected levels of only 66 mRNAs, but the global impact of microRNA-mediated epigenetic regulation was amplified by preferential targeting of mRNAs encoding transcription factors, kinases, and phosphatases exerting amplified secondary effects. Thus, an emergent cooperative property of stress-regulated microRNAs is orchestration of transcriptional and posttranslational events that help determine the stress-reactive cardiac phenotype. This global functionality explains how large end-organ effects can be induced through modest individual changes in target mRNA and protein content by microRNAs that sense and respond dynamically to a changing physiological milieu.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Myocardium/metabolism , Stress, Physiological/genetics , Transcription, Genetic , Animals , Mice
19.
J Cell Mol Med ; 18(3): 415-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24428157

ABSTRACT

Diabetic cardiomyopathy is a cascade of complex events leading to eventual failure of the heart and cardiac fibrosis being considered as one of its major causes. miR-133a is one of the most abundantly expressed microRNAs in the heart. We investigated the role of miR-133a during severe hyperglycaemia. And, our aim was to find out what role miR-133a plays during diabetes-induced cardiac fibrosis. We saw a drastic decrease in miR-133a expression in the hearts of streptozotocin-induced diabetic animals, as measured by RT-qPCR. This decrease was accompanied by an increase in the transcriptional co-activator EP300 mRNA and major markers of fibrosis [transforming growth factor-ß1, connective tissue growth factor, fibronectin (FN1) and COL4A1]; in addition, focal cardiac fibrosis assessed by Masson's trichome stain was increased. Interestingly, in diabetic mice with cardiac-specific miR-133aa overexpression, cardiac fibrosis was significantly decreased, as observed by RT-qPCR and immunoblotting of COL4A1, ELISA for FN1 and microscopic examination. Furthermore, Cardiac miR-133a overexpression prevented ERK1/2 and SMAD-2 phosphorylation. These findings show that miR-133a could be a potential therapeutic target for diabetes-induced cardiac fibrosis and related cardiac dysfunction.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , MicroRNAs/genetics , Myocardium/metabolism , Myocardium/pathology , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Biomarkers/metabolism , E1A-Associated p300 Protein/metabolism , Endothelin-1/genetics , Endothelin-1/metabolism , Fibrosis , Gene Expression Regulation , Mice , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
J Biol Chem ; 288(22): 15455-65, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23603904

ABSTRACT

Several Bcl2 family proteins are expressed both as mitochondrial-targeted full-length and as cytosolic truncated alternately spliced isoforms. Recombinantly expressed shorter Bcl2 family isoforms can heterotypically bind to and prevent mitochondrial localization of their full-length analogs, thus suppressing their activity by sequestration. This "sponge" role requires 1:1 expression stoichiometry; absent this an alternate role is suggested. Here, RNA sequencing revealed coordinate regulation of BH3-only protein Nix/Bnip3L (Nix) and its alternately spliced soluble form (sNix) in hearts, but relative sNix/Nix expression of ∼1:10. Accordingly, we examined other putative functions of sNix. Although Nix expressed in H9c2 rat myoblasts localized to mitochondria, sNix showed variable cytoplasmic and nuclear distribution. Tumor necrosis factor α (TNFα) induced rapid and complete sNix nucleoplasmic translocation concomitant with nuclear translocation of the p65/RelA subunit of NFκB. sNix co-localized and co-precipitated with p65/RelA after TNFα stimulation; TNFα-induced sNix nuclear translocation did not occur in p65/RelA null murine embryonic fibroblasts. ChIP sequencing of TNFα-stimulated H9c2 cells revealed sNix suppression of p65/RelA binding to a subset of weaker DNA binding sites, accounting for its ability to alter gene expression in cultured cells and in vivo mouse hearts. These findings reveal TNFα-stimulated cytoplasmic-nuclear shuttling of the alternately spliced non-mitochondrial Nix isoform and uncover a role for sNix as a modulator of TNFα/NFκB-stimulated cardiac gene expression. Transcriptional co-regulation of sNix and Nix, combined with sNix posttranslational regulation by TNFα, comprises a previously unknown mechanism for molecular cross-talk between extrinsic death receptor and intrinsic mitochondrial apoptosis pathways.


Subject(s)
Alternative Splicing/physiology , Cell Nucleus/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Myoblasts, Cardiac/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Cell Line , Cell Nucleus/genetics , Gene Expression Regulation/physiology , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Muscle Proteins/genetics , Myoblasts, Cardiac/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL