Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 9(1): 5230, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531796

ABSTRACT

Left ventricular hypertrophy (LVH) is a major risk factor for cardiovascular morbidity and mortality. Pathological LVH engages transcriptional programs including reactivation of canonical fetal genes and those inducing fibrosis. Histone lysine demethylases (KDMs) are emerging regulators of transcriptional reprogramming in cancer, though their potential role in abnormal heart growth and fibrosis remains little understood. Here, we investigate gain and loss of function of an H3K9me2 specific demethylase, Kdm3a, and show it promotes LVH and fibrosis in response to pressure-overload. Cardiomyocyte KDM3A activates Timp1 transcription with pro-fibrotic activity. By contrast, a pan-KDM inhibitor, JIB-04, suppresses pressure overload-induced LVH and fibrosis. JIB-04 inhibits KDM3A and suppresses the transcription of fibrotic genes that overlap with genes downregulated in Kdm3a-KO mice versus WT controls. Our study provides genetic and biochemical evidence for a pro-hypertrophic function of KDM3A and proof-of principle for pharmacological targeting of KDMs as an effective strategy to counter LVH and pathological fibrosis.


Subject(s)
Cardiomegaly/genetics , Gene Expression Regulation/genetics , Histone Demethylases/genetics , Myocardium/metabolism , Aminopyridines/pharmacology , Animals , Animals, Newborn , Cardiomegaly/enzymology , Cells, Cultured , Fibrosis/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Hydrazones/pharmacology , Mice, Knockout , Mice, Transgenic , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Rats, Sprague-Dawley
2.
Sci Adv ; 4(1): eaap9004, 2018 01.
Article in English | MEDLINE | ID: mdl-29404407

ABSTRACT

Genome editing with CRISPR/Cas9 is a promising new approach for correcting or mitigating disease-causing mutations. Duchenne muscular dystrophy (DMD) is associated with lethal degeneration of cardiac and skeletal muscle caused by more than 3000 different mutations in the X-linked dystrophin gene (DMD). Most of these mutations are clustered in "hotspots." There is a fortuitous correspondence between the eukaryotic splice acceptor and splice donor sequences and the protospacer adjacent motif sequences that govern prokaryotic CRISPR/Cas9 target gene recognition and cleavage. Taking advantage of this correspondence, we screened for optimal guide RNAs capable of introducing insertion/deletion (indel) mutations by nonhomologous end joining that abolish conserved RNA splice sites in 12 exons that potentially allow skipping of the most common mutant or out-of-frame DMD exons within or nearby mutational hotspots. We refer to the correction of DMD mutations by exon skipping as myoediting. In proof-of-concept studies, we performed myoediting in representative induced pluripotent stem cells from multiple patients with large deletions, point mutations, or duplications within the DMD gene and efficiently restored dystrophin protein expression in derivative cardiomyocytes. In three-dimensional engineered heart muscle (EHM), myoediting of DMD mutations restored dystrophin expression and the corresponding mechanical force of contraction. Correcting only a subset of cardiomyocytes (30 to 50%) was sufficient to rescue the mutant EHM phenotype to near-normal control levels. We conclude that abolishing conserved RNA splicing acceptor/donor sites and directing the splicing machinery to skip mutant or out-of-frame exons through myoediting allow correction of the cardiac abnormalities associated with DMD by eliminating the underlying genetic basis of the disease.


Subject(s)
Gene Editing , Genome, Human , Muscular Dystrophy, Duchenne/genetics , Mutation/genetics , Myocardium/pathology , Tissue Engineering/methods , Base Sequence , Dystrophin/genetics , Exons/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
Cardiovasc Res ; 114(1): 90-102, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29036556

ABSTRACT

Aims: Duchenne muscular dystrophy (DMD) is an inherited devastating muscle disease with severe and often lethal cardiac complications. Emerging evidence suggests that the evolution of the pathology in DMD is accompanied by the accumulation of mitochondria with defective structure and function. Here, we investigate whether defects in the housekeeping autophagic pathway contribute to mitochondrial and metabolic dysfunctions in dystrophic cardiomyopathy. Methods and results: We employed various biochemical and imaging techniques to assess mitochondrial structure and function as well as to evaluate autophagy, and specific mitochondrial autophagy (mitophagy), in hearts of mdx mice, an animal model of DMD. Our results indicate substantial structural damage of mitochondria and a significant decrease in ATP production in hearts of mdx animals, which developed cardiomyopathy. In these hearts, we also detected enhanced autophagy but paradoxically, mitophagy appeared to be suppressed. In addition, we found decreased levels of several proteins involved in the PINK1/PARKIN mitophagy pathway as well as an insignificant amount of PARKIN protein phosphorylation at the S65 residue upon induction of mitophagy. Conclusions: Our results suggest faulty mitophagy in dystrophic hearts due to defects in the PINK1/PARKIN pathway.


Subject(s)
Autophagy , Cardiomyopathies/enzymology , Mitochondria, Heart/enzymology , Mitophagy , Muscular Dystrophy, Duchenne/complications , Myocytes, Cardiac/enzymology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cellular Senescence , Disease Models, Animal , Mice, Inbred mdx , Microtubule-Associated Proteins/metabolism , Mitochondria, Heart/ultrastructure , Muscular Dystrophy, Duchenne/enzymology , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/ultrastructure , Phosphorylation , Signal Transduction
4.
JCI Insight ; 2(18)2017 09 21.
Article in English | MEDLINE | ID: mdl-28931764

ABSTRACT

Dystrophin maintains the integrity of striated muscles by linking the actin cytoskeleton with the cell membrane. Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD) that result in progressive, debilitating muscle weakness, cardiomyopathy, and a shortened lifespan. Mutations of dystrophin that disrupt the amino-terminal actin-binding domain 1 (ABD-1), encoded by exons 2-8, represent the second-most common cause of DMD. In the present study, we compared three different strategies for CRISPR/Cas9 genome editing to correct mutations in the ABD-1 region of the DMD gene by deleting exons 3-9, 6-9, or 7-11 in human induced pluripotent stem cells (iPSCs) and by assessing the function of iPSC-derived cardiomyocytes. All three exon deletion strategies enabled the expression of truncated dystrophin protein and restoration of cardiomyocyte contractility and calcium transients to varying degrees. We show that deletion of exons 3-9 by genomic editing provides an especially effective means of correcting disease-causing ABD-1 mutations. These findings represent an important step toward eventual correction of common DMD mutations and provide a means of rapidly assessing the expression and function of internally truncated forms of dystrophin-lacking portions of ABD-1.


Subject(s)
Actins/metabolism , Dystrophin/metabolism , Gene Editing , Muscular Dystrophy, Duchenne/genetics , Mutation , CRISPR-Cas Systems , Dystrophin/genetics , Exons , Humans , Induced Pluripotent Stem Cells/cytology , Protein Binding , Protein Domains
5.
Stem Cells ; 35(3): 597-610, 2017 03.
Article in English | MEDLINE | ID: mdl-27734557

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by the loss of the protein dystrophin, leading to muscle fragility, progressive weakening, and susceptibility to mechanical stress. Although dystrophin-negative mdx mouse models have classically been used to study DMD, phenotypes appear mild compared to patients. As a result, characterization of muscle pathology, especially in the heart, has proven difficult. We report that injection of mdx embryonic stem cells (ESCs) into Wild Type blastocysts produces adult mouse chimeras with severe DMD phenotypes in the heart and skeletal muscle. Inflammation, regeneration and fibrosis are observed at the whole organ level, both in dystrophin-negative and dystrophin-positive portions of the chimeric tissues. Skeletal and cardiac muscle function are also decreased to mdx levels. In contrast to mdx heterozygous carriers, which show no significant phenotypes, these effects are even observed in chimeras with low levels of mdx ESC incorporation (10%-30%). Chimeric mice lack typical compensatory utrophin upregulation, and show pathological remodeling of Connexin-43. In addition, dystrophin-negative and dystrophin-positive isolated cardiomyocytes show augmented calcium response to mechanical stress, similar to mdx cells. These global effects highlight a novel role of mdx ESCs in triggering muscular dystrophy even when only low amounts are present. Stem Cells 2017;35:597-610.


Subject(s)
Aging/pathology , Chimera/metabolism , Embryonic Stem Cells/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Myocardium/pathology , Animals , Calcium/metabolism , Connexin 43/metabolism , Dystrophin/metabolism , Female , Heart Function Tests , Humans , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Myocytes, Cardiac/metabolism , Regeneration
6.
Sci Signal ; 9(422): ra34, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27048565

ABSTRACT

Altering chromatin structure through histone posttranslational modifications has emerged as a key driver of transcriptional responses in cells. Modulation of these transcriptional responses by pharmacological inhibition of class I histone deacetylases (HDACs), a group of chromatin remodeling enzymes, has been successful in blocking the growth of some cancer cell types. These inhibitors also attenuate the pathogenesis of pathological cardiac remodeling by blunting and even reversing pathological hypertrophy. The mechanistic target of rapamycin (mTOR) is a critical sensor and regulator of cell growth that, as part of mTOR complex 1 (mTORC1), drives changes in protein synthesis and metabolism in both pathological and physiological hypertrophy. We demonstrated through pharmacological and genetic methods that inhibition of class I HDACs suppressed pathological cardiac hypertrophy through inhibition of mTOR activity. Mice genetically silenced for HDAC1 and HDAC2 had a reduced hypertrophic response to thoracic aortic constriction (TAC) and showed reduced mTOR activity. We determined that the abundance of tuberous sclerosis complex 2 (TSC2), an mTOR inhibitor, was increased through a transcriptional mechanism in cardiomyocytes when class I HDACs were inhibited. In neonatal rat cardiomyocytes, loss of TSC2 abolished HDAC-dependent inhibition of mTOR activity, and increased expression of TSC2 was sufficient to reduce hypertrophy in response to phenylephrine. These findings point to mTOR and TSC2-dependent control of mTOR as critical components of the mechanism by which HDAC inhibitors blunt pathological cardiac growth. These results also suggest a strategy to modulate mTOR activity and facilitate the translational exploitation of HDAC inhibitors in heart disease.


Subject(s)
Cardiomegaly/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Animals, Newborn , Blotting, Western , Cardiomegaly/genetics , Cell Line , Cells, Cultured , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Peptides, Cyclic/pharmacology , RNA Interference , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
7.
Circulation ; 133(17): 1668-87, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-26984939

ABSTRACT

BACKGROUND: The clinical use of doxorubicin is limited by cardiotoxicity. Histopathological changes include interstitial myocardial fibrosis and the appearance of vacuolated cardiomyocytes. Whereas dysregulation of autophagy in the myocardium has been implicated in a variety of cardiovascular diseases, the role of autophagy in doxorubicin cardiomyopathy remains poorly defined. METHODS AND RESULTS: Most models of doxorubicin cardiotoxicity involve intraperitoneal injection of high-dose drug, which elicits lethargy, anorexia, weight loss, and peritoneal fibrosis, all of which confound the interpretation of autophagy. Given this, we first established a model that provokes modest and progressive cardiotoxicity without constitutional symptoms, reminiscent of the effects seen in patients. We report that doxorubicin blocks cardiomyocyte autophagic flux in vivo and in cardiomyocytes in culture. This block was accompanied by robust accumulation of undegraded autolysosomes. We go on to localize the site of block as a defect in lysosome acidification. To test the functional relevance of doxorubicin-triggered autolysosome accumulation, we studied animals with diminished autophagic activity resulting from haploinsufficiency for Beclin 1. Beclin 1(+/-) mice exposed to doxorubicin were protected in terms of structural and functional changes within the myocardium. Conversely, animals overexpressing Beclin 1 manifested an amplified cardiotoxic response. CONCLUSIONS: Doxorubicin blocks autophagic flux in cardiomyocytes by impairing lysosome acidification and lysosomal function. Reducing autophagy initiation protects against doxorubicin cardiotoxicity.


Subject(s)
Autophagy/drug effects , Doxorubicin/pharmacology , Lysosomes/drug effects , Myocytes, Cardiac/drug effects , Animals , Antibiotics, Antineoplastic , Autophagy/physiology , Cell Line , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
8.
Cardiovasc Res ; 108(3): 324-34, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26503985

ABSTRACT

AIMS: Nicotinamide adenine dinucleotide oxidases (NOXs) are important contributors to cellular oxidative stress in the cardiovascular system. The NOX2 isoform is upregulated in numerous disorders, including dystrophic cardiomyopathy, where it drives the progression of the disease. However, mechanisms underlying NOX2 overexpression are still unknown. We investigated the role of microRNAs (miRs) in the regulation of NOX2 expression. METHODS AND RESULTS: Duchenne muscular dystrophy (DMD) was used as a model of cardiomyopathy. After screening with miRNA target prediction databases and following qRT-PCR analysis, we found drastic downregulation of miR-448-3p in hearts of mdx mice, an animal model of DMD. The downregulation correlated with overexpression of the Ncf1 gene, encoding the NOX2 regulatory subunit p47(phox). Specificity of Ncf1 targeting by miR-448-3p was validated by luciferase reporter assay. Silencing of miR-448-3p in wild-type mice had a dramatic effect on cellular and functional properties of cardiac muscle as assessed by western blotting, qRT-PCR, confocal imaging, echocardiography, and histology. Acute treatment of mice with LNA-miR-448 inhibitors led to increased Ncf1 expression, abnormally elevated reactive oxygen species (ROS) production and exacerbated Ca(2+) signalling in cardiomyocytes, reminiscent of features previously observed in dystrophic cardiac cells. In addition, chronic inhibition of miR-448-3p resulted in dilated cardiomyopathy and arrhythmia, hallmarks of dystrophic cardiomyopathy. CONCLUSIONS: Our studies suggest that downregulation of miR-448-3p leads to the increase in the expression of Ncf1 gene and p47(phox) protein, as well as to the substantial increase in NOX2-derived ROS production. Cellular oxidative stress subsequently triggers events that finally culminate in cardiac tissue damage and development of cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/enzymology , MicroRNAs/metabolism , Myocardium/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Calcium Signaling , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Silencing , Genetic Predisposition to Disease , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred mdx , MicroRNAs/genetics , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/genetics , Myocardium/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phenotype , Time Factors , Transfection , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...