Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(3): e0282151, 2023.
Article in English | MEDLINE | ID: mdl-36888581

ABSTRACT

BACKGROUND: SARS-CoV-2-mediated COVID-19 may cause sudden cardiac death (SCD). Factors contributing to this increased risk of potentially fatal arrhythmias include thrombosis, exaggerated immune response, and treatment with QT-prolonging drugs. However, the intrinsic arrhythmic potential of direct SARS-CoV-2 infection of the heart remains unknown. OBJECTIVE: To assess the cellular and electrophysiological effects of direct SARS-CoV-2 infection of the heart using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS: hiPSC-CMs were transfected with recombinant SARS-CoV-2 spike protein (CoV-2 S) or CoV-2 S fused to a modified Emerald fluorescence protein (CoV-2 S-mEm). Cell morphology was visualized using immunofluorescence microscopy. Action potential duration (APD) and cellular arrhythmias were measured by whole cell patch-clamp. Calcium handling was assessed using the Fluo-4 Ca2+ indicator. RESULTS: Transfection of hiPSC-CMs with CoV-2 S-mEm produced multinucleated giant cells (syncytia) displaying increased cellular capacitance (75±7 pF, n = 10 vs. 26±3 pF, n = 10; P<0.0001) consistent with increased cell size. The APD90 was prolonged significantly from 419±26 ms (n = 10) in untransfected hiPSC-CMs to 590±67 ms (n = 10; P<0.05) in CoV-2 S-mEm-transfected hiPSC-CMs. CoV-2 S-induced syncytia displayed delayed afterdepolarizations, erratic beating frequency, and calcium handling abnormalities including calcium sparks, large "tsunami"-like waves, and increased calcium transient amplitude. After furin protease inhibitor treatment or mutating the CoV-2 S furin cleavage site, cell-cell fusion was no longer evident and Ca2+ handling returned to normal. CONCLUSION: The SARS-CoV-2 spike protein can directly perturb both the cardiomyocyte's repolarization reserve and intracellular calcium handling that may confer the intrinsic, mechanistic substrate for the increased risk of SCD observed during this COVID-19 pandemic.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Long QT Syndrome , Humans , Myocytes, Cardiac/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Calcium/metabolism , Furin/metabolism , Long QT Syndrome/metabolism , Pandemics , COVID-19/metabolism , SARS-CoV-2/metabolism , Arrhythmias, Cardiac/metabolism , Action Potentials/physiology
2.
J Virol ; 95(24): e0136821, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34613786

ABSTRACT

Severe cardiovascular complications can occur in coronavirus disease of 2019 (COVID-19) patients. Cardiac damage is attributed mostly to the aberrant host response to acute respiratory infection. However, direct infection of cardiac tissue by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) also occurs. We examined here the cardiac tropism of SARS-CoV-2 in spontaneously beating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). These cardiomyocytes express the angiotensin-converting enzyme 2 (ACE2) receptor but not the transmembrane protease serine 2 (TMPRSS2) that mediates spike protein cleavage in the lungs. Nevertheless, SARS-CoV-2 infection of hiPSC-CMs was prolific; viral transcripts accounted for about 88% of total mRNA. In the cytoplasm of infected hiPSC-CMs, smooth-walled exocytic vesicles contained numerous 65- to 90-nm particles with canonical ribonucleocapsid structures, and virus-like particles with knob-like spikes covered the cell surface. To better understand how SARS-CoV-2 spreads in hiPSC-CMs, we engineered an expression vector coding for the spike protein with a monomeric emerald-green fluorescent protein fused to its cytoplasmic tail (S-mEm). Proteolytic processing of S-mEm and the parental spike were equivalent. Live cell imaging tracked spread of S-mEm cell-to-cell and documented formation of syncytia. A cell-permeable, peptide-based molecule that blocks the catalytic site of furin and furin-like proteases abolished cell fusion. A spike mutant with the single amino acid change R682S that disrupts the multibasic furin cleavage motif was fusion inactive. Thus, SARS-CoV-2 replicates efficiently in hiPSC-CMs and furin, and/or furin-like-protease activation of its spike protein is required for fusion-based cytopathology. This hiPSC-CM platform enables target-based drug discovery in cardiac COVID-19. IMPORTANCE Cardiac complications frequently observed in COVID-19 patients are tentatively attributed to systemic inflammation and thrombosis, but viral replication has occasionally been confirmed in cardiac tissue autopsy materials. We developed an in vitro model of SARS-CoV-2 spread in myocardium using induced pluripotent stem cell-derived cardiomyocytes. In these highly differentiated cells, viral transcription levels exceeded those previously documented in permissive transformed cell lines. To better understand the mechanisms of SARS-CoV-2 spread, we expressed a fluorescent version of its spike protein that allowed us to characterize a fusion-based cytopathic effect. A mutant of the spike protein with a single amino acid mutation in the furin/furin-like protease cleavage site lost cytopathic function. Of note, the fusion activities of the spike protein of other coronaviruses correlated with the level of cardiovascular complications observed in infections with the respective viruses. These data indicate that SARS-CoV-2 may cause cardiac damage by fusing cardiomyocytes.


Subject(s)
COVID-19/virology , Myocytes, Cardiac/virology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Animals , Cathepsin B/metabolism , Cell Fusion , Chlorocebus aethiops , Embryonic Stem Cells/metabolism , Exocytosis , Humans , Induced Pluripotent Stem Cells/metabolism , Microscopy, Confocal , Serine Endopeptidases/metabolism , Vero Cells , Viral Proteins/metabolism , Virus Internalization , Virus Replication
4.
Nat Med ; 26(11): 1788-1800, 2020 11.
Article in English | MEDLINE | ID: mdl-33188278

ABSTRACT

Ribonucleoprotein (RNP) granules are biomolecular condensates-liquid-liquid phase-separated droplets that organize and manage messenger RNA metabolism, cell signaling, biopolymer assembly, biochemical reactions and stress granule responses to cellular adversity. Dysregulated RNP granules drive neuromuscular degenerative disease but have not previously been linked to heart failure. By exploring the molecular basis of congenital dilated cardiomyopathy (DCM) in genome-edited pigs homozygous for an RBM20 allele encoding the pathogenic R636S variant of human RNA-binding motif protein-20 (RBM20), we discovered that RNP granules accumulated abnormally in the sarcoplasm, and we confirmed this finding in myocardium and reprogrammed cardiomyocytes from patients with DCM carrying the R636S allele. Dysregulated sarcoplasmic RBM20 RNP granules displayed liquid-like material properties, docked at precisely spaced intervals along cytoskeletal elements, promoted phase partitioning of cardiac biomolecules and fused with stress granules. Our results link dysregulated RNP granules to myocardial cellular pathobiology and heart failure in gene-edited pigs and patients with DCM caused by RBM20 mutation.


Subject(s)
Cardiomyopathy, Dilated/genetics , Myocardium/metabolism , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics , Alleles , Animals , Cardiomyopathy, Dilated/physiopathology , Cellular Reprogramming , Disease Models, Animal , Female , Gene Editing , Humans , Male , Mutation/genetics , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Messenger/genetics , Sarcoplasmic Reticulum/genetics , Sarcoplasmic Reticulum/metabolism , Secretory Vesicles/genetics , Secretory Vesicles/metabolism , Swine
5.
Dev Biol ; 462(2): 197-207, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32197890

ABSTRACT

Three-dimensional (3D) organoid models derived from human pluripotent stem cells provide a platform for studying human development and understanding disease mechanisms. Most studies that examine biallelic inactivation of the cell cycle regulator Retinoblastoma 1 (RB1) and the link to retinoblastoma is in mice, however, less is known regarding the pathophysiological role of RB1 during human retinal development. To study the role of RB1 in early human retinal development and tumor formation, we generated retinal organoids from CRISPR/Cas9-derived RB1-null human embryonic stem cells (hESCs). We showed that RB is abundantly expressed in retinal progenitor cells in retinal organoids and loss of RB1 promotes S-phase entry. Furthermore, loss of RB1 resulted in widespread apoptosis and reduced the number of photoreceptor, ganglion, and bipolar cells. Interestingly, RB1 mutation in retinal organoids did not result in retinoblastoma formation in vitro or in the vitreous body of NOD/SCID immunodeficient mice. Together, our work identifies a crucial function for RB1 in human retinal development and suggests that RB1 deletion alone is not sufficient for tumor development, at least in human retinal organoids.


Subject(s)
Human Embryonic Stem Cells/metabolism , Retina/embryology , Retinoblastoma Binding Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/physiology , CRISPR-Cas Systems , Cell Differentiation/genetics , Human Embryonic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Organoids/cytology , Pluripotent Stem Cells/cytology , Retina/physiology , Retinal Ganglion Cells/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Retinoblastoma Binding Proteins/physiology , Ubiquitin-Protein Ligases/physiology
6.
Circulation ; 140(11): 921-936, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31220931

ABSTRACT

BACKGROUND: Polycystin-1 (PC1) is a transmembrane protein originally identified in autosomal dominant polycystic kidney disease where it regulates the calcium-permeant cation channel polycystin-2. Autosomal dominant polycystic kidney disease patients develop renal failure, hypertension, left ventricular hypertrophy, and diastolic dysfunction, among other cardiovascular disorders. These individuals harbor PC1 loss-of-function mutations in their cardiomyocytes, but the functional consequences are unknown. PC1 is ubiquitously expressed, and its experimental ablation in cardiomyocyte-specific knockout mice reduces contractile function. Here, we set out to determine the pathophysiological role of PC1 in cardiomyocytes. METHODS: Wild-type and cardiomyocyte-specific PC1 knockout mice were analyzed by echocardiography. Excitation-contraction coupling was assessed in isolated cardiomyocytes and human embryonic stem cell-derived cardiomyocytes, and functional consequences were explored in heterologous expression systems. Protein-protein interactions were analyzed biochemically and by means of ab initio calculations. RESULTS: PC1 ablation reduced action potential duration in cardiomyocytes, decreased Ca2+ transients, and myocyte contractility. PC1-deficient cardiomyocytes manifested a reduction in sarcoendoplasmic reticulum Ca2+ stores attributable to a reduced action potential duration and sarcoendoplasmic reticulum Ca2+ ATPase (SERCA) activity. An increase in outward K+ currents decreased action potential duration in cardiomyocytes lacking PC1. Overexpression of full-length PC1 in HEK293 cells significantly reduced the current density of heterologously expressed Kv4.3, Kv1.5 and Kv2.1 potassium channels. PC1 C terminus inhibited Kv4.3 currents to the same degree as full-length PC1. Additionally, PC1 coimmunoprecipitated with Kv4.3, and a modeled PC1 C-terminal structure suggested the existence of 2 docking sites for PC1 within the N terminus of Kv4.3, supporting a physical interaction. Finally, a naturally occurring human mutant PC1R4228X manifested no suppressive effects on Kv4.3 channel activity. CONCLUSIONS: Our findings uncover a role for PC1 in regulating multiple Kv channels, governing membrane repolarization and alterations in SERCA activity that reduce cardiomyocyte contractility.


Subject(s)
Action Potentials/physiology , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , TRPP Cation Channels/deficiency , Animals , Humans , Mice , Mice, Knockout , Mice, Transgenic , TRPP Cation Channels/genetics
7.
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
8.
Circ Res ; 122(6): e20-e33, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29362227

ABSTRACT

RATIONALE: The regulator of calcineurin 1 (RCAN1) inhibits CN (calcineurin), a Ca2+-activated protein phosphatase important in cardiac remodeling. In humans, RCAN1 is located on chromosome 21 in proximity to the Down syndrome critical region. The hearts and brains of Rcan1 KO mice are more susceptible to damage from ischemia/reperfusion (I/R); however, the underlying cause is not known. OBJECTIVE: Mitochondria are key mediators of I/R damage. The goal of these studies was to determine the impact of RCAN1 on mitochondrial dynamics and function. METHODS AND RESULTS: Using both neonatal and isolated adult cardiomyocytes, we show that, when RCAN1 is depleted, the mitochondrial network is more fragmented because of increased CN-dependent activation of the fission protein, DRP1 (dynamin-1-like). Mitochondria in RCAN1-depleted cardiomyocytes have reduced membrane potential, O2 consumption, and generation of reactive oxygen species, as well as a reduced capacity for mitochondrial Ca2+ uptake. RCAN1-depleted cardiomyocytes were more sensitive to I/R; however, pharmacological inhibition of CN, DRP1, or CAPN (calpains; Ca2+-activated proteases) restored protection, suggesting that in the absence of RCAN1, CAPN-mediated damage after I/R is greater because of a decrease in the capacity of mitochondria to buffer cytoplasmic Ca2+. Increasing RCAN1 levels by adenoviral infection was sufficient to enhance fusion and confer protection from I/R. To examine the impact of more modest, and biologically relevant, increases in RCAN1, we compared the mitochondrial network in induced pluripotent stem cells derived from individuals with Down syndrome to that of isogenic, disomic controls. Mitochondria were more fused, and O2 consumption was greater in the trisomic induced pluripotent stem cells; however, coupling efficiency and metabolic flexibility were compromised compared with disomic induced pluripotent stem cells. Depletion of RCAN1 from trisomic induced pluripotent stem cells was sufficient to normalize mitochondrial dynamics and function. CONCLUSIONS: RCAN1 helps maintain a more interconnected mitochondrial network, and maintaining appropriate RCAN1 levels is important to human health and disease.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Muscle Proteins/genetics , Myocardial Reperfusion Injury/genetics , Animals , Calcium-Binding Proteins , Calpain/genetics , Calpain/metabolism , Cell Line , Cells, Cultured , Dynamins/genetics , Dynamins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Oxygen/metabolism , Rats , Rats, Sprague-Dawley
9.
J Inherit Metab Dis ; 41(2): 231-238, 2018 03.
Article in English | MEDLINE | ID: mdl-29110178

ABSTRACT

Fabry disease is a glycosphingolipidosis caused by deficient activity of α-galactosidase A; it is one of a few diseases that are associated with priapism, an abnormal prolonged erection of the penis. The goal of this study was to investigate the pathogenesis of Fabry disease-associated priapism in a mouse model of the disease. We found that Fabry mice develop late-onset priapism. Neuronal nitric oxide synthase (nNOS), which was predominantly present as the 120-kDa N-terminus-truncated form, was significantly upregulated in the penis of 18-month-old Fabry mice compared to wild type controls (~fivefold). Endothelial NOS (eNOS) was also upregulated (~twofold). NO level in penile tissues of Fabry mice was significantly higher than wild type controls at 18 months. Gene transfer-mediated enzyme replacement therapy reversed abnormal nNOS expression in the Fabry mouse penis. The penile nNOS level was restored by antiandrogen treatment, suggesting that hyperactive androgen receptor signaling in Fabry mice may contribute to nNOS upregulation. However, the phosphodiesterase-5A expression level and the adenosine content in the penis, which are known to play roles in the development of priapism in other etiologies, were unchanged in Fabry mice. In conclusion, these data suggested that increased nNOS (and probably eNOS) content and the consequential elevated NO production and high arterial blood flow in the penis may be the underlying mechanism of priapism in Fabry mice. Furthermore, in combination with previous findings, this study suggested that regulation of NOS expression is susceptible to α-galactosidase A deficiency, and this may represent a general pathogenic mechanism of Fabry vasculopathy.


Subject(s)
Fabry Disease/complications , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type I/metabolism , Penile Erection , Penis/enzymology , Priapism/etiology , Animals , Disease Models, Animal , Enzyme Replacement Therapy/methods , Fabry Disease/enzymology , Fabry Disease/physiopathology , Fabry Disease/therapy , Genetic Therapy/methods , Male , Mice, 129 Strain , Mice, Inbred C57BL , Nitric Oxide/metabolism , Penis/physiopathology , Priapism/enzymology , Priapism/physiopathology , Priapism/therapy , Regional Blood Flow , Signal Transduction , Up-Regulation , alpha-Galactosidase/biosynthesis , alpha-Galactosidase/genetics
10.
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
11.
Development ; 144(6): 1025-1034, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28087635

ABSTRACT

The tumor suppressor retinoblastoma protein (RB) regulates S-phase cell cycle entry via E2F transcription factors. Knockout (KO) mice have shown that RB plays roles in cell migration, differentiation and apoptosis, in developing and adult brain. In addition, the RB family is required for self-renewal and survival of human embryonic stem cells (hESCs). Since little is known about the role of RB in human brain development, we investigated its function in cerebral organoids differentiated from gene-edited hESCs lacking RB. We show that RB is abundantly expressed in neural stem and progenitor cells in organoids at 15 and 28 days of culture. RB loss promoted S-phase entry in DCX+ cells and increased apoptosis in Sox2+ neural stem and progenitor cells, and in DCX+ and Tuj1+ neurons. Associated with these cell cycle and pro-apoptotic effects, we observed increased CCNA2 and BAX gene expression, respectively. Moreover, we observed aberrant Tuj1+ neuronal migration in RB-KO organoids and upregulation of the gene encoding VLDLR, a receptor important in reelin signaling. Corroborating the results in RB-KO organoids in vitro, we observed ectopically localized Tuj1+ cells in RB-KO teratomas grown in vivo Taken together, these results identify crucial functions for RB in the cerebral organoid model of human brain development.


Subject(s)
Cell Movement , Cerebrum/cytology , Neurons/cytology , Organoids/cytology , Organoids/metabolism , Retinoblastoma Protein/metabolism , Cell Death , Cell Line , Cell Proliferation , Cell Survival , Doublecortin Protein , Embryonic Stem Cells/cytology , Gene Deletion , Gene Knockout Techniques , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/metabolism , Reelin Protein , S Phase
12.
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
13.
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
14.
ACS Chem Biol ; 11(4): 1128-36, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26828310

ABSTRACT

Novel strategies are needed to modulate ß-cell differentiation and function as potential ß-cell replacement or restorative therapies for diabetes. We previously demonstrated that small molecules based on the isoxazole scaffold drive neuroendocrine phenotypes. The nature of the effects of isoxazole compounds on ß-cells was incompletely defined. We find that isoxazole induces genes that support neuroendocrine and ß-cell phenotypes and suppresses genes important for proliferation. Isoxazole alters ß-cell metabolites and protects glucose-responsive signaling pathways under lipotoxic conditions. Finally, we show that isoxazole improves glycemia in a mouse model of ß-cell regeneration. Isoxazole is a prime candidate to alter cell fate in different contexts.


Subject(s)
Cell Proliferation/drug effects , Gene Expression/drug effects , Islets of Langerhans/drug effects , Isoxazoles/pharmacology , Humans , Islets of Langerhans/cytology
15.
J Investig Med ; 64(1): 50-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26755814

ABSTRACT

Adipose-derived stem cells (ADSCs) have myocardial regeneration potential, and transplantation of these cells following myocardial infarction (MI) in animal models leads to modest improvements in cardiac function. We hypothesized that pharmacological priming of pre-transplanted ADSCs would further improve left ventricular functional recovery after MI. We previously identified a compound from a family of 3,5-disubstituted isoxazoles, ISX1, capable of activating an Nkx2-5-driven promoter construct. Here, using ADSCs, we found that ISX1 (20 mM, 4 days) triggered a robust, dose-dependent, fourfold increase in Nkx2-5 expression, an early marker of cardiac myocyte differentiation and increased ADSC viability in vitro. Co-culturing neonatal cardiomyocytes with ISX1-treated ADSCs increased early and late cardiac gene expression. Whereas ISX1 promoted ADSC differentiation toward a cardiogenic lineage, it did not elicit their complete differentiation or their differentiation into mature adipocytes, osteoblasts, or chondrocytes, suggesting that re-programming is cardiomyocyte specific. Cardiac transplantation of ADSCs improved left ventricular functional recovery following MI, a response which was significantly augmented by transplantation of ISX1- pretreated cells. Moreover, ISX1-treated and transplanted ADSCs engrafted and were detectable in the myocardium 3 weeks following MI, albeit at relatively small numbers. ISX1 treatment increased histone acetyltransferase (HAT) activity in ADSCs, which was associated with histone 3 and histone 4 acetylation. Finally, hearts transplanted with ISX1-treated ADSCs manifested significant increases in neovascularization, which may account for the improved cardiac function. These findings suggest that a strategy of drug-facilitated initiation of myocyte differentiation enhances exogenously transplanted ADSC persistence in vivo, and consequent tissue neovascularization, to improve cardiac function.


Subject(s)
Adipose Tissue/cytology , Myocardium/pathology , Stem Cell Transplantation , Stem Cells/cytology , Wound Healing , Acetylation/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Chondrogenesis/drug effects , Chondrogenesis/genetics , Chromatin Assembly and Disassembly/drug effects , Coculture Techniques , Female , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Homeobox Protein Nkx-2.5/metabolism , Isoxazoles/pharmacology , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Neovascularization, Physiologic/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , Stem Cells/drug effects , Stem Cells/metabolism , Wound Healing/drug effects
16.
Hum Mol Genet ; 24(11): 3181-91, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25701874

ABSTRACT

Fabry disease is caused by deficient activity of lysosomal enzyme α-galactosidase A. The enzyme deficiency results in intracellular accumulation of glycosphingolipids, leading to a variety of clinical manifestations including hypertrophic cardiomyopathy and renal insufficiency. The mechanism through which glycosphingolipid accumulation causes these manifestations remains unclear. Current treatment, especially when initiated at later stage of the disease, does not produce completely satisfactory results. Elucidation of the pathogenesis of Fabry disease is therefore crucial to developing new treatments. We found increased activity of androgen receptor (AR) signaling in Fabry disease. We subsequently also found that blockade of AR signaling either through castration or AR-antagonist prevented and reversed cardiac and kidney hypertrophic phenotype in a mouse model of Fabry disease. Our findings implicate abnormal AR pathway in the pathogenesis of Fabry disease and suggest blocking AR signaling as a novel therapeutic approach.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Fabry Disease/metabolism , Hypertrophy, Left Ventricular/metabolism , Kidney Diseases/metabolism , Receptors, Androgen/metabolism , Animals , Fabry Disease/drug therapy , Female , Hypertrophy, Left Ventricular/drug therapy , Kidney/metabolism , Kidney/pathology , Kidney Diseases/drug therapy , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Androgen/genetics , Signal Transduction , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 111(1): E129-38, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24367119

ABSTRACT

Mammalian skeletal muscle can remodel, repair, and regenerate itself by mobilizing satellite cells, a resident population of myogenic progenitor cells. Muscle injury and subsequent activation of myogenic progenitor cells is associated with oxidative stress. Cytoglobin is a hemoprotein expressed in response to oxidative stress in a variety of tissues, including striated muscle. In this study, we demonstrate that cytoglobin is up-regulated in activated myogenic progenitor cells, where it localizes to the nucleus and contributes to cell viability. siRNA-mediated depletion of cytoglobin from C2C12 myoblasts increased levels of reactive oxygen species and apoptotic cell death both at baseline and in response to stress stimuli. Conversely, overexpression of cytoglobin reduced reactive oxygen species levels, caspase activity, and cell death. Mice in which cytoglobin was knocked out specifically in skeletal muscle were generated to examine the role of cytoglobin in vivo. Myogenic progenitor cells isolated from these mice were severely deficient in their ability to form myotubes as compared with myogenic progenitor cells from wild-type littermates. Consistent with this finding, the capacity for muscle regeneration was severely impaired in mice deficient for skeletal-muscle cytoglobin. Collectively, these data demonstrate that cytoglobin serves an important role in muscle repair and regeneration.


Subject(s)
Gene Expression Regulation , Globins/metabolism , Muscles/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Animals , Apoptosis , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Cytoglobin , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/metabolism , Reactive Oxygen Species , Satellite Cells, Skeletal Muscle/pathology , Stem Cells/cytology , Time Factors
18.
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
20.
ACS Chem Neurosci ; 3(7): 557-68, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22860225

ABSTRACT

Acidosis, a critical aspect of central nervous system (CNS) pathophysiology and a metabolic corollary of the hypoxic stem cell niche, could be an expedient trigger for hippocampal neurogenesis and brain repair. We recently tracked the function of our isoxazole stem cell-modulator small molecules (Isx) through a chemical biology-target discovery strategy to GPR68, a proton (pH) sensing G protein-coupled receptor with no known function in brain. Isx and GPR68 coregulated neuronal target genes such as Bex1 (brain-enriched X-linked protein-1) in hippocampal neural progenitors (HCN cells), which further amplified GPR68 signaling by producing metabolic acid in response to Isx. To evaluate this proneurogenic small molecule/proton signaling circuit in vivo, we explored GPR68 and BEX1 expression in brain and probed brain function with Isx. We localized proton-sensing GPR68 to radial processes of hippocampal type 1 neural stem cells (NSCs) and, conversely, localized BEX1 to neurons. At the transcriptome level, Isx demonstrated unrivaled proneurogenic activity in primary hippocampal NSC cultures. In vivo, Isx pharmacologically targeted type 1 NSCs, promoting neurogenesis in young mice, depleting the progenitor pool without adversely affecting hippocampal learning and memory function. After traumatic brain injury, cerebral cortical astrocytes abundantly expressed GPR68, suggesting an additional role for proton-GPCR signaling in reactive astrogliosis. Thus, probing a novel proneurogenic synthetic small molecule's mechanism-of-action, candidate target, and pharmacological activity, we identified a new GPR68 regulatory pathway for integrating neural stem and astroglial cell functions with brain pH.


Subject(s)
Brain/physiology , Hippocampus/physiology , Neurogenesis/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Cells, Cultured , Hippocampus/cytology , Hydrogen-Ion Concentration , Maze Learning/physiology , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Receptors, G-Protein-Coupled/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...