Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Circulation ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39185559

ABSTRACT

BACKGROUND: The Hippo pathway effector YAP (Yes-associated protein) plays an essential role in cardiomyocyte proliferation and heart regeneration. In response to physiological changes, YAP moves in and out of the nucleus. The pathophysiological mechanisms regulating YAP subcellular localization after myocardial infarction remain poorly defined. METHODS: We identified YAP acetylation at site K265 by in vitro acetylation followed by mass spectrometry analysis. We used adeno-associated virus to express YAP-containing mutations that either abolished acetylation (YAP-K265R) or mimicked acetylation (YAP-K265Q) and studied how acetylation regulates YAP subcellular localization in mouse hearts. We generated a cell line with YAP-K265R mutation and investigated the protein-protein interactors by YAP immunoprecipitation followed by mass spectrometry, then validated the YAP interaction in neonatal rat ventricular myocytes. We examined colocalization of YAP and TUBA4A (tubulin α 4A) by superresolution imaging. Furthermore, we developed YAP-K265R and αMHC-MerCreMer (MCM); Yap-loxP/K265R mutant mice to examine the pathophysiological role of YAP acetylation in cardiomyocytes during cardiac regeneration. RESULTS: We found that YAP is acetylated at K265 by CBP (CREB-binding protein)/P300 (E1A-binding protein P300) and is deacetylated by nicotinamide phosphoribosyltransferase/nicotinamide adenine dinucleotide/sirtuins axis in cardiomyocytes. After myocardial infarction, YAP acetylation is increased, which promotes YAP cytoplasmic localization. Compared with controls, mice that were genetically engineered to express a K265R mutation that prevents YAP K256 acetylation showed improved cardiac regenerative ability and increased YAP nuclear localization. Mechanistically, YAP acetylation facilitates its interaction with TUBA4A, a component of the microtubule network that sequesters acetylated YAP in the cytoplasm. After myocardial infarction, the microtubule network increased in cardiomyocytes, resulting in the accumulation of YAP in the cytoplasm. CONCLUSIONS: After myocardial infarction, decreased sirtuin activity enriches YAP acetylation at K265. The growing TUBA4A network sequesters acetylated YAP within the cytoplasm, which is detrimental to cardiac regeneration.

2.
Nat Cardiovasc Res ; 3(3): 283-300, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38510108

ABSTRACT

After myocardial infarction (MI), mammalian hearts do not regenerate, and the microenvironment is disrupted. Hippo signaling loss of function with activation of transcriptional co-factor YAP induces heart renewal and rebuilds the post-MI microenvironment. In this study, we investigated adult renewal-competent mouse hearts expressing an active version of YAP, called YAP5SA, in cardiomyocytes (CMs). Spatial transcriptomics and single-cell RNA sequencing revealed a conserved, renewal-competent CM cell state called adult (a)CM2 with high YAP activity. aCM2 co-localized with cardiac fibroblasts (CFs) expressing complement pathway component C3 and macrophages (MPs) expressing C3ar1 receptor to form a cellular triad in YAP5SA hearts and renewal-competent neonatal hearts. Although aCM2 was detected in adult mouse and human hearts, the cellular triad failed to co-localize in these non-renewing hearts. C3 and C3ar1 loss-of-function experiments indicated that C3a signaling between MPs and CFs was required to assemble the pro-renewal aCM2, C3+ CF and C3ar1+ MP cellular triad.

3.
Life Sci Alliance ; 7(2)2024 02.
Article in English | MEDLINE | ID: mdl-38012001

ABSTRACT

Modulation of the heart's immune microenvironment is crucial for recovery after ischemic events such as myocardial infarction (MI). Endothelial cells (ECs) can have immune regulatory functions; however, interactions between ECs and the immune environment in the heart after MI remain poorly understood. We identified an EC-specific IFN responsive and immune regulatory gene signature in adult and pediatric heart failure (HF) tissues. Single-cell transcriptomic analysis of murine hearts subjected to MI uncovered an EC population (IFN-ECs) with immunologic gene signatures similar to those in human HF. IFN-ECs were enriched in regenerative-stage mouse hearts and expressed genes encoding immune responsive transcription factors (Irf7, Batf2, and Stat1). Single-cell chromatin accessibility studies revealed an enrichment of these TF motifs at IFN-EC signature genes. Expression of immune regulatory ligand genes by IFN-ECs suggests bidirectional signaling between IFN-ECs and macrophages in regenerative-stage hearts. Our data suggest that ECs may adopt immune regulatory signatures after cardiac injury to accompany the reparative response. The presence of these signatures in human HF and murine MI models suggests a potential role for EC-mediated immune regulation in responding to stress induced by acute injury in MI and chronic adverse remodeling in HF.


Subject(s)
Heart Failure , Myocardial Infarction , Mice , Humans , Animals , Child , Endothelial Cells/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Heart , Signal Transduction/genetics
4.
Nature ; 608(7921): 181-191, 2022 08.
Article in English | MEDLINE | ID: mdl-35732239

ABSTRACT

The heart, the first organ to develop in the embryo, undergoes complex morphogenesis that when defective results in congenital heart disease (CHD). With current therapies, more than 90% of patients with CHD survive into adulthood, but many suffer premature death from heart failure and non-cardiac causes1. Here, to gain insight into this disease progression, we performed single-nucleus RNA sequencing on 157,273 nuclei from control hearts and hearts from patients with CHD, including those with hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot, two common forms of cyanotic CHD lesions, as well as dilated and hypertrophic cardiomyopathies. We observed CHD-specific cell states in cardiomyocytes, which showed evidence of insulin resistance and increased expression of genes associated with FOXO signalling and CRIM1. Cardiac fibroblasts in HLHS were enriched in a low-Hippo and high-YAP cell state characteristic of activated cardiac fibroblasts. Imaging mass cytometry uncovered a spatially resolved perivascular microenvironment consistent with an immunodeficient state in CHD. Peripheral immune cell profiling suggested deficient monocytic immunity in CHD, in agreement with the predilection in CHD to infection and cancer2. Our comprehensive phenotyping of CHD provides a roadmap towards future personalized treatments for CHD.


Subject(s)
Heart Defects, Congenital , Phenotype , Bone Morphogenetic Protein Receptors/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/immunology , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Disease Progression , Fibroblasts/metabolism , Fibroblasts/pathology , Forkhead Transcription Factors/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/immunology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Hypoplastic Left Heart Syndrome/genetics , Hypoplastic Left Heart Syndrome/immunology , Hypoplastic Left Heart Syndrome/metabolism , Hypoplastic Left Heart Syndrome/pathology , Image Cytometry , Insulin Resistance , Monocytes/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Tetralogy of Fallot/genetics , Tetralogy of Fallot/immunology , Tetralogy of Fallot/metabolism , Tetralogy of Fallot/pathology , YAP-Signaling Proteins/metabolism
5.
Nat Cell Biol ; 22(11): 1346-1356, 2020 11.
Article in English | MEDLINE | ID: mdl-33046882

ABSTRACT

Cardiomyocyte loss after injury results in adverse remodelling and fibrosis, inevitably leading to heart failure. The ERBB2-Neuregulin and Hippo-YAP signalling pathways are key mediators of heart regeneration, yet the crosstalk between them is unclear. We demonstrate that transient overexpression of activated ERBB2 in cardiomyocytes (OE CMs) promotes cardiac regeneration in a heart failure model. OE CMs present an epithelial-mesenchymal transition (EMT)-like regenerative response manifested by cytoskeletal remodelling, junction dissolution, migration and extracellular matrix turnover. We identified YAP as a critical mediator of ERBB2 signalling. In OE CMs, YAP interacts with nuclear-envelope and cytoskeletal components, reflecting an altered mechanical state elicited by ERBB2. We identified two YAP-activating phosphorylations on S352 and S274 in OE CMs, which peak during metaphase, that are ERK dependent and Hippo independent. Viral overexpression of YAP phospho-mutants dampened the proliferative competence of OE CMs. Together, we reveal a potent ERBB2-mediated YAP mechanotransduction signalling, involving EMT-like characteristics, resulting in robust heart regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Heart Failure/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Receptor, ErbB-2/metabolism , Regeneration , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Mechanotransduction, Cellular , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Phosphorylation , Receptor, ErbB-2/genetics , YAP-Signaling Proteins
6.
Hypertens Res ; 43(11): 1204-1213, 2020 11.
Article in English | MEDLINE | ID: mdl-32616846

ABSTRACT

Esaxerenone is a novel selective mineralocorticoid receptor (MR) blocker that was recently approved in Japan to treat hypertension. In phase II and III studies, esaxerenone plus a renin-angiotensin system inhibitor markedly reduced the urinary albumin-to-creatinine ratio (UACR) in hypertensive patients with diabetic nephropathy. To evaluate a direct renoprotective effect by MR blockade independent of an antihypertensive effect in the context of diabetic nephropathy, esaxerenone (3 mg/kg), olmesartan (an angiotensin II receptor blocker; 1 mg/kg), or both were orally administered to KK-Ay mice, a type 2 diabetes model, once daily for 56 days. Urinary albumin (Ualb), UACR, and markers, such as podocalyxin, monocyte chemoattractant protein-1 (MCP-1), and 8-hydroxy-2'-deoxyguanosine (8-OHdG), were measured, along with systolic blood pressure (SBP), fasting blood glucose, and serum K+ levels. Prior to the initiation of drug administration, KK-Ay mice showed higher blood glucose, insulin, Ualb excretion, and UACR levels than C57BL/6 J mice, a nondiabetic control, indicating the development of diabetic renal injury. Combined treatment with esaxerenone and olmesartan significantly reduced the change in UACR from baseline compared with the change associated with vehicle at week 8 (-1.750 vs. 0.339 g/gCre; P < 0.002) and significantly inhibited the change in Ualb from baseline compared with the change associated with vehicle at week 8 (P < 0.002). The combination treatment also reduced urinary excretion of podocalyxin and MCP-1, but did not influence 8-OHdG excretion, SBP, blood glucose, or serum K+ levels. Overall, esaxerenone plus olmesartan treatment ameliorated diabetic nephropathy in KK-Ay mice without affecting SBP, suggesting that the renoprotective effects of esaxerenone could be exerted independently of its antihypertensive effect.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Diabetic Nephropathies/drug therapy , Imidazoles/therapeutic use , Mineralocorticoid Receptor Antagonists/therapeutic use , Pyrroles/therapeutic use , Sulfones/therapeutic use , Tetrazoles/therapeutic use , Albuminuria/drug therapy , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Type 2/complications , Drug Evaluation, Preclinical , Drug Therapy, Combination , Imidazoles/pharmacology , Male , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/pharmacology , Pyrroles/pharmacology , Sulfones/pharmacology , Tetrazoles/pharmacology
7.
Dev Cell ; 48(6): 765-779.e7, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30773489

ABSTRACT

Specialized adult somatic cells, such as cardiomyocytes (CMs), are highly differentiated with poor renewal capacity, an integral reason underlying organ failure in disease and aging. Among the least renewable cells in the human body, CMs renew approximately 1% annually. Consistent with poor CM turnover, heart failure is the leading cause of death. Here, we show that an active version of the Hippo pathway effector YAP, termed YAP5SA, partially reprograms adult mouse CMs to a more fetal and proliferative state. One week after induction, 19% of CMs that enter S-phase do so twice, CM number increases by 40%, and YAP5SA lineage CMs couple to pre-existing CMs. Genomic studies showed that YAP5SA increases chromatin accessibility and expression of fetal genes, partially reprogramming long-lived somatic cells in vivo to a primitive, fetal-like, and proliferative state.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Aging/physiology , Chromatin/metabolism , Heart/growth & development , Organogenesis , Phosphoproteins/metabolism , Action Potentials , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Cycle , Cell Cycle Proteins , Cell Lineage , Cell Proliferation , Diploidy , Enhancer Elements, Genetic/genetics , Gain of Function Mutation/genetics , Gene Expression Regulation, Developmental , Heart Ventricles/anatomy & histology , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Promoter Regions, Genetic/genetics , Transcription Factor AP-1/metabolism , Transgenes , YAP-Signaling Proteins
8.
Development ; 145(18)2018 09 26.
Article in English | MEDLINE | ID: mdl-30143541

ABSTRACT

Loss of the paired-like homeodomain transcription factor 2 (Pitx2) in cardiomyocytes predisposes mice to atrial fibrillation and compromises neonatal regenerative capacity. In addition, Pitx2 gain-of-function protects mature cardiomyocytes from ischemic injury and promotes heart repair. Here, we characterized the long-term myocardial phenotype following myocardial infarction (MI) in Pitx2 conditional-knockout (Pitx2 CKO) mice. We found adipose-like tissue in Pitx2 CKO hearts 60 days after MI induced surgically at postnatal day 2 but not at day 8. Molecular and cellular analyses showed the onset of adipogenic signaling in mutant hearts after MI. Lineage tracing experiments showed a non-cardiomyocyte origin of the de novo adipose-like tissue. Interestingly, we found that Pitx2 promotes mitochondrial function through its gene regulatory network, and that the knockdown of a key mitochondrial Pitx2 target gene, Cox7c, also leads to the accumulation of myocardial fat tissue. Single-nuclei RNA-seq revealed that Pitx2-deficient hearts were oxidatively stressed. Our findings reveal a role for Pitx2 in maintaining proper cardiac cellular composition during heart regeneration via the maintenance of proper mitochondrial structure and function.


Subject(s)
Adipogenesis/physiology , Homeodomain Proteins/metabolism , Mitochondria/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Regeneration/physiology , Transcription Factors/metabolism , Adipose Tissue/pathology , Animals , Cell Line , Electron Transport Complex IV/genetics , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Myocardial Infarction/genetics , Oxidative Stress/genetics , Regeneration/genetics , Transcription Factors/genetics , Homeobox Protein PITX2
9.
Sci Rep ; 8(1): 9955, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29967338

ABSTRACT

Vasodilator-stimulated phosphoprotein (VASP) is a member of actin regulatory proteins implicated in platelet adhesion. In addition, phosphorylation of VASP is utilised for the assessment of platelet reactivity in patients treated with P2Y12 receptor antagonists, a class of antiplatelet agents. However, the role of VASP in platelet aggregation, thrombogenesis, haemostasis, and the antiplatelet effect of P2Y12 receptor antagonists remains unclear. We investigated these effects using heterozygous and homozygous VASP knockout rats generated with a CRISPR/Cas9 system. Baseline characteristics, such as haematology and other biochemical parameters, were comparable among the genotypes. In vitro platelet aggregation stimulated by adenosine diphosphate (ADP) or collagen, P-selectin expression of rat platelets treated with ADP, and in vivo thrombocytopenia induced by collagen were also comparable among the genotypes. In addition, in vivo thrombogenesis in a ferric chloride-induced arterial thrombosis model and bleeding time were also comparable among the genotypes. Furthermore, the in vitro antiplatelet effect of prasugrel, a third-generation P2Y12 receptor antagonist, was unaffected by VASP knockout. Although phosphorylated VASP is still an important surrogate marker specific for P2Y12 antagonists, our findings demonstrate that VASP is not a major mediator of platelet aggregation, thrombogenesis, haemostasis, and the antiplatelet effect of prasugrel in rats.


Subject(s)
Cell Adhesion Molecules/metabolism , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Platelet Aggregation Inhibitors/pharmacology , Prasugrel Hydrochloride/pharmacology , Thrombosis/genetics , Animals , Cell Adhesion Molecules/genetics , Collagen/toxicity , Female , Hemostasis/drug effects , Hemostasis/physiology , Microfilament Proteins/genetics , P-Selectin/metabolism , Phosphoproteins/genetics , Phosphorylation , Piperazines/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Rats, Mutant Strains , Rats, Sprague-Dawley , Thrombocytopenia/chemically induced , Thrombocytopenia/genetics
10.
Dev Cell ; 45(2): 153-169.e6, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29689192

ABSTRACT

During development, progenitors progress through transition states. The cardiac epicardium contains progenitors of essential non-cardiomyocytes. The Hippo pathway, a kinase cascade that inhibits the Yap transcriptional co-factor, controls organ size in developing hearts. Here, we investigated Hippo kinases Lats1 and Lats2 in epicardial diversification. Epicardial-specific deletion of Lats1/2 was embryonic lethal, and mutant embryos had defective coronary vasculature remodeling. Single-cell RNA sequencing revealed that Lats1/2 mutant cells failed to activate fibroblast differentiation but remained in an intermediate cell state with both epicardial and fibroblast characteristics. Lats1/2 mutant cells displayed an arrested developmental trajectory with persistence of epicardial markers and expanded expression of Yap targets Dhrs3, an inhibitor of retinoic acid synthesis, and Dpp4, a protease that modulates extracellular matrix (ECM) composition. Genetic and pharmacologic manipulation revealed that Yap inhibits fibroblast differentiation, prolonging a subepicardial-like cell state, and promotes expression of matricellular factors, such as Dpp4, that define ECM characteristics.


Subject(s)
Fibroblasts/cytology , Heart/embryology , Organogenesis/physiology , Pericardium/cytology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Extracellular Matrix , Female , Fibroblasts/metabolism , Gene Expression Profiling , Heart/physiology , Hippo Signaling Pathway , Mice , Mice, Knockout , Pericardium/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Single-Cell Analysis , YAP-Signaling Proteins
11.
Nature ; 550(7675): 260-264, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28976966

ABSTRACT

Mammalian organs vary widely in regenerative capacity. Poorly regenerative organs, such as the heart are particularly vulnerable to organ failure. Once established, heart failure commonly results in mortality. The Hippo pathway, a kinase cascade that prevents adult cardiomyocyte proliferation and regeneration, is upregulated in human heart failure. Here we show that deletion of the Hippo pathway component Salvador (Salv) in mouse hearts with established ischaemic heart failure after myocardial infarction induces a reparative genetic program with increased scar border vascularity, reduced fibrosis, and recovery of pumping function compared with controls. Using translating ribosomal affinity purification, we isolate cardiomyocyte-specific translating messenger RNA. Hippo-deficient cardiomyocytes have increased expression of proliferative genes and stress response genes, such as the mitochondrial quality control gene, Park2. Genetic studies indicate that Park2 is essential for heart repair, suggesting a requirement for mitochondrial quality control in regenerating myocardium. Gene therapy with a virus encoding Salv short hairpin RNA improves heart function when delivered at the time of infarct or after ischaemic heart failure following myocardial infarction was established. Our findings indicate that the failing heart has a previously unrecognized reparative capacity involving more than cardiomyocyte renewal.


Subject(s)
Cell Cycle Proteins/deficiency , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/therapy , Myocardial Infarction/complications , Protein Serine-Threonine Kinases/deficiency , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Genetic Therapy , Heart Failure, Systolic/etiology , Heart Failure, Systolic/pathology , Hippo Signaling Pathway , Humans , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Quality Control , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Ubiquitin-Protein Ligases/genetics
12.
Nature ; 547(7662): 227-231, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28581498

ABSTRACT

The regenerative capacity of the adult mammalian heart is limited, because of the reduced ability of cardiomyocytes to progress through mitosis. Endogenous cardiomyocytes have regenerative capacity at birth but this capacity is lost postnatally, with subsequent organ growth occurring through cardiomyocyte hypertrophy. The Hippo pathway, a conserved kinase cascade, inhibits cardiomyocyte proliferation in the developing heart to control heart size and prevents regeneration in the adult heart. The dystrophin-glycoprotein complex (DGC), a multicomponent transmembrane complex linking the actin cytoskeleton to extracellular matrix, is essential for cardiomyocyte homeostasis. DGC deficiency in humans results in muscular dystrophy, including the lethal Duchenne muscular dystrophy. Here we show that the DGC component dystroglycan 1 (Dag1) directly binds to the Hippo pathway effector Yap to inhibit cardiomyocyte proliferation in mice. The Yap-Dag1 interaction was enhanced by Hippo-induced Yap phosphorylation, revealing a connection between Hippo pathway function and the DGC. After injury, Hippo-deficient postnatal mouse hearts maintained organ size control by repairing the defect with correct dimensions, whereas postnatal hearts deficient in both Hippo and the DGC showed cardiomyocyte overproliferation at the injury site. In the hearts of mature Mdx mice (which have a point mutation in Dmd)-a model of Duchenne muscular dystrophy-Hippo deficiency protected against overload-induced heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dystrophin/metabolism , Glycoproteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Myocytes, Cardiac/cytology , Phosphoproteins/metabolism , Animals , Cardiomyopathies , Cell Cycle Proteins , Cell Proliferation , Dystroglycans/metabolism , Dystrophin/deficiency , Dystrophin/genetics , Glycoproteins/deficiency , Heart Failure/genetics , Heart Failure/prevention & control , Hippo Signaling Pathway , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Multiprotein Complexes/deficiency , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/metabolism , Organ Size , Phosphorylation , Pressure , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , YAP-Signaling Proteins
13.
J Pharmacol Exp Ther ; 358(3): 548-57, 2016 09.
Article in English | MEDLINE | ID: mdl-27384074

ABSTRACT

The present study was designed to assess both preventive and therapeutic effects of (S)-1-(2-Hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl) phenyl]-5-[2-(trifluoromethyl) phenyl]-1H-pyrrole-3-carboxamide (CS-3150), a novel nonsteroidal mineralocorticoid receptor antagonist, on renal injury in deoxycorticosterone acetate (DOCA)/salt-induced hypertensive rats (DOCA rats). From 7 weeks of age, DOCA was subcutaneously administered once a week for 4 weeks to uninephrectomized rats fed a high-salt diet. In experiment 1, CS-3150 (0.3-3 mg/kg) was orally administered once a day for 4 weeks coincident with DOCA administration. In experiment 2, after establishment of renal injury by 4 weeks of DOCA/salt loading, CS-3150 (3 mg/kg) was orally administered once a day for 4 weeks with or without continuous DOCA administration. In experiment 1, DOCA/salt loading significantly increased systolic blood pressure (SBP), which was prevented by CS-3150 in a dose-dependent manner. Development of renal injury (proteinuria, renal hypertrophy, and histopathological changes in glomeruli and tubule) was also suppressed by CS-3150 with inhibition of mRNA expression of fibrosis, inflammation, and oxidative stress markers. In experiment 2, under continuous DOCA treatment, CS-3150 clearly ameliorated existing renal injury without lowering SBP, indicating that CS-3150 regressed renal injury independent of its antihypertensive action. Moreover, CS-3150 treatment in combination with withdrawal of DOCA showed further therapeutic effect on renal injury accompanied by reduction in SBP. These results demonstrate that CS-3150 not only prevents but also ameliorates hypertension and renal injury in DOCA rats. Therefore, CS-3150 could be a promising agent for the treatment of hypertension and renal disorders, and may have potential to promote regression of renal injury.


Subject(s)
Desoxycorticosterone Acetate/adverse effects , Hypertension/chemically induced , Hypertension/prevention & control , Kidney/drug effects , Pyrroles/pharmacology , Receptors, Mineralocorticoid/metabolism , Sodium Chloride, Dietary/adverse effects , Sulfones/pharmacology , Animals , Blood Pressure/drug effects , Gene Expression Regulation/drug effects , Hypertension/metabolism , Hypertension/pathology , Kidney/injuries , Kidney/metabolism , Kidney/pathology , Male , Mineralocorticoid Receptor Antagonists/pharmacology , Organ Size/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
14.
Nature ; 534(7605): 119-23, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27251288

ABSTRACT

Myocardial infarction results in compromised myocardial function and heart failure owing to insufficient cardiomyocyte self-renewal. Unlike many vertebrates, mammalian hearts have only a transient neonatal renewal capacity. Reactivating primitive reparative ability in the mature mammalian heart requires knowledge of the mechanisms that promote early heart repair. By testing an established Hippo-deficient heart regeneration mouse model for factors that promote renewal, here we show that the expression of Pitx2 is induced in injured, Hippo-deficient ventricles. Pitx2-deficient neonatal mouse hearts failed to repair after apex resection, whereas adult mouse cardiomyocytes with Pitx2 gain-of-function efficiently regenerated after myocardial infarction. Genomic analyses indicated that Pitx2 activated genes encoding electron transport chain components and reactive oxygen species scavengers. A subset of Pitx2 target genes was cooperatively regulated with the Hippo pathway effector Yap. Furthermore, Nrf2, a regulator of the antioxidant response, directly regulated the expression and subcellular localization of Pitx2. Pitx2 mutant myocardium had increased levels of reactive oxygen species, while antioxidant supplementation suppressed the Pitx2 loss-of-function phenotype. These findings reveal a genetic pathway activated by tissue damage that is essential for cardiac repair.


Subject(s)
Antioxidants/metabolism , Heart Injuries/metabolism , Homeodomain Proteins/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Regeneration/physiology , Transcription Factors/metabolism , Wound Healing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Antioxidants/pharmacology , Cell Cycle Proteins , Disease Models, Animal , Electron Transport/drug effects , Electron Transport/genetics , Female , Free Radical Scavengers/metabolism , Heart Injuries/genetics , Heart Injuries/pathology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hippo Signaling Pathway , Homeodomain Proteins/genetics , Male , Mice , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NF-E2-Related Factor 2/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Reactive Oxygen Species/metabolism , Regeneration/drug effects , Regeneration/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Wound Healing/drug effects , Wound Healing/genetics , YAP-Signaling Proteins , Homeobox Protein PITX2
15.
Eur J Pharmacol ; 761: 226-34, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26073023

ABSTRACT

The present study was designed to characterize the pharmacological profile of CS-3150, a novel non-steroidal mineralocorticoid receptor antagonist. In the radioligand-binding assay, CS-3150 inhibited (3)H-aldosterone binding to mineralocorticoid receptor with an IC50 value of 9.4nM, and its potency was superior to that of spironolactone and eplerenone, whose IC50s were 36 and 713nM, respectively. CS-3150 also showed at least 1000-fold higher selectivity for mineralocorticoid receptor over other steroid hormone receptors, glucocorticoid receptor, androgen receptor and progesterone receptor. In the reporter gene assay, CS-3150 inhibited aldosterone-induced transcriptional activation of human mineralocorticoid receptor with an IC50 value of 3.7nM, and its potency was superior to that of spironolactone and eplerenone, whose IC50s were 66 and 970nM, respectively. CS-3150 had no agonistic effect on mineralocorticoid receptor and did not show any antagonistic or agonistic effect on glucocorticoid receptor, androgen receptor and progesterone receptor even at the high concentration of 5µM. In adrenalectomized rats, single oral administration of CS-3150 suppressed aldosterone-induced decrease in urinary Na(+)/K(+) ratio, an index of in vivo mineralocorticoid receptor activation, and this suppressive effect was more potent and longer-lasting than that of spironolactone and eplerenone. Chronic treatment with CS-3150 inhibited blood pressure elevation induced by deoxycorticosterone acetate (DOCA)/salt-loading to rats, and this antihypertensive effect was more potent than that of spironolactone and eplerenone. These findings indicate that CS-3150 is a selective and highly potent mineralocorticoid receptor antagonist with long-lasting oral activity. This agent could be useful for the treatment of hypertension, cardiovascular and renal disorders.


Subject(s)
Mineralocorticoid Receptor Antagonists/pharmacology , Pyrroles/pharmacology , Receptors, Mineralocorticoid/drug effects , Sulfones/pharmacology , Administration, Oral , Adrenalectomy , Aldosterone/metabolism , Aldosterone/pharmacology , Animals , Antihypertensive Agents/pharmacology , Binding, Competitive , Blood Pressure/drug effects , Desoxycorticosterone Acetate , Disease Models, Animal , Dose-Response Relationship, Drug , Eplerenone , Female , HEK293 Cells , Humans , Hypertension/chemically induced , Hypertension/physiopathology , Hypertension/prevention & control , Male , Mineralocorticoid Receptor Antagonists/administration & dosage , Mineralocorticoid Receptor Antagonists/pharmacokinetics , Potassium/urine , Protein Binding , Pyrroles/administration & dosage , Pyrroles/pharmacokinetics , Rabbits , Radioligand Assay , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Sodium/urine , Spironolactone/analogs & derivatives , Spironolactone/metabolism , Spironolactone/pharmacology , Sulfones/administration & dosage , Sulfones/pharmacokinetics , Transcriptional Activation/drug effects , Transfection , Urological Agents/pharmacology , Water-Electrolyte Balance/drug effects
16.
Sci Signal ; 8(375): ra41, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25943351

ABSTRACT

The mammalian heart regenerates poorly, and damage commonly leads to heart failure. Hippo signaling is an evolutionarily conserved kinase cascade that regulates organ size during development and prevents adult mammalian cardiomyocyte regeneration by inhibiting the transcriptional coactivator Yap, which also responds to mechanical signaling in cultured cells to promote cell proliferation. To identify Yap target genes that are activated during cardiomyocyte renewal and regeneration, we performed Yap chromatin immunoprecipitation sequencing (ChIP-Seq) and mRNA expression profiling in Hippo signaling-deficient mouse hearts. We found that Yap directly regulated genes encoding cell cycle progression proteins, as well as genes encoding proteins that promote F-actin polymerization and that link the actin cytoskeleton to the extracellular matrix. Included in the latter group were components of the dystrophin glycoprotein complex, a large molecular complex that, when defective, results in muscular dystrophy in humans. Cardiomyocytes near the scar tissue of injured Hippo signaling-deficient mouse hearts showed cellular protrusions suggestive of cytoskeletal remodeling. The hearts of mdx mutant mice, which lack functional dystrophin and are a model for muscular dystrophy, showed impaired regeneration and cytoskeleton remodeling, but normal cardiomyocyte proliferation, after injury. Our data showed that, in addition to genes encoding cell cycle progression proteins, Yap regulated genes that enhance cytoskeletal remodeling. Thus, blocking the Hippo pathway input to Yap may tip the balance so that Yap responds to mechanical changes associated with heart injury to promote repair.


Subject(s)
Actins/metabolism , Cell Proliferation/physiology , Cytoskeleton/metabolism , Heart/physiology , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , Actins/genetics , Animals , Cytoskeleton/genetics , Hippo Signaling Pathway , Humans , Mice , Mice, Inbred mdx , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology
17.
Behav Brain Res ; 274: 108-17, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25108246

ABSTRACT

Actions are usually made of several action steps gearing towards an overarching goal. During observation of such action episodes the overarching action goal becomes more and more clear and upcoming action steps can be predicted with increasing precision. To tap this process, the present fMRI study investigated the dynamic changes of neural activity during the observation of distinct action steps that cohere by an overarching goal. Our hypotheses specifically addressed the role of the inferior frontal gyrus (IFG), a region assumed to be a key hub for integration functions during action processing, as well as the role of regions involved in action perception (often referred to as action observation network or AON) that should benefit from the predictability of forthcoming action steps. Participants watched separate action steps that formed a coherent action goal or not (factor goal coherence) and were performed by a single actor or not (factor actor coherence). Independent of actor coherence, neural activity in IFG and occipitotemporal cortex decreased as a function of goal predictability during the unfolding of goal-coherent episodes. In addition, we identified a network (precuneus, dorsolateral prefrontal and orbitofrontal cortex, angular gyrus, and middle temporal gyrus) that showed increased activity for goal coherence. We conclude that IFG fosters the integration of action steps to build overarching goals. Identifying the unifying goal of an action episode allows anticipation, and thus efficient processing, of forthcoming action steps. To this end, past action steps of the action episode are buffered and recollected with recourse to episodic memory.


Subject(s)
Brain Mapping , Cerebral Cortex/blood supply , Cerebral Cortex/physiology , Goals , Adult , Female , Functional Laterality , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Oxygen/blood , Photic Stimulation , Predictive Value of Tests , Young Adult
18.
Biomed Opt Express ; 5(7): 1980-92, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25071943

ABSTRACT

We report on a quantitative optical elastographic method based on shear wave imaging optical coherence tomography (SWI-OCT) for biomechanical characterization of cardiac muscle through noncontact elasticity measurement. The SWI-OCT system employs a focused air-puff device for localized loading of the cardiac muscle and utilizes phase-sensitive OCT to monitor the induced tissue deformation. Phase information from the optical interferometry is used to reconstruct 2-D depth-resolved shear wave propagation inside the muscle tissue. Cross-correlation of the displacement profiles at various spatial locations in the propagation direction is applied to measure the group velocity of the shear waves, based on which the Young's modulus of tissue is quantified. The quantitative feature and measurement accuracy of this method is demonstrated from the experiments on tissue-mimicking phantoms with the verification using uniaxial compression test. The experiments are performed on ex vivo cardiac muscle tissue from mice with normal and genetically altered myocardium. Our results indicate this optical elastographic technique is useful as a noncontact tool to assist the cardiac muscle studies.

19.
Development ; 140(23): 4683-90, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24255096

ABSTRACT

Heart failure due to cardiomyocyte loss after ischemic heart disease is the leading cause of death in the United States in large part because heart muscle regenerates poorly. The endogenous mechanisms preventing mammalian cardiomyocyte regeneration are poorly understood. Hippo signaling, an ancient organ size control pathway, is a kinase cascade that inhibits developing cardiomyocyte proliferation but it has not been studied postnatally or in fully mature adult cardiomyocytes. Here, we investigated Hippo signaling in adult cardiomyocyte renewal and regeneration. We found that unstressed Hippo-deficient adult mouse cardiomyocytes re-enter the cell cycle and undergo cytokinesis. Moreover, Hippo deficiency enhances cardiomyocyte regeneration with functional recovery after adult myocardial infarction as well as after postnatal day eight (P8) cardiac apex resection and P8 myocardial infarction. In damaged hearts, Hippo mutant cardiomyocytes also have elevated proliferation. Our findings reveal that Hippo signaling is an endogenous repressor of adult cardiomyocyte renewal and regeneration. Targeting the Hippo pathway in human disease might be beneficial for the treatment of heart disease.


Subject(s)
Heart/physiology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , Animals , Cell Cycle , Cell Line , Cell Proliferation , Hippo Signaling Pathway , Mice , Mice, Transgenic , Myocardial Infarction , Myocytes, Cardiac/cytology , RNA Interference , RNA, Small Interfering , Signal Transduction
20.
Nat Cell Biol ; 15(11): 1282-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24161931

ABSTRACT

Although aberrant reactivation of embryonic gene programs is intricately linked to pathological heart disease, the transcription factors driving these gene programs remain ill-defined. Here we report that increased calcineurin/Nfat signalling and decreased miR-25 expression integrate to re-express the basic helix-loop-helix (bHLH) transcription factor dHAND (also known as Hand2) in the diseased human and mouse myocardium. In line, mutant mice overexpressing Hand2 in otherwise healthy heart muscle cells developed a phenotype of pathological hypertrophy. Conversely, conditional gene-targeted Hand2 mice demonstrated a marked resistance to pressure-overload-induced hypertrophy, fibrosis, ventricular dysfunction and induction of a fetal gene program. Furthermore, in vivo inhibition of miR-25 by a specific antagomir evoked spontaneous cardiac dysfunction and sensitized the murine myocardium to heart failure in a Hand2-dependent manner. Our results reveal that signalling cascades integrate with microRNAs to induce the expression of the bHLH transcription factor Hand2 in the postnatal mammalian myocardium with impact on embryonic gene programs in heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Heart Failure/metabolism , MicroRNAs/physiology , NFATC Transcription Factors/physiology , Animals , Base Sequence , Gene Expression Profiling , Gene Silencing , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , NFATC Transcription Factors/metabolism , RNA Processing, Post-Transcriptional , Sequence Homology, Nucleic Acid , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL