Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 259
Filter
1.
Nat Commun ; 15(1): 4885, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849353

ABSTRACT

Inherited cardiomyopathies are common cardiac diseases worldwide, leading in the late stage to heart failure and death. The most promising treatments against these diseases are small molecules directly modulating the force produced by ß-cardiac myosin, the molecular motor driving heart contraction. Omecamtiv mecarbil and Mavacamten are two such molecules that completed phase 3 clinical trials, and the inhibitor Mavacamten is now approved by the FDA. In contrast to Mavacamten, Omecamtiv mecarbil acts as an activator of cardiac contractility. Here, we reveal by X-ray crystallography that both drugs target the same pocket and stabilize a pre-stroke structural state, with only few local differences. All-atom molecular dynamics simulations reveal how these molecules produce distinct effects in motor allostery thus impacting force production in opposite way. Altogether, our results provide the framework for rational drug development for the purpose of personalized medicine.


Subject(s)
Molecular Dynamics Simulation , Myocardial Contraction , Urea , Myocardial Contraction/drug effects , Crystallography, X-Ray , Humans , Urea/analogs & derivatives , Urea/pharmacology , Urea/chemistry , Cardiac Myosins/metabolism , Cardiac Myosins/chemistry , Cardiac Myosins/genetics , Ventricular Myosins/metabolism , Ventricular Myosins/chemistry , Ventricular Myosins/genetics , Animals , Benzylamines , Uracil/analogs & derivatives
2.
J Gen Physiol ; 156(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38709176

ABSTRACT

Dilated cardiomyopathy (DCM) is a condition characterized by impaired cardiac function, due to myocardial hypo-contractility, and is associated with point mutations in ß-cardiac myosin, the molecular motor that powers cardiac contraction. Myocardial function can be modulated through sequestration of myosin motors into an auto-inhibited "super-relaxed" state (SRX), which may be further stabilized by a structural state known as the "interacting heads motif" (IHM). Here, we sought to determine whether hypo-contractility of DCM myocardium results from reduced function of individual myosin molecules or from decreased myosin availability to interact with actin due to increased IHM/SRX stabilization. We used an established DCM myosin mutation, E525K, and characterized the biochemical and mechanical activity of wild-type and mutant human ß-cardiac myosin constructs that differed in the length of their coiled-coil tail, which dictates their ability to form the IHM/SRX state. We found that short-tailed myosin constructs exhibited low IHM/SRX content, elevated actin-activated ATPase activity, and fast velocities in unloaded motility assays. Conversely, longer-tailed constructs exhibited higher IHM/SRX content and reduced actomyosin ATPase and velocity. Our modeling suggests that reduced velocities may be attributed to IHM/SRX-dependent sequestration of myosin heads. Interestingly, longer-tailed E525K mutants showed no apparent impact on velocity or actomyosin ATPase at low ionic strength but stabilized IHM/SRX state at higher ionic strength. Therefore, the hypo-contractility observed in DCM may be attributable to reduced myosin head availability caused by enhanced IHM/SRX stability in E525K mutants.


Subject(s)
Cardiac Myosins , Cardiomyopathy, Dilated , Ventricular Myosins , Animals , Humans , Actins/metabolism , Actins/genetics , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Mutation , Myocardial Contraction/physiology , Ventricular Myosins/genetics , Ventricular Myosins/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism
3.
Sci Rep ; 14(1): 3915, 2024 02 16.
Article in English | MEDLINE | ID: mdl-38365813

ABSTRACT

Human induced pluripotent stem cells and their differentiation into cardiac myocytes (hiPSC-CMs) provides a unique and valuable platform for studies of cardiac muscle structure-function. This includes studies centered on disease etiology, drug development, and for potential clinical applications in heart regeneration/repair. Ultimately, for these applications to achieve success, a thorough assessment and physiological advancement of the structure and function of hiPSC-CMs is required. HiPSC-CMs are well noted for their immature and sub-physiological cardiac muscle state, and this represents a major hurdle for the field. To address this roadblock, we have developed a hiPSC-CMs (ß-MHC dominant) experimental platform focused on directed physiological enhancement of the sarcomere, the functional unit of cardiac muscle. We focus here on the myosin heavy chain (MyHC) protein isoform profile, the molecular motor of the heart, which is essential to cardiac physiological performance. We hypothesized that inducing increased expression of α-MyHC in ß-MyHC dominant hiPSC-CMs would enhance contractile performance of hiPSC-CMs. To test this hypothesis, we used gene editing with an inducible α-MyHC expression cassette into isogeneic hiPSC-CMs, and separately by gene transfer, and then investigated the direct effects of increased α-MyHC expression on hiPSC-CMs contractility and relaxation function. Data show improved cardiac functional parameters in hiPSC-CMs induced with α-MyHC. Positive inotropy and relaxation was evident in comparison to ß-MyHC dominant isogenic controls both at baseline and during pacing induced stress. This approach should facilitate studies of hiPSC-CMs disease modeling and drug screening, as well as advancing fundamental aspects of cardiac function parameters for the optimization of future cardiac regeneration, repair and re-muscularization applications.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Ventricular Myosins/genetics , Ventricular Myosins/metabolism , Ventricular Myosins/pharmacology , Gene Editing , Myocardium , Myocytes, Cardiac/metabolism , Cell Differentiation , Myosins/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism
4.
Proc Natl Acad Sci U S A ; 121(9): e2315472121, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38377203

ABSTRACT

Mutations at a highly conserved homologous residue in three closely related muscle myosins cause three distinct diseases involving muscle defects: R671C in ß-cardiac myosin causes hypertrophic cardiomyopathy, R672C and R672H in embryonic skeletal myosin cause Freeman-Sheldon syndrome, and R674Q in perinatal skeletal myosin causes trismus-pseudocamptodactyly syndrome. It is not known whether their effects at the molecular level are similar to one another or correlate with disease phenotype and severity. To this end, we investigated the effects of the homologous mutations on key factors of molecular power production using recombinantly expressed human ß, embryonic, and perinatal myosin subfragment-1. We found large effects in the developmental myosins but minimal effects in ß myosin, and magnitude of changes correlated partially with clinical severity. The mutations in the developmental myosins dramatically decreased the step size and load-sensitive actin-detachment rate of single molecules measured by optical tweezers, in addition to decreasing overall enzymatic (ATPase) cycle rate. In contrast, the only measured effect of R671C in ß myosin was a larger step size. Our measurements of step size and bound times predicted velocities consistent with those measured in an in vitro motility assay. Finally, molecular dynamics simulations predicted that the arginine to cysteine mutation in embryonic, but not ß, myosin may reduce pre-powerstroke lever arm priming and ADP pocket opening, providing a possible structural mechanism consistent with the experimental observations. This paper presents direct comparisons of homologous mutations in several different myosin isoforms, whose divergent functional effects are a testament to myosin's highly allosteric nature.


Subject(s)
Myosins , Ventricular Myosins , Humans , Ventricular Myosins/genetics , Myosins/metabolism , Adenosine Triphosphatases/metabolism , Mutation , Actins/metabolism , Muscle, Skeletal/metabolism
5.
Nat Commun ; 14(1): 3166, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37258552

ABSTRACT

To save energy and precisely regulate cardiac contractility, cardiac muscle myosin heads are sequestered in an 'off' state that can be converted to an 'on' state when exertion is increased. The 'off' state is equated with a folded-back structure known as the interacting-heads motif (IHM), which is a regulatory feature of all class-2 muscle and non-muscle myosins. We report here the human ß-cardiac myosin IHM structure determined by cryo-electron microscopy to 3.6 Å resolution, providing details of all the interfaces stabilizing the 'off' state. The structure shows that these interfaces are hot spots of hypertrophic cardiomyopathy mutations that are thought to cause hypercontractility by destabilizing the 'off' state. Importantly, the cardiac and smooth muscle myosin IHM structures dramatically differ, providing structural evidence for the divergent physiological regulation of these muscle types. The cardiac IHM structure will facilitate development of clinically useful new molecules that modulate IHM stability.


Subject(s)
Cardiac Myosins , Cardiomyopathy, Hypertrophic , Humans , Ventricular Myosins/chemistry , Ventricular Myosins/genetics , Cryoelectron Microscopy , Heart , Cardiomyopathy, Hypertrophic/genetics
6.
Elife ; 112022 11 24.
Article in English | MEDLINE | ID: mdl-36422472

ABSTRACT

The auto-inhibited, super-relaxed (SRX) state of cardiac myosin is thought to be crucial for regulating contraction, relaxation, and energy conservation in the heart. We used single ATP turnover experiments to demonstrate that a dilated cardiomyopathy (DCM) mutation (E525K) in human beta-cardiac myosin increases the fraction of myosin heads in the SRX state (with slow ATP turnover), especially in physiological ionic strength conditions. We also utilized FRET between a C-terminal GFP tag on the myosin tail and Cy3ATP bound to the active site of the motor domain to estimate the fraction of heads in the closed, interacting-heads motif (IHM); we found a strong correlation between the IHM and SRX state. Negative stain electron microscopy and 2D class averaging of the construct demonstrated that the E525K mutation increased the fraction of molecules adopting the IHM. Overall, our results demonstrate that the E525K DCM mutation may reduce muscle force and power by stabilizing the auto-inhibited SRX state. Our studies also provide direct evidence for a correlation between the SRX biochemical state and the IHM structural state in cardiac muscle myosin. Furthermore, the E525 residue may be implicated in crucial electrostatic interactions that modulate this conserved, auto-inhibited conformation of myosin.


Subject(s)
Cardiomyopathy, Dilated , Ventricular Myosins , Humans , Ventricular Myosins/genetics , Cardiac Myosins , Cardiomyopathy, Dilated/genetics , Myosins/genetics , Mutation , Myocardium , Adenosine Triphosphate
7.
Genes (Basel) ; 13(9)2022 08 29.
Article in English | MEDLINE | ID: mdl-36140722

ABSTRACT

Cardiac dysfunction accelerates the risk of heart failure, and its pathogenesis involves a complex interaction between genetic and environmental factors. Variations in myosin affect contractile abilities of cardiomyocytes and cause structural and functional abnormalities in myocardium. The study aims to find the association of MYH7 rs121913642 (c.1594 T>C) and rs121913645 (c.667G>A) variants with cardiac dysfunction in the Punjabi Pakistani population. Patients with heart failure (n = 232) and healthy controls (n = 205) were enrolled in this study. MYH7 variant genotyping was performed using tetra ARMS-PCR. MYH7 rs121913642 TC genotype was significantly more prevalent in the patient group (p < 0.001). However, MYH7 rs121913645 genotype frequencies were not significantly different between the patient and control groups (p < 0.666). Regression analysis also revealed that the rs121913642 C allele increases the risk of cardiac failure by ~2 [OR:1.98, CI: 1.31−2.98, p < 0.001] in comparison to the T allele. High levels of the cardiac enzymes cardiac troponin I (cTnI) and CK-MB were observed in patients. There was also an increase in total cholesterol, LDL cholesterol, and uric acid in patients compared to the healthy control group (p < 0.001). In conclusion, the MYH7 gene variant rs121913642 is genetically associated with cardiac dysfunction and involved in the pathogenesis of HF.


Subject(s)
Heart Diseases , Heart Failure , Myosin Heavy Chains/genetics , Cardiac Myosins/genetics , Cholesterol, LDL/genetics , Heart Failure/genetics , Humans , Mutation , Phenotype , Troponin I/genetics , Uric Acid , Ventricular Myosins/genetics
8.
Elife ; 112022 06 29.
Article in English | MEDLINE | ID: mdl-35767336

ABSTRACT

Mutations in the lever arm of ß-cardiac myosin are a frequent cause of hypertrophic cardiomyopathy, a disease characterized by hypercontractility and eventual hypertrophy of the left ventricle. Here, we studied five such mutations: three in the pliant region of the lever arm (D778V, L781P, and S782N) and two in the light chain-binding region (A797T and F834L). We investigated their effects on both motor function and myosin subfragment 2 (S2) tail-based autoinhibition. The pliant region mutations had varying effects on the motor function of a myosin construct lacking the S2 tail: overall, D778V increased power output, L781P reduced power output, and S782N had little effect on power output, while all three reduced the external force sensitivity of the actin detachment rate. With a myosin containing the motor domain and the proximal S2 tail, the pliant region mutations also attenuated autoinhibition in the presence of filamentous actin but had no impact in the absence of actin. By contrast, the light chain-binding region mutations had little effect on motor activity but produced marked reductions in autoinhibition in both the presence and absence of actin. Thus, mutations in the lever arm of ß-cardiac myosin have divergent allosteric effects on myosin function, depending on whether they are in the pliant or light chain-binding regions.


Subject(s)
Cardiomyopathy, Hypertrophic , Ventricular Myosins , Actins/genetics , Actins/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Humans , Mutation , Structure-Activity Relationship , Ventricular Myosins/chemistry , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
9.
Cardiovasc Ther ; 2021: 5554569, 2021.
Article in English | MEDLINE | ID: mdl-34257705

ABSTRACT

Ginkgolide B (GB) is an active ingredient extracted from Ginkgo biloba leaves. However, the effects of GB on cardiac hypertrophy remain unclear. The study is aimed at determining whether GB could alleviate cardiac hypertrophy and exploring its underlying molecular mechanism. Rat cardiomyocyte cell line H9c2 cells were pretreated with GB and incubated with angiotensin II (Ang II) to simulate an in vitro cardiac hypertrophy model. Cell viability, cell size, hypertrophy markers, and autophagy were determined in H9c2 cells after Ang II treatment. Proteins involved in autophagy and the SIRT1 pathway were determined by western blot. Our data demonstrated that GB attenuated Ang II-induced cardiac hypertrophy and reduced the mRNA expressions of hypertrophy marker, atrial natriuretic peptide (ANP), and ß-myosin heavy chain (ß-MHC). GB further increased Ang II-induced autophagy in H9c2 cells and modulated expressions of autophagy-related proteins Beclin1 and P62. Modulation of autophagy using autophagy inhibitor 3-methyladenine (3-MA) could abrogate GB-downregulated transcription of NPPA. We then showed that GB attenuated Ang II-induced oxidative stress and reduction in SIRT1 and FoxO1 protein expression. Finally, the effect of GB on autophagy and cardiac hypertrophy could be reversed by SIRT1 inhibitor EX-527. GB inhibits Ang II-induced cardiac hypertrophy by enhancing autophagy via the SIRT1-FoxO1 signaling pathway and might be a potential agent in treating pathological cardiac hypertrophy.


Subject(s)
Angiotensin II/toxicity , Autophagy/drug effects , Ginkgolides/pharmacology , Lactones/pharmacology , Myocytes, Cardiac/drug effects , Nerve Tissue Proteins/metabolism , Sirtuin 1/metabolism , Animals , Atrial Natriuretic Factor/genetics , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Line , Myocytes, Cardiac/pathology , Protective Agents/pharmacology , Rats , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Ventricular Myosins/genetics
10.
Proc Natl Acad Sci U S A ; 118(24)2021 06 15.
Article in English | MEDLINE | ID: mdl-34117120

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is the most common inherited form of heart disease, associated with over 1,000 mutations, many in ß-cardiac myosin (MYH7). Molecular studies of myosin with different HCM mutations have revealed a diversity of effects on ATPase and load-sensitive rate of detachment from actin. It has been difficult to predict how such diverse molecular effects combine to influence forces at the cellular level and further influence cellular phenotypes. This study focused on the P710R mutation that dramatically decreased in vitro motility velocity and actin-activated ATPase, in contrast to other MYH7 mutations. Optical trap measurements of single myosin molecules revealed that this mutation reduced the step size of the myosin motor and the load sensitivity of the actin detachment rate. Conversely, this mutation destabilized the super relaxed state in longer, two-headed myosin constructs, freeing more heads to generate force. Micropatterned human induced pluripotent derived stem cell (hiPSC)-cardiomyocytes CRISPR-edited with the P710R mutation produced significantly increased force (measured by traction force microscopy) compared with isogenic control cells. The P710R mutation also caused cardiomyocyte hypertrophy and cytoskeletal remodeling as measured by immunostaining and electron microscopy. Cellular hypertrophy was prevented in the P710R cells by inhibition of ERK or Akt. Finally, we used a computational model that integrated the measured molecular changes to predict the measured traction forces. These results confirm a key role for regulation of the super relaxed state in driving hypercontractility in HCM with the P710R mutation and demonstrate the value of a multiscale approach in revealing key mechanisms of disease.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Mutation/genetics , Myocardial Contraction/genetics , Ventricular Myosins/genetics , Actins/metabolism , Animals , Biomechanical Phenomena , Calcium/metabolism , Cell Line , Cell Size , Genetic Predisposition to Disease , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Myofibrils/metabolism
11.
Front Immunol ; 12: 620541, 2021.
Article in English | MEDLINE | ID: mdl-33763067

ABSTRACT

Tenascin-C (TNC) is an extracellular matrix glycoprotein that is expressed during embryogenesis. It is not expressed in normal adults, but is up-regulated under pathological conditions. Although TNC knockout mice do not show a distinct phenotype, analyses of disease models using TNC knockout mice combined with in vitro experiments revealed the diverse functions of TNC. Since high TNC levels often predict a poor prognosis in various clinical settings, we developed a transgenic mouse that overexpresses TNC through Cre recombinase-mediated activation. Genomic walking showed that the transgene was integrated into and truncated the Atp8a2 gene. While homozygous transgenic mice showed a severe neurological phenotype, heterozygous mice were viable, fertile, and did not exhibit any distinct abnormalities. Breeding hemizygous mice with Nkx2.5 promoter-Cre or α-myosin heavy chain promoter Cre mice induced the heart-specific overexpression of TNC in embryos and adults. TNC-overexpressing mouse hearts did not have distinct histological or functional abnormalities. However, the expression of proinflammatory cytokines/chemokines was significantly up-regulated and mortality rates during the acute stage after myocardial infarction were significantly higher than those of the controls. Our novel transgenic mouse may be applied to investigations on the role of TNC overexpression in vivo in various tissue/organ pathologies using different Cre donors.


Subject(s)
Myocardial Infarction/immunology , Neurodegenerative Diseases/genetics , Tenascin/genetics , Animals , Chromosome Walking , Cytokines/metabolism , Gene Expression Regulation, Developmental , Genome , Homozygote , Inflammation Mediators/metabolism , Integrases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Promoter Regions, Genetic/genetics , Tenascin/metabolism , Ventricular Myosins/genetics
12.
Elife ; 102021 02 19.
Article in English | MEDLINE | ID: mdl-33605878

ABSTRACT

Hypertrophic cardiomyopathies (HCMs) are the leading cause of acute cardiac failure in young individuals. Over 300 mutations throughout ß-cardiac myosin, including in the motor domain, are associated with HCM. A ß-cardiac myosin motor mutation (R712L) leads to a severe form of HCM. Actin-gliding motility of R712L-myosin is inhibited, despite near-normal ATPase kinetics. By optical trapping, the working stroke of R712L-myosin was decreased 4-fold, but actin-attachment durations were normal. A prevalent hypothesis that HCM mutants are hypercontractile is thus not universal. R712 is adjacent to the binding site of the heart failure drug omecamtiv mecarbil (OM). OM suppresses the working stroke of normal ß-cardiac myosin, but remarkably, OM rescues the R712L-myosin working stroke. Using a flow chamber to interrogate a single molecule during buffer exchange, we found OM rescue to be reversible. Thus, the R712L mutation uncouples lever arm rotation from ATPase activity and this inhibition is rescued by OM.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Mutation , Urea/analogs & derivatives , Ventricular Myosins/genetics , Humans , Urea/pharmacology , Ventricular Myosins/chemistry
13.
Int J Mol Sci ; 22(2)2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33466825

ABSTRACT

Dilated cardiomyopathy (DCM) is a potentially lethal disorder characterized by progressive impairment of cardiac function. Chronic myocarditis has long been hypothesized to be one of the causes of DCM. However, owing to the lack of suitable animal models of chronic myocarditis, its pathophysiology remains unclear. Here, we report a novel mouse model of chronic myocarditis induced by recombinant bacille Calmette-Guérin (rBCG) expressing a CD4+ T-cell epitope of cardiac myosin heavy chain-α (rBCG-MyHCα). Mice immunized with rBCG-MyHCα developed chronic myocarditis, and echocardiography revealed dilation and impaired contraction of ventricles, similar to those observed in human DCM. In the heart, CD62L-CD4+ T cells were increased and produced significant amounts of IFN-γ and IL-17 in response to cardiac myosin. Adoptive transfer of CD62L-CD4+ T cells induced myocarditis in the recipient mice, which indicated that CD62L-CD4+ T cells were the effector cells in this model. rBCG-MyHCα-infected dendritic cells produced proinflammatory cytokines and induced MyHCα-specific T-cell proliferation and Th1 and Th17 polarization. This novel chronic myocarditis mouse model may allow the identification of the central pathophysiological and immunological processes involved in the progression to DCM.


Subject(s)
BCG Vaccine/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Myocarditis/immunology , Ventricular Myosins/immunology , Animals , BCG Vaccine/genetics , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Chronic Disease , Cytokines/immunology , Cytokines/metabolism , Echocardiography , Epitopes, T-Lymphocyte/genetics , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Lymphocyte Activation , Male , Mice, Inbred BALB C , Myocarditis/pathology , Myocarditis/physiopathology , Recombinant Proteins/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Ventricular Myosins/genetics
14.
Eur J Pharmacol ; 882: 173287, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32585157

ABSTRACT

Expression of the ß-myosin heavy chain (ß-MHC), a major component of the cardiac contractile apparatus, is tightly regulated as even modest increases can be detrimental to heart under stress. In healthy hearts, continuous inhibition of ß-adrenergic tone upregulates ß-MHC expression. However, it is unknown whether the duration of the ß-adrenergic inhibition and ß-MHC expression are related. Here, we evaluated the effects of intermittent ß-blockade on cardiac ß-MHC expression. To this end, the ß-blocker propranolol, at the dose of 15mg/kg, was administered once a day in mice for 14 days. This dosing schedule caused daily drug-free periods of at least 6 h as evidenced by propranolol plasma concentrations and cardiac ß-adrenergic responsiveness. Under these conditions, ß-MHC expression decreased by about 75% compared to controls. This effect was abolished in mice lacking ß1- but not ß2-adrenergic receptors (ß-AR) indicating that ß-MHC expression is regulated in a ß1-AR-dependent manner. In ß1-AR knockout mice, the baseline ß-MHC expression was fourfold higher than in wild-type mice. Also, we evaluated the impact of intermittent ß-blockade on ß-MHC expression in mice with systolic dysfunction, in which an increased ß-MHC expression occurs. At 3 weeks after myocardial infarction, mice showed systolic dysfunction and upregulation of ß-MHC expression. Intermittent ß-blockade decreased ß-MHC expression while attenuating cardiac dysfunction. In vitro studies showed that propranolol does not affect ß-MHC expression on its own but antagonizes catecholamine effects on ß-MHC expression. In conclusion, a direct relationship occurs between the duration of the ß-adrenergic inhibition and ß-MHC expression through the ß1-AR.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Myocardium/metabolism , Myosin Heavy Chains/genetics , Propranolol/pharmacology , Receptors, Adrenergic, beta/genetics , Ventricular Myosins/genetics , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/blood , Adrenergic beta-Antagonists/pharmacokinetics , Adrenergic beta-Antagonists/therapeutic use , Animals , Down-Regulation/drug effects , Female , Isoproterenol/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Propranolol/blood , Propranolol/pharmacokinetics , Propranolol/therapeutic use
15.
Sci Adv ; 6(14): eaax0069, 2020 04.
Article in English | MEDLINE | ID: mdl-32284968

ABSTRACT

Hypertrophic cardiomyopathy (HCM) mutations in ß-cardiac myosin and myosin binding protein-C (MyBP-C) lead to hypercontractility of the heart, an early hallmark of HCM. We show that hypercontractility caused by the HCM-causing mutation R663H cannot be explained by changes in fundamental myosin contractile parameters, much like the HCM-causing mutation R403Q. Using enzymatic assays with purified human ß-cardiac myosin, we provide evidence that both mutations cause hypercontractility by increasing the number of functionally accessible myosin heads. We also demonstrate that the myosin mutation R403Q, but not R663H, ablates the binding of myosin with the C0-C7 fragment of MyBP-C. Furthermore, addition of C0-C7 decreases the wild-type myosin basal ATPase single turnover rate, while the mutants do not show a similar reduction. These data suggest that a primary mechanism of action for these mutations is to increase the number of myosin heads functionally available for interaction with actin, which could contribute to hypercontractility.


Subject(s)
Actins/metabolism , Alleles , Amino Acid Substitution , Cardiomyopathy, Hypertrophic/genetics , Mutation , Myosins/genetics , Myosins/metabolism , Actins/chemistry , Binding Sites , Cardiomyopathy, Hypertrophic/physiopathology , Genetic Predisposition to Disease , Humans , Models, Molecular , Myocardial Contraction/genetics , Myosins/chemistry , Protein Binding , Protein Conformation , Structure-Activity Relationship , Ventricular Myosins/genetics
16.
J Biol Chem ; 294(46): 17451-17462, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31582565

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a common genetic disorder characterized by left ventricular hypertrophy and cardiac hyper-contractility. Mutations in the ß-cardiac myosin heavy chain gene (ß-MyHC) are a major cause of HCM, but the specific mechanistic changes to myosin function that lead to this disease remain incompletely understood. Predicting the severity of any ß-MyHC mutation is hindered by a lack of detailed examinations at the molecular level. Moreover, because HCM can take ≥20 years to develop, the severity of the mutations must be somewhat subtle. We hypothesized that mutations that result in early onset disease would have more severe changes in function than do later onset mutations. Here, we performed steady-state and transient kinetic analyses of myosins carrying one of seven missense mutations in the motor domain. Of these seven, four were previously identified in early onset cardiomyopathy screens. We used the parameters derived from these analyses to model the ATP-driven cross-bridge cycle. Contrary to our hypothesis, the results indicated no clear differences between early and late onset HCM mutations. Despite the lack of distinction between early and late onset HCM, the predicted occupancy of the force-holding actin·myosin·ADP complex at [Actin] = 3 Kapp along with the closely related duty ratio (the fraction of myosin in strongly attached force-holding states), and the measured ATPases all changed in parallel (in both sign and degree of change) compared with wildtype (WT) values. Six of the seven HCM mutations were clearly distinct from a set of previously characterized DCM mutations.


Subject(s)
Adenosine Triphosphatases/genetics , Cardiomyopathy, Hypertrophic/genetics , Myosins/genetics , Ventricular Myosins/genetics , Actin Cytoskeleton/genetics , Actins/chemistry , Actins/genetics , Adenosine Triphosphatases/chemistry , Age of Onset , Cardiomyopathy, Hypertrophic/pathology , Female , Humans , Kinetics , Male , Mutation, Missense/genetics , Myocardial Contraction/genetics , Myosin Light Chains/chemistry , Myosin Light Chains/genetics , Myosins/chemistry , Severity of Illness Index , Ventricular Myosins/chemistry
17.
J Biol Chem ; 294(46): 17314-17325, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31578282

ABSTRACT

We investigated a dilated cardiomyopathy (DCM) mutation (F764L) in human ß-cardiac myosin by determining its motor properties in the presence and absence of the heart failure drug omecamtive mecarbil (OM). The mutation is located in the converter domain, a key region of communication between the catalytic motor and lever arm in myosins, and is nearby but not directly in the OM-binding site. We expressed and purified human ß-cardiac myosin subfragment 1 (M2ß-S1) containing the F764L mutation, and compared it to WT with in vitro motility as well as steady-state and transient kinetics measurements. In the absence of OM we demonstrate that the F764L mutation does not significantly change maximum actin-activated ATPase activity but slows actin sliding velocity (15%) and the actomyosin ADP release rate constant (25%). The transient kinetic analysis without OM demonstrates that F764L has a similar duty ratio as WT in unloaded conditions. OM is known to enhance force generation in cardiac muscle while it inhibits the myosin power stroke and enhances actin-attachment duration. We found that OM has a reduced impact on F764L ATPase and sliding velocity compared with WT. Specifically, the EC50 for OM induced inhibition of in vitro motility was 3-fold weaker in F764L. Also, OM reduces maximum actin-activated ATPase 2-fold in F764L, compared with 4-fold with WT. Overall, our results suggest that F764L attenuates the impact of OM on actin-attachment duration and/or the power stroke. Our work highlights the importance of mutation-specific considerations when pursuing small molecule therapies for cardiomyopathies.


Subject(s)
Cardiomyopathy, Dilated/genetics , Heart Failure/genetics , Urea/analogs & derivatives , Ventricular Myosins/genetics , Actin Cytoskeleton/drug effects , Actins/genetics , Actins/metabolism , Actomyosin/genetics , Adenosine Triphosphatases/genetics , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/pathology , Heart Failure/drug therapy , Heart Failure/pathology , Humans , Kinetics , Motor Activity/genetics , Mutation , Myocardial Contraction/drug effects , Protein Domains/genetics , Urea/pharmacology , Ventricular Myosins/chemistry , Ventricular Myosins/metabolism
18.
Appl Biochem Biotechnol ; 189(2): 396-410, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31025171

ABSTRACT

Although embryonic stem (ES) cells (ESCs) may be a promising donor source for the repair of infarcted or ischemic heart tissues, their successful application in regenerative medicine has been hampered by difficulties in enriching, identifying, and selecting cardiomyocytes from the differentiating cells. We established transgenic human ES cell lines by transcriptional control of the α-cardiac myosin heavy chain (α-MHC) promoter driving green fluorescent protein (GFP) expression. Differentiated GFP-expressing cells display the characteristics of cardiomyocytes (CMs). Apela, a recently identified short peptide, up-regulated the expression of the cardiac-restricted transcription factors Tbx5 and GATA4 as well as differentiated the cardiomyocyte markers α-MHC and ß-MHC. Flow cytometric analysis showed that apela increased the percentage of GFP-expressing cells in the beating foci of the embryoid bodies. The percentage of cardiac troponin T (TNT)-positive cells and the protein expression of TNT were increased in the ES cell-derived CMs with apela treatment. Functionally, the contractile frequency of the ES-derived CMs responded appropriately to the vasoactive drugs isoprenaline and carbachol. Our work presented a protocol for specially labelling and enriching CMs by combining transgenic human ES cell lines and exogenous growth factor treatment.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Peptide Hormones/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Human Embryonic Stem Cells/cytology , Humans , Myocytes, Cardiac/cytology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Peptide Hormones/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Troponin T/metabolism , Up-Regulation , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
19.
Pflugers Arch ; 471(5): 701-717, 2019 05.
Article in English | MEDLINE | ID: mdl-30767072

ABSTRACT

Several lines of evidence suggest that the primary effect of hypertrophic cardiomyopathy mutations in human ß-cardiac myosin is hypercontractility of the heart, which leads to subsequent hypertrophy, fibrosis, and myofilament disarray. Here, I describe three perspectives on the molecular basis of this hypercontractility. The first is that hypercontractility results from changes in the fundamental parameters of the actin-activated ß-cardiac myosin chemo-mechanical ATPase cycle. The second considers that hypercontractility results from an increase in the number of functionally accessible heads in the sarcomere for interaction with actin. The final and third perspective is that load dependence of contractility is affected by cardiomyopathy mutations and small-molecule effectors in a manner that changes the power output of cardiac contraction. Experimental approaches associated with each perspective are described along with concepts of therapeutic approaches that could prove valuable in treating hypertrophic cardiomyopathy.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Myocardial Contraction , Ventricular Myosins/genetics , Animals , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/physiopathology , Humans , Mutation , Ventricular Myosins/metabolism
20.
Cardiovasc Diabetol ; 18(1): 13, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30696455

ABSTRACT

BACKGROUND: The diabetic heart undergoes remodelling contributing to an increased incidence of heart failure in individuals with diabetes at a later stage. The molecular regulators that drive this process in the diabetic heart are still unknown. METHODS: Real-time (RT) PCR analysis was performed to determine the expression of cardiac specific microRNA-208a in right atrial appendage (RAA) and left ventricular (LV) biopsy tissues collected from diabetic and non-diabetic patients undergoing coronary artery bypass graft surgery. To determine the time-dependent changes, cardiac tissue were collected from type 2 diabetic mice at different age groups. A western blotting analysis was conducted to determine the expression of contractile proteins α- and ß-myosin heavy chain (MHC) and thyroid hormone receptor-α (TR-α), the negative regulator of ß-MHC. To determine the beneficial effects of therapeutic modulation of miR-208a, high glucose treated adult mouse HL-1 cardiomyocytes were transfected with anti-miR-208a. RESULTS: RT-PCR analysis showed marked upregulation of miR-208a from early stages of diabetes in type 2 diabetic mouse heart, which was associated with a marked increase in the expression of pro-hypertrophic ß-MHC and downregulation of TR-α. Interestingly, upregulation of miR-208a preceded the switch of α-/ß-MHC isoforms and the development of diastolic and systolic dysfunction. We also observed significant upregulation of miR-208a and modulation of miR-208a associated proteins in the type 2 human diabetic heart. Therapeutic inhibition of miR-208a activity in high glucose treated HL-1 cardiomyocytes prevented the activation of ß-MHC and hence the hypertrophic response. CONCLUSION: Our results provide the first evidence that early modulation of miR-208a in the diabetic heart induces alterations in the downstream signaling pathway leading to cardiac remodelling and that therapeutic inhibition of miR-208a may be beneficial in preventing diabetes-induced adverse remodelling of the heart.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/metabolism , MicroRNAs/metabolism , Ventricular Function, Left , Ventricular Remodeling , Aged , Aged, 80 and over , Animals , Cell Line , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation , Heart Ventricles/physiopathology , Humans , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Middle Aged , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Signal Transduction , Time Factors , Ventricular Myosins/genetics , Ventricular Myosins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...