Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 9(4): 354-358, 2018 Apr 12.
Article in English | MEDLINE | ID: mdl-29670700

ABSTRACT

The identification and optimization of the first activators of fast skeletal muscle are reported. Compound 1 was identified from high-throughput screening (HTS) and subsequently found to improve muscle function via interaction with the troponin complex. Optimization of 1 for potency, metabolic stability, and physical properties led to the discovery of tirasemtiv (25), which has been extensively characterized in clinical trials for the treatment of amyotrophic lateral sclerosis.

2.
J Biol Chem ; 292(40): 16571-16577, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28808052

ABSTRACT

Mavacamten, formerly known as MYK-461 is a recently discovered novel small-molecule modulator of cardiac myosin that targets the underlying sarcomere hypercontractility of hypertrophic cardiomyopathy, one of the most prevalent heritable cardiovascular disorders. Studies on isolated cells and muscle fibers as well as intact animals have shown that mavacamten inhibits sarcomere force production, thereby reducing cardiac contractility. Initial mechanistic studies have suggested that mavacamten primarily reduces the steady-state ATPase activity by inhibiting the rate of phosphate release of ß-cardiac myosin-S1, but the molecular mechanism of action of mavacamten has not been described. Here we used steady-state and presteady-state kinetic analyses to investigate the mechanism of action of mavacamten. Transient kinetic analyses revealed that mavacamten modulates multiple steps of the myosin chemomechanical cycle. In addition to decreasing the rate-limiting step of the cycle (phosphate release), mavacamten reduced the number of myosin-S1 heads that can interact with the actin thin filament during transition from the weakly to the strongly bound state without affecting the intrinsic rate. Mavacamten also decreased the rate of myosin binding to actin in the ADP-bound state and the ADP-release rate from myosin-S1 alone. We, therefore, conclude that mavacamten acts on multiple stages of the myosin chemomechanical cycle. Although the primary mechanism of mavacamten-mediated inhibition of cardiac myosin is the decrease of phosphate release from ß-cardiac myosin-S1, a secondary mechanism decreases the number of actin-binding heads transitioning from the weakly to the strongly bound state, which occurs before phosphate release and may provide an additional method to modulate myosin function.


Subject(s)
Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Benzylamines/chemistry , Cardiac Myosins/chemistry , Myosin Subfragments/chemistry , Sarcomeres/chemistry , Uracil/analogs & derivatives , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cardiac Myosins/metabolism , Cardiomegaly/metabolism , Cattle , Myosin Subfragments/metabolism , Sarcomeres/metabolism , Uracil/chemistry
3.
Science ; 351(6273): 617-21, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26912705

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited disease of heart muscle that can be caused by mutations in sarcomere proteins. Clinical diagnosis depends on an abnormal thickening of the heart, but the earliest signs of disease are hyperdynamic contraction and impaired relaxation. Whereas some in vitro studies of power generation by mutant and wild-type sarcomere proteins are consistent with mutant sarcomeres exhibiting enhanced contractile power, others are not. We identified a small molecule, MYK-461, that reduces contractility by decreasing the adenosine triphosphatase activity of the cardiac myosin heavy chain. Here we demonstrate that early, chronic administration of MYK-461 suppresses the development of ventricular hypertrophy, cardiomyocyte disarray, and myocardial fibrosis and attenuates hypertrophic and profibrotic gene expression in mice harboring heterozygous human mutations in the myosin heavy chain. These data indicate that hyperdynamic contraction is essential for HCM pathobiology and that inhibitors of sarcomere contraction may be a valuable therapeutic approach for HCM.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Benzylamines/administration & dosage , Cardiac Myosins/antagonists & inhibitors , Cardiomyopathy, Hypertrophic, Familial/drug therapy , Myocardial Contraction/drug effects , Myosin Heavy Chains/antagonists & inhibitors , Sarcomeres/drug effects , Uracil/analogs & derivatives , Animals , Benzylamines/chemistry , Cardiac Myosins/genetics , Cardiomyopathy, Hypertrophic, Familial/pathology , Cardiomyopathy, Hypertrophic, Familial/physiopathology , Cells, Cultured , Disease Models, Animal , Fibrosis , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heterozygote , Humans , Male , Mice , Mice, Inbred Strains , Mutation , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosin Heavy Chains/genetics , Rats , Uracil/administration & dosage , Uracil/chemistry
4.
Nat Med ; 18(3): 452-5, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344294

ABSTRACT

Limited neural input results in muscle weakness in neuromuscular disease because of a reduction in the density of muscle innervation, the rate of neuromuscular junction activation or the efficiency of synaptic transmission. We developed a small-molecule fast-skeletal-troponin activator, CK-2017357, as a means to increase muscle strength by amplifying the response of muscle when neural input is otherwise diminished secondary to neuromuscular disease. Binding selectively to the fast-skeletal-troponin complex, CK-2017357 slows the rate of calcium release from troponin C and sensitizes muscle to calcium. As a consequence, the force-calcium relationship of muscle fibers shifts leftwards, as does the force-frequency relationship of a nerve-muscle pair, so that CK-2017357 increases the production of muscle force in situ at sub-maximal nerve stimulation rates. Notably, we show that sensitization of the fast-skeletal-troponin complex to calcium improves muscle force and grip strength immediately after administration of single doses of CK-2017357 in a model of the neuromuscular disease myasthenia gravis. Troponin activation may provide a new therapeutic approach to improve physical activity in diseases where neuromuscular function is compromised.


Subject(s)
Calcium/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Diseases/metabolism , Troponin C/agonists , Troponin C/metabolism , Adenosine Triphosphatases/metabolism , Animals , Cattle , Humans , Imidazoles/chemistry , Imidazoles/therapeutic use , Molecular Targeted Therapy , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/pathology , Myasthenia Gravis/drug therapy , Myasthenia Gravis/metabolism , Myasthenia Gravis/pathology , Myosins/isolation & purification , Myosins/metabolism , Neuromuscular Diseases/drug therapy , Neuromuscular Diseases/pathology , Pyrazines/chemistry , Pyrazines/therapeutic use , Rabbits , Rats , Troponin/metabolism , Troponin/physiology
5.
Science ; 331(6023): 1439-43, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21415352

ABSTRACT

Decreased cardiac contractility is a central feature of systolic heart failure. Existing drugs increase cardiac contractility indirectly through signaling cascades but are limited by their mechanism-related adverse effects. To avoid these limitations, we previously developed omecamtiv mecarbil, a small-molecule, direct activator of cardiac myosin. Here, we show that it binds to the myosin catalytic domain and operates by an allosteric mechanism to increase the transition rate of myosin into the strongly actin-bound force-generating state. Paradoxically, it inhibits adenosine 5'-triphosphate turnover in the absence of actin, which suggests that it stabilizes an actin-bound conformation of myosin. In animal models, omecamtiv mecarbil increases cardiac function by increasing the duration of ejection without changing the rates of contraction. Cardiac myosin activation may provide a new therapeutic approach for systolic heart failure.


Subject(s)
Cardiac Myosins/metabolism , Heart Failure, Systolic/drug therapy , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Urea/analogs & derivatives , Actin Cytoskeleton/metabolism , Actins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Adrenergic beta-Agonists/pharmacology , Allosteric Regulation , Animals , Binding Sites , Calcium/metabolism , Cardiac Myosins/chemistry , Cardiac Output/drug effects , Dogs , Female , Heart Failure, Systolic/physiopathology , Isoproterenol/pharmacology , Male , Myocytes, Cardiac/physiology , Phosphates/metabolism , Protein Binding , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Urea/chemistry , Urea/metabolism , Urea/pharmacology , Ventricular Function, Left/drug effects
6.
ACS Med Chem Lett ; 1(9): 472-7, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-24900233

ABSTRACT

We report the design, synthesis, and optimization of the first, selective activators of cardiac myosin. Starting with a poorly soluble, nitro-aromatic hit compound (1), potent, selective, and soluble myosin activators were designed culminating in the discovery of omecamtiv mecarbil (24). Compound 24 is currently in clinical trials for the treatment of systolic heart failure.

SELECTION OF CITATIONS
SEARCH DETAIL
...