Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David J. Morgans is active.

Publication


Featured researches published by David J. Morgans.


Science | 2011

Cardiac Myosin Activation: A Potential Therapeutic Approach for Systolic Heart Failure

Fady Malik; James J. Hartman; Kathleen A. Elias; Bradley P. Morgan; Hector Rodriguez; Katjuša Brejc; Robert L. Anderson; Sandra H. Sueoka; Kenneth H. Lee; Jeffrey T. Finer; Roman Sakowicz; Ramesh Baliga; D. R. Cox; Marc Garard; Guillermo Godinez; Raja Kawas; Erica Anne Kraynack; David Lenzi; Pu Ping Lu; Alexander Ramon Muci; Congrong Niu; Xiangping Qian; Daniel W. Pierce; Maria V. Pokrovskii; Ion Suehiro; Sheila Sylvester; Todd Tochimoto; Corey Valdez; Wenyue Wang; Tatsuo Katori

A small molecule improves cardiac function by accelerating the transition of myosin into a force-producing state. 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.


Circulation-heart Failure | 2010

Improvement of Cardiac Function by a Cardiac Myosin Activator in Conscious Dogs With Systolic Heart Failure

You-Tang Shen; Fady Malik; Xin Zhao; Christophe Depre; Sunil K. Dhar; Patricio Abarzúa; David J. Morgans; Stephen F. Vatner

Background—Therapy for chronic systolic heart failure (sHF) has improved over the past 2 decades, but the armamentarium of drugs is limited and consequently sHF remains a leading cause of death and disability. In this investigation, we examined the effects of a novel cardiac myosin activator, omecamtiv mecarbil (formerly CK-1827452) in 2 different models of heart failure. Methods and Results—Two different models of sHF were used: (1) pacing-induced sHF after myocardial infarction (MI-sHF) and (2) pacing-induced sHF after 1 year of chronic pressure overload left ventricular hypertrophy (LVH-sHF). Omecamtiv mecarbil increased systolic function in sHF dogs, chronically instrumented to measure LV pressure, wall thickness, and cardiac output. Omecamtiv mecarbil, infused for 24 hours, induced a sustained increase without desensitization (P<0.05) in wall thickening (25±6.2%), stroke volume (44±6.5%) and cardiac output (22±2.8%), and decreased heart rate (15±3.0%). The major differences between the effect of omecamtiv mecarbil on cardiac function and the effect induced by a catecholamine, for example, dobutamine, is that omecamtiv mecarbil did not increase LV dP/dt but rather increased LV systolic ejection time by 26±2.9% in sHF. Another key difference is that myocardial O2 consumption (MVO2), which increases with catecholamines, was not significantly affected by omecamtiv mecarbil. Conclusions—These results demonstrate that chronic infusion of the cardiac myosin activator, omecamtiv mecarbil, improves LV function in sHF without the limitations of progressive desensitization and increased MVO2. This unique profile may provide a new therapeutic approach for patients with sHF.


Nature Medicine | 2012

Activation of fast skeletal muscle troponin as a potential therapeutic approach for treating neuromuscular diseases

Alan Russell; James J. Hartman; Aaron C. Hinken; Alexander Ramon Muci; Raja Kawas; Lena Driscoll; Guillermo Godinez; Kenneth H. Lee; David Marquez; William F. Browne; Michael M. Chen; David Clarke; Scott Collibee; Marc Garard; Richard Hansen; Zhiheng Jia; Pu Ping Lu; Hector Rodriguez; Khalil G. Saikali; Julia Schaletzky; Vipin Vijayakumar; Daniel L. Albertus; Dennis R. Claflin; David J. Morgans; Bradley P. Morgan; Fady Malik

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.


ACS Medicinal Chemistry Letters | 2010

Discovery of Omecamtiv Mecarbil the First, Selective, Small Molecule Activator of Cardiac Myosin

Bradley P. Morgan; Alexander Ramon Muci; Pu-Ping Lu; Xiangping Qian; Todd Tochimoto; Whitney W. Smith; Marc Garard; Erica Anne Kraynack; Scott Collibee; Ion Suehiro; Adam Lewis Tomasi; S. Corey Valdez; Wenyue Wang; Hong Jiang; James J. Hartman; Hector Rodriguez; Raja Kawas; Sheila Sylvester; Kathleen A. Elias; Guillermo Godinez; Kenneth H. Lee; Robert L. Anderson; Sandra H. Sueoka; Donghong Xu; Zhengping Wang; Nebojsa Djordjevic; Fady Malik; David J. Morgans

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.


PLOS ONE | 2014

Fast Skeletal Muscle Troponin Activator tirasemtiv Increases Muscle Function and Performance in the B6SJL-SOD1G93A ALS Mouse Model

Darren T. Hwee; Adam D. Kennedy; Julie Ryans; Alan J. Russell; Zhiheng Jia; Aaron C. Hinken; David J. Morgans; Fady Malik; Jeffrey R. Jasper

Amyotrophic Lateral Sclerosis (ALS) is a motor neuron disease characterized by progressive motor neuron loss resulting in muscle atrophy, declining muscle function, and eventual paralysis. Patients typically die from respiratory failure 3 to 5 years from the onset of symptoms. Tirasemtiv is a fast skeletal troponin activator that sensitizes the sarcomere to calcium; this mechanism of action amplifies the response of muscle to neuromuscular input producing greater force when nerve input is reduced. Here, we demonstrate that a single dose of tirasemtiv significantly increases submaximal isometric force, forelimb grip strength, grid hang time, and rotarod performance in a female transgenic mouse model (B6SJL-SOD1G93A) of ALS with functional deficits. Additionally, diaphragm force and tidal volume are significantly higher in tirasemtiv-treated female B6SJL-SOD1G93A mice. These results support the potential of fast skeletal troponin activators to improve muscle function in neuromuscular diseases.


Muscle & Nerve | 2014

TIRASEMTIV AMPLIFIES SKELETAL MUSCLE RESPONSE TO NERVE ACTIVATION IN HUMANS

Richard Hansen; Khalil G. Saikali; Willis Chou; Alan J. Russell; Michael M. Chen; Vipin Vijayakumar; Randall Stoltz; Stéphane Baudry; Roger M. Enoka; David J. Morgans; Andrew A. Wolff; Fady Malik

Introduction: In this study we tested the hypothesis that tirasemtiv, a selective fast skeletal muscle troponin activator that sensitizes the sarcomere to calcium, could amplify the response of muscle to neuromuscular input in humans. Methods: Healthy men received tirasemtiv and placebo in a randomized, double‐blind, 4‐period, crossover design. The deep fibular nerve was stimulated transcutaneously to activate the tibialis anterior muscle and produce dorsiflexion of the foot. The force–frequency relationship of tibialis anterior dorsiflexion was assessed after dosing. Results: Tirasemtiv increased force produced by the tibialis anterior in a dose‐, concentration‐, and frequency‐dependent manner with the largest increases [up to 24.5% (SE 3.1), Pu2009<u20090.0001] produced at subtetanic nerve stimulation frequencies (10 Hz). Conclusions: The data confirm that tirasemtiv amplifies the response of skeletal muscle to nerve input in humans. This outcome provides support for further studies of tirasemtiv as a potential therapy in conditions marked by diminished neuromuscular input. Muscle Nerve 50: 925–931, 2014


Journal of Pharmacology and Experimental Therapeutics | 2011

Inhibition of Smooth Muscle Myosin as a Novel Therapeutic Target for Hypertension

Xin Zhao; David Ho; Patricio Abarzúa; Sunil K. Dhar; Xi Wang; Zhiheng Jia; Malar Pannirselvam; David J. Morgans; Fady Malik; Stephen F. Vatner

We examined a novel therapeutic approach for hypertension, a small-molecule direct inhibitor of smooth muscle myosin, CK-2018448 (CK-448), which is an N,N-alkylurea (U.S. Patent Publication 2009–0275537 A1) in conscious dogs with renal hypertension and compared its efficacy with that of a calcium channel blocker, amlodipine. Dogs were instrumented with a miniature left ventricular pressure gauge, an aortic pressure catheter, and ultrasonic flow probes in the ascending aorta and renal and iliac arteries for measurement of cardiac output and regional blood flow. In the hypertensive state, mean arterial pressure increased from 101 ± 3.8 to 142 ± 1.9 mm Hg. At the doses selected, CK-448 and amlodipine increased cardiac output similarly (30 ± 11% versus 33 ± 6.4%) and similarly reduced mean arterial pressure (−22 ± 3.6% versus −16 ± 3.4%) and total peripheral resistance (−36 ± 5.9% versus −37 ± 5.8%). CK-448 had the greatest vasodilator effect in the renal bed, where renal blood flow increased by 46 ± 9.0%, versus 11 ± 3.4% for amlodipine (p < 0.01). CK-488 produced significantly less vasodilation in the limb, where iliac blood flow did not change; in contrast, it rose by 48 ± 12% with amlodipine (p < 0.01). The minimal effects on limb blood flow could limit the development of peripheral edema, an adverse side effect of Ca2+ channel blockers. In addition, in a rodent model of hypertension, oral administration of a smooth muscle myosin inhibitor resulted in a sustained antihypertensive effect. Thus, the smooth muscle myosin inhibitors preferential effect on renal blood flow makes this drug mechanism particularly appealing, because many patients with hypertension have renal insufficiency, and patients with heart failure could benefit from afterload reduction coupled with enhanced renal blood flow.


PLOS ONE | 2012

Smooth Muscle Myosin Inhibition: A Novel Therapeutic Approach for Pulmonary Hypertension

David Ho; Li Chen; Xin Zhao; Nicquanna Durham; Malar Pannirselvam; Dorothy E. Vatner; David J. Morgans; Fady Malik; Stephen F. Vatner; You-Tang Shen

Objective Pulmonary hypertension remains a major clinical problem despite current therapies. In this study, we examine for the first time a novel pharmacological target, smooth muscle myosin, and determine if the smooth muscle myosin inhibitor, CK-2019165 (CK-165) ameliorates pulmonary hypertension. Materials and Methods Six domestic female pigs were surgically instrumented to measure pulmonary blood flow and systemic and pulmonary vascular dynamics. Pulmonary hypertension was induced by hypoxia, or infusion of the thromboxane analog (U-46619, 0.1 µg/kg/min, i.v.). In rats, chronic pulmonary hypertension was induced by monocrotaline. Results CK-165 (4 mg/kg, i.v.) reduced pulmonary vascular resistance by 22±3 and 28±6% from baseline in hypoxia and thromboxane pig models, respectively (p<0.01 and 0.01), while mean arterial pressure also fell and heart rate rose slightly. When CK-165 was delivered via inhalation in the hypoxia model, pulmonary vascular resistance fell by 17±6% (p<0.05) while mean arterial pressure and heart rate were unchanged. In the monocrotaline model of chronic pulmonary hypertension, inhaled CK-165 resulted in a similar (18.0±3.8%) reduction in right ventricular systolic pressure as compared with sildenafil (20.3±4.5%). Conclusion Inhibition of smooth muscle myosin may be a novel therapeutic target for treatment of pulmonary hypertension.


ACS Medicinal Chemistry Letters | 2018

Discovery of Tirasemtiv, the First Direct Fast Skeletal Muscle Troponin Activator

Scott Collibee; Gustave Bergnes; Alexander Ramon Muci; William F. Browne; Marc Garard; Aaron C. Hinken; Alan J. Russell; Ion Suehiro; James J. Hartman; Raja Kawas; Pu-Ping Lu; Kenneth H. Lee; David Marquez; Matthew Tomlinson; Donghong Xu; Adam D. Kennedy; Darren Hwee; Julia Schaletzky; Kwan Leung; Fady Malik; David J. Morgans; Bradley P. Morgan

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.


Journal of Cardiac Failure | 2006

Activating Cardiac Myosin, a Novel Inotropic Mechanism To Improve Cardiac Function in Conscious Dogs with Congestive Heart Failure

You-Tang Shen; Stephen F. Vatner; David J. Morgans; Fady Malik

Collaboration


Dive into the David J. Morgans's collaboration.

Top Co-Authors

Avatar

Fady Malik

University of California

View shared research outputs
Top Co-Authors

Avatar

Stephen F. Vatner

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

You-Tang Shen

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xin Zhao

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Alan J. Russell

Carnegie Mellon University

View shared research outputs
Researchain Logo
Decentralizing Knowledge