Bio


Dr. Kawana joined the Advanced Heart Failure and Transplant Cardiology group in 2018. He completed his internal medicine, cardiovascular medicine, and heart failure training at Stanford. He also completed a postdoctoral research fellowship under Dr. James Spudich in the Department of Biochemistry. He sees advanced heart failure patients in the clinic and attends CCU/heart failure service, and post-heart transplant and MCS service. His research interests are in the fundamental mechanism of inherited cardiomyopathies, and he studies the effect of gene mutation on the cardiac sarcomere function using cutting-edge biochemical and biophysical approaches, which would lead to the development of novel pharmacotherapy that directly modulates cardiac muscle protein. He is involved in multiple clinical trials for pharmacotherapy in inherited cardiomyopathy and also conducting a device study in heart failure.

Clinical Focus


  • Heart Failure
  • Heart Transplantation
  • Mechanical Circulatory Support
  • Left Ventricular Assist Device
  • Inherited Cardiomyopathy
  • Hypertrophic Cardiomyopathy
  • Dilated Cardiomyopathy
  • Nonischemic cardiomyopathy
  • Cirrhotic Cardiomyopathy
  • Iron overload cardiomyopathy
  • Advanced Heart Failure and Transplant Cardiology

Academic Appointments


Professional Education


  • Board Certification: National Board of Echocardiography, Adult Echocardiography (2017)
  • Board Certification: American Board of Internal Medicine, Advanced Heart Failure and Transplant Cardiology (2018)
  • Residency: Stanford University Internal Medicine Residency (2012) CA
  • Board Certification, American Board of Internal Medicine, Advanced Heart Failure and Transplant Cardiology (2018)
  • Fellowship: Stanford University Advanced Heart Failure and Transplant Fellowship (2018) CA
  • Board Certification: American Board of Internal Medicine, Cardiovascular Disease (2017)
  • Fellowship: Stanford University Cardiovascular Medicine Fellowship (2017) CA
  • Board Certification: American Board of Internal Medicine, Internal Medicine (2012)
  • Medical Education: Warren Alpert Medical School Brown University (2009) RI
  • Fellow, Stanford University Medical Center, Advanced Heart Failure and Transplant Cardiology (2018)
  • Fellow, Stanford University Medical Center, Cardiovascular Medicine (2017)
  • Postdoctoral Fellow, Stanford University School of Medicine, Biochemistry (2015)
  • Resident, Stanford University Medical Center, Internal Medicine (2012)
  • MD, Brown University, Medicine (2009)

Clinical Trials


  • Study for the Evaluation of a Non-invasive Hemodynamic Measurement in Heart Failure Patients Recruiting

    Track changes in non-invasive central venous pressure across hospital stay and relationship with readmission

    View full details

  • Phase 3 Trial to Evaluate the Efficacy and Safety of Aficamten Compared to Placebo in Adults With Symptomatic oHCM (SEQUOIA-HCM) Not Recruiting

    The purpose of this study is to evaluate the efficacy and safety of aficamten (CK-3773274) in adults with symptomatic hypertrophic cardiomyopathy and left ventricular outflow tract obstruction

    Stanford is currently not accepting patients for this trial. For more information, please contact Jennifer Marino, 650-725-6911.

    View full details

  • Single-Ascending Dose Study of JK07 in Subjects With HFpEF Not Recruiting

    A phase 1, randomized, double-blind, placebo-controlled single-ascending dose study to assess JK07 in adult subjects with heart failure with preserved ejection fraction.

    Stanford is currently not accepting patients for this trial. For more information, please contact Jennifer Marino, 650-725-6911.

    View full details

  • Study of JK07 in Subjects With Heart Failure With Reduced Ejection Fraction (HFrEF) Not Recruiting

    This is a Phase 1, randomized, double-blind, placebo-controlled, single-ascending dose study to assess the safety, tolerability, immunogenicity, PK, and exploratory efficacy of JK07 in subjects 18 to 80 years of age with HFrEF ≤40%. Initially 5 cohorts are planned with the option to expand the study to a total of 7 cohorts. The size of the cohorts will range from 5 to 9 subjects. Each cohort will include one single active unblinded sentinel subject receiving a single IV dose of JK07 prior to randomized single dose administration of JK07 or placebo [3:1] in the remainder of the cohort.

    Stanford is currently not accepting patients for this trial. For more information, please contact Nicole Howard, (650) 725 - 6911.

    View full details

All Publications


  • Improved Cardiac Performance and Decreased Arrhythmia in Hypertrophic Cardiomyopathy With Non-β-Blocking R-Enantiomer Carvedilol. Circulation Seo, K., Yamamoto, Y., Kirillova, A., Kawana, M., Yadav, S., Huang, Y., Wang, Q., Lane, K. V., Pruitt, B. L., Perez, M. V., Bernstein, D., Wu, J. C., Wheeler, M. T., Parikh, V. N., Ashley, E. A. 2023

    Abstract

    Hypercontractility and arrhythmia are key pathophysiologic features of hypertrophic cardiomyopathy (HCM), the most common inherited heart disease. β-Adrenergic receptor antagonists (β-blockers) are the first-line therapy for HCM. However, β-blockers commonly selected for this disease are often poorly tolerated in patients, where heart-rate reduction and noncardiac effects can lead to reduced cardiac output and fatigue. Mavacamten, myosin ATPase inhibitor recently approved by the US Food and Drug Administration, has demonstrated the ability to ameliorate hypercontractility without lowering heart rate, but its benefits are so far limited to patients with left ventricular (LV) outflow tract obstruction, and its effect on arrhythmia is unknown.We screened 21 β-blockers for their impact on myocyte contractility and evaluated the antiarrhythmic properties of the most promising drug in a ventricular myocyte arrhythmia model. We then examined its in vivo effect on LV function by hemodynamic pressure-volume loop analysis. The efficacy of the drug was tested in vitro and in vivo compared with current therapeutic options (metoprolol, verapamil, and mavacamten) for HCM in an established mouse model of HCM (Myh6R403Q/+ [myosin heavy chain 6]) and iPSC cardiomyocytes derived from patients with HCM (MYH7R403Q/+) [myosin heavy chain 7]).We identified that carvedilol, a β-blocker not commonly used in HCM, suppresses contractile function and arrhythmia by inhibiting RyR2 (ryanodine receptor type 2). Unlike metoprolol (a β1-blocker), carvedilol markedly reduced LV contractility through RyR2 inhibition, while maintaining stroke volume through α1-adrenergic receptor inhibition in vivo. Clinically available carvedilol is a racemic mixture, and the R-enantiomer, devoid of β-blocking effect, retains the ability to inhibit both α1-receptor and RyR2, thereby suppressing contractile function and arrhythmias without lowering heart rate and cardiac output. In Myh6R403Q/+ mice, R-carvedilol normalized hyperdynamic contraction, suppressed arrhythmia, and increased cardiac output better than metoprolol, verapamil, and mavacamten. The ability of R-carvedilol to suppress contractile function was well retained in MYH7R403Q/+ induced pluripotent stem cell cardiomyocytes.R-enantiomer carvedilol attenuates hyperdynamic contraction, suppresses arrhythmia, and at the same time, improves cardiac output without lowering heart rate by dual blockade of α1-adrenergic receptor and RyR2 in mouse and human models of HCM. This combination of therapeutic effects is unique among current therapeutic options for HCM and may particularly benefit patients without LV outflow tract obstruction.

    View details for DOI 10.1161/CIRCULATIONAHA.123.065017

    View details for PubMedID 37850394

  • Implementation of evidence-based heart failure management: Regional variations between Japan and the USA. Journal of cardiology Ichihara, Y. K., Kohsaka, S., Kisanuki, M., Sandhu, A. T., Kawana, M. 2023

    Abstract

    The implementation of optimal medical therapy is a crucial step in the management of heart failure with reduced ejection fraction (HFrEF). Over the prior three decades, there have been substantial advancements in this field. Early and accurate detection and diagnosis of the disease allow for the appropriate initiation of optimal therapies. The initiation and uptitration of optimal medical therapy including renin-angiotensin system inhibitor, beta-blocker, mineralocorticoid receptor antagonist, and sodium-glucose cotransporter 2 inhibitor in the early stage would prevent the progression and morbidity of HF. Concurrently, individualized surveillance to recognize and treat signs of disease progression is critical given the progressive nature of HF, even among stable patients on optimal therapy. However, there remains a wide variation in regional practice regarding the initiation, titration, and long-term monitoring of this therapy. To cover the differences in approaches toward HFrEF management and the implementation of guideline-based medical therapy, we discuss the current evidence in this arena, differences in present guideline recommendations, and compare practice patterns in Japan and the USA using a case of new-onset HF as an example. We will discuss pros and cons of the way HF is managed in each region, and highlight potential areas for improvement in care.

    View details for DOI 10.1016/j.jjcc.2023.07.019

    View details for PubMedID 37543194

  • Molecular characterization of a novel MYH7 mutation Q222H in a patient with severe dilated cardiomyopathy. Biophysical journal Kawana, M., Reddy Goluguri, R., Dawood, A., Spudich, J. A., Ruppel, K. 2023; 122 (3S1): 258a

    View details for DOI 10.1016/j.bpj.2022.11.1488

    View details for PubMedID 36783268

  • Molecular characterization of a novel MYH7 mutation Q222H in a patient with severe dilated cardiomyopathy Kawana, M., Goluguri, R., Dawood, A., Spudich, J. A., Ruppel, K. CELL PRESS. 2023: 258A
  • Severe Cardiovascular Complications Following Liver Transplantation in Patients With Iron Overload. JACC. Case reports Rhee, J., Zhang, S., Gallo, A., Ahmed, A., Kawana, M. 2022; 4 (11): 677-681

    Abstract

    We report 4 cases of our institutional experience with liver transplantation that illustrate the high risk of heart failure and cardiogenic shock in the setting of cardiac iron overload. We then discuss a pragmatic approach to assess the cardiovascular risk in liver transplantation candidates with cardiac iron overload. (Level of Difficulty: Advanced.).

    View details for DOI 10.1016/j.jaccas.2021.12.012

    View details for PubMedID 35677787

  • Multimorbidity, guideline-directed medical therapies, and associated outcomes among hospitalized heart failure patients. ESC heart failure Takeuchi, S., Kohno, T., Goda, A., Shiraishi, Y., Kawana, M., Saji, M., Nagatomo, Y., Nishihata, Y., Takei, M., Nakano, S., Soejima, K., Kohsaka, S., Yoshikawa, T., West Tokyo Heart Failure Registry Investigators 2022

    Abstract

    AIMS: Multimorbidity is common among heart failure (HF) patients and may attenuate guideline-directed medical therapy (GDMT). Multimorbid patients are under-represented in clinical trials; therefore, the effect of multimorbidity clustering on the prognosis of HF patients remains unknown. We evaluated the prevalence of multimorbidity clusters among consecutively registered hospitalized HF patients and assessed whether GDMT attenuated outcomes.METHODS AND RESULTS: We examined 1924 hospitalized HF patients with reduced left ventricular ejection fraction (<50%) in a multicentre registry (West Tokyo HF Registry: WET-HF). Ten comorbid conditions in the WET-HF were abstracted: coronary artery disease, atrial fibrillation, stroke, anaemia, chronic obstructive pulmonary disease, renal dysfunction, obesity, hypertension, dyslipidaemia, and diabetes. Patients were divided into three groups (0-2: n=451; 3-4: n=787; and ≥5: n=686) based on the number of comorbid conditions. The primary composite endpoint was all-cause mortality and HF rehospitalization. The most prevalent comorbidities were renal dysfunction (67.9%), hypertension (66.0%), and anaemia (53.8%). Increased comorbidity was associated with increased adverse outcomes [3-4: hazard ratio (HR) 1.42, 95% confidence interval (CI) 1.13-1.77, P=0.003; ≥5: HR 2.12, 95%CI 1.69-2.65, P<0.001; and reference: 0-2] and lower GDMT prescription rate (0-2: 69.2%; 3-4: 57.7%; and ≥5: 57.6%). GDMT was associated with decreased adverse outcomes; this association was maintained even as the comorbidity burden increased but tended to weaken (0-2: HR 0.53, 95%CI 0.35-0.78; P=0.001; 3-4: HR 0.82, 95%CI 0.65-1.04, P=0.095; and ≥5: HR 0.81, 95%CI 0.65-1.00, P=0.053; P for interaction=0.156).CONCLUSIONS: Comorbidity clusters were prevalent and associated with poorer outcomes. GDMT remained beneficial regardless of the comorbidity burden but tended to weaken with increasing comorbidity burden. Further research is required to optimize medical care in these patients.

    View details for DOI 10.1002/ehf2.13954

    View details for PubMedID 35561100

  • Effective Sphygmomanometer Based Non-invasive Central Venous Pressure Measurement In Hospitalized Heart Failure Patients Kawana, M., Cheng, P., Morimoto, H., Fowler, M. CHURCHILL LIVINGSTONE INC MEDICAL PUBLISHERS. 2022: S18
  • Hypertrophic cardiomyopathy: Mutations to mechanisms to therapies. Frontiers in physiology Kawana, M., Spudich, J. A., Ruppel, K. M. 2022; 13: 975076

    Abstract

    Hypertrophic cardiomyopathy (HCM) affects more than 1 in 500 people in the general population with an extensive burden of morbidity in the form of arrhythmia, heart failure, and sudden death. More than 25years since the discovery of the genetic underpinnings of HCM, the field has unveiled significant insights into the primary effects of these genetic mutations, especially for the myosin heavy chain gene, which is one of the most commonly mutated genes. Our group has studied the molecular effects of HCM mutations on human beta-cardiac myosin heavy chain using state-of-the-art biochemical and biophysical tools for the past 10years, combining insights from clinical genetics and structural analyses of cardiac myosin. The overarching hypothesis is that HCM-causing mutations in sarcomere proteins cause hypercontractility at the sarcomere level, and we have shown that an increase in the number of myosin molecules available for interaction with actin is a primary driver. Recently, two pharmaceutical companies have developed small molecule inhibitors of human cardiac myosin to counteract the molecular consequences of HCM pathogenesis. One of these inhibitors (mavacamten) has recently been approved by the FDA after completing a successful phase III trial in HCM patients, and the other (aficamten) is currently being evaluated in a phase III trial. Myosin inhibitors will be the first class of medication used to treat HCM that has both robust clinical trial evidence of efficacy and that targets the fundamental mechanism of HCM pathogenesis. The success of myosin inhibitors in HCM opens the door to finding other new drugs that target the sarcomere directly, as we learn more about the genetics and fundamental mechanisms of this disease.

    View details for DOI 10.3389/fphys.2022.975076

    View details for PubMedID 36225299

  • Assessment of Physical Activity Using Waist-Worn Accelerometers in Hospitalized Heart Failure Patients and Its Relationship with Kansas City Cardiomyopathy Questionnaire. Journal of clinical medicine Shiraishi, Y., Niimi, N., Goda, A., Takei, M., Kimura, T., Kohno, T., Kawana, M., Fukuda, K., Kohsaka, S. 2021; 10 (18)

    Abstract

    The health benefits of physical activity have been widely recognized, yet there is limited information on associations between accelerometer-related parameters and established patient-reported health status. This study investigated the association between the waist-worn accelerometer measurements, cardiopulmonary exercise testing (CPX), and results of the Kansas City Cardiomyopathy Questionnaire (KCCQ) in heart failure (HF) patients hospitalized for acute decompensation. A total of 31 patients were enrolled and wore a validated three-axis accelerometer for 2 weeks and completed the short version of the KCCQ after removing the device. Daily step counts, exercise time (metabolic equivalents × hours), and %sedentary time (sedentary time/device-equipped time) were measured. Among the measured parameters, the best correlation was observed between %sedentary time and the KCCQ overall and clinical summary scores (r = -0.65 and -0.65, each p < 0.001). All of the individual domains of the KCCQ (physical limitation, symptom frequency, and quality of life), with the exception of the social limitation domain, showed moderate correlations with %sedentary time. Finally, oxygen consumption assessed by CPX demonstrated only weak associations with the accelerometer-measured parameters. An accelerometer could complement the KCCQ results in accurately assessing the physical activity in HF patients immediately after hospitalization, albeit its correlation with CPX was at most moderate.

    View details for DOI 10.3390/jcm10184103

    View details for PubMedID 34575215

  • Time-sensitive approach in the management of acute heart failure. ESC heart failure Shiraishi, Y., Kawana, M., Nakata, J., Sato, N., Fukuda, K., Kohsaka, S. 2020

    Abstract

    Acute heart failure (AHF) has become a global public health burden largely because of the associated high morbidity, mortality, and cost. The treatment options for AHF have remained relatively unchanged over the past decades. Historically, clinical congestion alone has been considered the main target for treatment of acute decompensation in patients with AHF; however, this is an oversimplification of the complex pathophysiology. Within the similar clinical presentation of congestion, significant differences in pathophysiological mechanisms exist between the fluid accumulation and redistribution. Tissue hypoperfusion is another vital characteristic of AHF and should be promptly treated with appropriate interventions. In addition, recent clinical trials of novel therapeutic strategies have shown that heart failure management is 'time sensitive' and suggested that treatment selection based on individual aetiologies, triggers, and risk factor profiles could lead to better outcomes. In this review, we aim to describe the specifics of the 'time-sensitive' approach by the clinical phenotypes, for example, pulmonary/systemic congestion and tissue hypoperfusion, wherein patients are classified based on pathophysiological conditions. This mechanistic classification, in parallel with the comprehensive risk assessment, has become a cornerstone in the management of patients with AHF and thus supports effective decision making by clinicians. We will also highlight how therapeutic modalities should be individualized according to each clinical phenotype.

    View details for DOI 10.1002/ehf2.13139

    View details for PubMedID 33295126

  • Hospital meal intake in acute heart failure patients and its association with long-term outcomes. Open heart Yoshida, T. n., Shoji, S. n., Shiraishi, Y. n., Kawana, M. n., Kohno, T. n., Inoue, K. n., Fukuda, K. n., Heidenreich, P. A., Kohsaka, S. n. 2020; 7 (1)

    Abstract

    Risk prediction for hospitalised heart failure (HF, HHF) patients remains suboptimal. We aimed to determine the prognostic value of hospital food intake (FI) immediately before discharge among HHF patients.We analysed the data of 255 HHF patients extracted from the records of a single university hospital. The FI percentage of the three meals the day before hospital discharge was averaged. Patients were stratified into adequate FI (100% consumption) and inadequate FI (less than 100% consumption) groups. The primary outcome was the composite of all-cause mortality and/or HF readmission within 1 year.Only 49.3% of HHF patients consumed 100% of their meals. Patients with inadequate FI were older; predominantly women; and had a lower body mass index, higher brain natriuretic peptide levels and Clinical Frailty Scale scores at discharge than those with adequate FI. Inadequate FI was significantly associated with adverse outcomes after adjustments (HR 2.00; 95% CI 1.09 to 3.67; p=0.026). The effect of interaction by ejection fraction (EF) was highly significant: HF with preserved EF (≥40%) was significantly associated with inadequate FI with adverse outcomes (HR 4.95; 95% CI 1.71 to 14.36; p=0.003) but HF with reduced EF (<40%) was not (HR 0.77; 95% CI 0.31 to 1.95; p=0.590).The hospital FI assessment might be a simple, useful tool for predicting and stratifying risk for HHF patients.

    View details for DOI 10.1136/openhrt-2020-001248

    View details for PubMedID 32393659

  • Long-term outcome of orthotopic heart transplantation in Asians: An analysis of the United Network of Organ Sharing database. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation Kohsaka, S. n., Shudo, Y. n., Wang, H. n., Lingala, B. n., Kawana, M. n., Woo, Y. J. 2020

    View details for DOI 10.1016/j.healun.2020.07.016

    View details for PubMedID 32948416

  • Interpreting the evidence from tolvaptan clinical trials. Journal of cardiology Tanaka, T. n., Kawana, M. n., Kohsaka, S. n. 2019

    View details for DOI 10.1016/j.jjcc.2019.02.018

    View details for PubMedID 30948184

  • Physical therapy in successful venoarterial extracorporeal membrane oxygenation bridge to orthotopic heart transplantation. Journal of cardiac surgery Rinewalt, D. n., Shudo, Y. n., Kawana, M. n., Woo, Y. J. 2019

    Abstract

    Venoarterial extracorporeal membrane oxygenation (VA-ECMO) is a temporary mechanical circulatory support system that may be used as a lifesaving therapy for patients in acute heart failure and as a bridge to definitive management. Physical therapy in these patients remains challenging, with limited protocols to guide practitioners.We describe a case of a 37-year-old gentleman who presented with familial cardiomyopathy and cardiogenic shock.Our patient underwent urgent peripheral venoarterial extracorporeal membrane oxygenation (VA-ECMO) followed by successful heart transplantation. While on ECMO support he was enrolled in a physical therapy program that included the VitalGo Tilt Bed to improve lower body weight bearing while avoiding hip flexion and damage to the peripheral ECMO cannulae. The patient was discharged home expeditiously after heart transplant due to aggressive physical rehabilitation while on full VA-ECMO support.Early intensive physical rehabilitation is feasible and safe and may result in improved outcomes and expeditious discharge in VA ECMO patients. Protocol driven multidisciplinary physical therapy with a patient on femorally cannulated VA-ECMO retains the advantages of lower extremity peripheral cannulation while eliminating the risks of immobility. The new UNOS allocation system may result in a successful bridge to transplantation in patients on VA-ECMO due to the increased prioritization of this population to receive donor organs.

    View details for DOI 10.1111/jocs.14220

    View details for PubMedID 31441558

  • Dilated cardiomyopathy myosin mutants have reduced force-generating capacity JOURNAL OF BIOLOGICAL CHEMISTRY Ujfalusi, Z., Vera, C. D., Mijailovich, S. M., Svicevic, M., Yu, E., Kawana, M., Ruppel, K. M., Spudich, J. A., Geeves, M. A., Leinwand, L. A. 2018; 293 (23): 9017–29

    Abstract

    Dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) can cause arrhythmias, heart failure, and cardiac death. Here, we functionally characterized the motor domains of five DCM-causing mutations in human β-cardiac myosin. Kinetic analyses of the individual events in the ATPase cycle revealed that each mutation alters different steps in this cycle. For example, different mutations gave enhanced or reduced rate constants of ATP binding, ATP hydrolysis, or ADP release or exhibited altered ATP, ADP, or actin affinity. Local effects dominated, no common pattern accounted for the similar mutant phenotype, and there was no distinct set of changes that distinguished DCM mutations from previously analyzed HCM myosin mutations. That said, using our data to model the complete ATPase contraction cycle revealed additional critical insights. Four of the DCM mutations lowered the duty ratio (the ATPase cycle portion when myosin strongly binds actin) because of reduced occupancy of the force-holding A·M·D complex in the steady state. Under load, the A·M·D state is predicted to increase owing to a reduced rate constant for ADP release, and this effect was blunted for all five DCM mutations. We observed the opposite effects for two HCM mutations, namely R403Q and R453C. Moreover, the analysis predicted more economical use of ATP by the DCM mutants than by WT and the HCM mutants. Our findings indicate that DCM mutants have a deficit in force generation and force-holding capacity due to the reduced occupancy of the force-holding state.

    View details for PubMedID 29666183

    View details for PubMedCentralID PMC5995530

  • Controlling load-dependent kinetics of beta-cardiac myosin at the single-molecule level. Nature structural & molecular biology Liu, C., Kawana, M., Song, D., Ruppel, K. M., Spudich, J. A. 2018; 25 (6): 505–14

    Abstract

    Concepts in molecular tension sensing in biology are growing and have their origins in studies of muscle contraction. In the heart muscle, a key parameter of contractility is the detachment rate of myosin from actin, which determines the time that myosin is bound to actin in a force-producing state and, importantly, depends on the load (force) against which myosin works. Here we measure the detachment rate of single molecules of human beta-cardiac myosin and its load dependence. We find that both can be modulated by both small-molecule compounds and cardiomyopathy-causing mutations. Furthermore, effects of mutations can be reversed by introducing appropriate compounds. Our results suggest that activating versus inhibitory perturbations of cardiac myosin are discriminated by the aggregate result on duty ratio, average force, and ultimately average power output and suggest that cardiac contractility can be controlled by tuning the load-dependent kinetics of single myosin molecules.

    View details for PubMedID 29867217

  • Controlling Cardiac Contractility at the Single Molecule Level Liu, C., Song, D. L., Kawana, M., Ruppel, K. M., Spudich, J. A. CELL PRESS. 2018: 37A
  • Biophysical properties of human ß-cardiac myosin with converter mutations that cause hypertrophic cardiomyopathy. Science advances Kawana, M., Sarkar, S. S., Sutton, S., Ruppel, K. M., Spudich, J. A. 2017; 3 (2)

    Abstract

    Hypertrophic cardiomyopathy (HCM) affects 1 in 500 individuals and is an important cause of arrhythmias and heart failure. Clinically, HCM is characterized as causing hypercontractility, and therapies are aimed toward controlling the hyperactive physiology. Mutations in the β-cardiac myosin comprise ~40% of genetic mutations associated with HCM, and the converter domain of myosin is a hotspot for HCM-causing mutations; however, the underlying primary effects of these mutations on myosin's biomechanical function remain elusive. We hypothesize that these mutations affect the biomechanical properties of myosin, such as increasing its intrinsic force and/or its duty ratio and therefore the ensemble force of the sarcomere. Using recombinant human β-cardiac myosin, we characterize the molecular effects of three severe HCM-causing converter domain mutations: R719W, R723G, and G741R. Contrary to our hypothesis, the intrinsic forces of R719W and R723G mutant myosins are decreased compared to wild type and unchanged for G741R. Actin and regulated thin filament gliding velocities are ~15% faster for R719W and R723G myosins, whereas there is no change in velocity for G741R. Adenosine triphosphatase activities and the load-dependent velocity change profiles of all three mutant proteins are very similar to those of wild type. These results indicate that the net biomechanical properties of human β-cardiac myosin carrying these converter domain mutations are very similar to those of wild type or are even slightly hypocontractile, leading us to consider an alternative mechanism for the clinically observed hypercontractility. Future work includes how these mutations affect protein interactions within the sarcomere that increase the availability of myosin heads participating in force production.

    View details for DOI 10.1126/sciadv.1601959

    View details for PubMedID 28246639

    View details for PubMedCentralID PMC5302870

  • Acute Right Ventricular Failure After Successful Opening of Chronic Total Occlusion in Right Coronary Artery Caused by a Large Intramural Hematoma. Circulation. Cardiovascular interventions Kawana, M., Lee, A. M., Liang, D. H., Yeung, A. C. 2017; 10 (2)
  • Effects of hypertrophic and dilated cardiomyopathy mutations on power output by human beta-cardiac myosin JOURNAL OF EXPERIMENTAL BIOLOGY Spudich, J. A., Aksel, T., Bartholomew, S. R., Nag, S., Kawana, M., Yu, E. C., Sarkar, S. S., Sung, J., Sommese, R. F., Sutton, S., Cho, C., Adhikari, A. S., Taylor, R., Liu, C., Trivedi, D., Ruppel, K. M. 2016; 219 (2): 161-167

    Abstract

    Hypertrophic cardiomyopathy is the most frequently occurring inherited cardiovascular disease, with a prevalence of more than one in 500 individuals worldwide. Genetically acquired dilated cardiomyopathy is a related disease that is less prevalent. Both are caused by mutations in the genes encoding the fundamental force-generating protein machinery of the cardiac muscle sarcomere, including human β-cardiac myosin, the motor protein that powers ventricular contraction. Despite numerous studies, most performed with non-human or non-cardiac myosin, there is no clear consensus about the mechanism of action of these mutations on the function of human β-cardiac myosin. We are using a recombinantly expressed human β-cardiac myosin motor domain along with conventional and new methodologies to characterize the forces and velocities of the mutant myosins compared with wild type. Our studies are extending beyond myosin interactions with pure actin filaments to include the interaction of myosin with regulated actin filaments containing tropomyosin and troponin, the roles of regulatory light chain phosphorylation on the functions of the system, and the possible roles of myosin binding protein-C and titin, important regulatory components of both cardiac and skeletal muscles.

    View details for DOI 10.1242/jeb.125930

    View details for Web of Science ID 000368546300006

  • Effects of hypertrophic and dilated cardiomyopathy mutations on power output by human β-cardiac myosin. The Journal of experimental biology Spudich, J. A., Aksel, T., Bartholomew, S. R., Nag, S., Kawana, M., Yu, E. C., Sarkar, S. S., Sung, J., Sommese, R. F., Sutton, S., Cho, C., Adhikari, A. S., Taylor, R., Liu, C., Trivedi, D., Ruppel, K. M. 2016; 219 (Pt 2): 161-7

    Abstract

    Hypertrophic cardiomyopathy is the most frequently occurring inherited cardiovascular disease, with a prevalence of more than one in 500 individuals worldwide. Genetically acquired dilated cardiomyopathy is a related disease that is less prevalent. Both are caused by mutations in the genes encoding the fundamental force-generating protein machinery of the cardiac muscle sarcomere, including human β-cardiac myosin, the motor protein that powers ventricular contraction. Despite numerous studies, most performed with non-human or non-cardiac myosin, there is no clear consensus about the mechanism of action of these mutations on the function of human β-cardiac myosin. We are using a recombinantly expressed human β-cardiac myosin motor domain along with conventional and new methodologies to characterize the forces and velocities of the mutant myosins compared with wild type. Our studies are extending beyond myosin interactions with pure actin filaments to include the interaction of myosin with regulated actin filaments containing tropomyosin and troponin, the roles of regulatory light chain phosphorylation on the functions of the system, and the possible roles of myosin binding protein-C and titin, important regulatory components of both cardiac and skeletal muscles.

    View details for DOI 10.1242/jeb.125930

    View details for PubMedID 26792326

  • Understanding the Effects of Cardiomyopathy Causing Mutations on Human Beta Cardiac Myosin Biomechanical Function Nag, S., Sommese, R., Sung, J., Choe, E., Kawana, M., Cho, C., Taylor, R., Liu, C., Sutton, S., Ruppel, K., Spudich, J. CELL PRESS. 2014: 156A
  • Improved Loaded In Vitro Motility Assay and Actin Filament Tracking Software Delineates the Effect of Hypertrophic and Dilated Cardiomyopathy Mutations on the Power Output of Cardiac Myosin Aksel, T., Kawana, M., Adhikari, A., Sutton, S., Ruppel, K., Spudich, J. CELL PRESS. 2014: 562A
  • Quantification of gene transcripts with deep sequencing analysis of gene expression (DSAGE) using 1 to 2 µg total RNA. Current protocols in molecular biology Christodoulou, D. C., Gorham, J. M., Kawana, M., DePalma, S. R., Herman, D. S., Wakimoto, H. 2011; Chapter 25: Unit25B 9-?

    Abstract

    Deep sequencing analysis of gene expression (DSAGE) measures global gene transcript levels from only 1 to 2 µg total RNA by massively parallel sequencing of cDNA tags. This unit describes the construction of 21-bp cDNA tag libraries appropriate for massively parallel sequencing and analysis of the resulting sequence data. The adapter oligonucleotides used are optimized for sequencing with current Illumina massively parallel sequencers, and a step-by-step implementation of the analysis protocol is described. The expression profiles obtained are highly reproducible, enabling sensitive detection of differences between experimental conditions as well as assessment of the relative transcript abundance of different genes.

    View details for DOI 10.1002/0471142727.mb25b09s93

    View details for PubMedID 21225638

    View details for PubMedCentralID PMC4139004

  • Heterogeneous myocyte enhancer factor-2 (Mef2) activation in myocytes predicts focal scarring in hypertrophic cardiomyopathy PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Konno, T., Chen, D., Wang, L., Wakimoto, H., Teekakirikul, P., Nayor, M., Kawana, M., Eminaga, S., Gorham, J. M., Pandya, K., Smithies, O., Naya, F. J., Olson, E. N., Seidman, J. G., Seidman, C. E. 2010; 107 (42): 18097–102

    Abstract

    Unknown molecular responses to sarcomere protein gene mutations account for pathologic remodeling in hypertrophic cardiomyopathy (HCM), producing myocyte growth and increased cardiac fibrosis. To determine if hypertrophic signals activated myocyte enhancer factor-2 (Mef2), we studied mice carrying the HCM mutation, myosin heavy-chain Arg403Gln, (MHC(403/+)) and an Mef2-dependent β-galactosidase reporter transgene. In young, prehypertrophic MHC(403/+) mice the reporter was not activated. In hypertrophic hearts, activation of the Mef2-dependent reporter was remarkably heterogeneous and was observed consistently in myocytes that bordered fibrotic foci with necrotic cells, MHC(403/+) myocytes with Mef2-dependent reporter activation reexpressed the fetal myosin isoform (βMHC), a molecular marker of hypertrophy, although MHC(403/+) myocytes with or without βMHC expression were comparably enlarged over WT myocytes. To consider Mef2 roles in severe HCM, we studied homozygous MHC(403/403) mice, which have accelerated remodeling, widespread myocyte necrosis, and neonatal lethality. Levels of phosphorylated class II histone deacetylases that activate Mef2 were substantially increased in MHC(403/403) hearts, but Mef2-dependent reporter activation was patchy. Sequential analyses showed myocytes increased Mef2-dependent reporter activity before death. Our data dissociate myocyte hypertrophy, a consistent response in HCM, from heterogeneous Mef2 activation and reexpression of a fetal gene program. The temporal and spatial relationship of Mef2-dependent gene activation with myocyte necrosis and fibrosis in MHC(403/+) and MHC(403/403) hearts defines Mef2 activation as a molecular signature of stressed HCM myocytes that are poised to die.

    View details for PubMedID 20923879

  • Endogenous regulation of cardiovascular function by apelin-APJ AMERICAN JOURNAL OF PHYSIOLOGY-HEART AND CIRCULATORY PHYSIOLOGY Charo, D. N., Ho, M., Fajardo, G., Kawana, M., Kundu, R. K., Sheikh, A. Y., Finsterbach, T. P., Leeper, N. J., Ernst, K. V., Chen, M. M., Ho, Y. D., Chun, H. J., Bernstein, D., Ashley, E. A., Quertermous, T. 2009; 297 (5): H1904-H1913

    Abstract

    Studies have shown significant cardiovascular effects of exogenous apelin administration, including the potent activation of cardiac contraction. However, the role of the endogenous apelin-APJ pathway is less clear. To study the loss of endogenous apelin-APJ signaling, we generated mice lacking either the ligand (apelin) or the receptor (APJ). Apelin-deficient mice were viable, fertile, and showed normal development. In contrast, APJ-deficient mice were not born in the expected Mendelian ratio, and many showed cardiovascular developmental defects. Under basal conditions, both apelin and APJ null mice that survived to adulthood manifested modest decrements in contractile function. However, with exercise stress both mutant lines demonstrated consistent and striking decreases in exercise capacity. To explain these findings, we explored the role of autocrine signaling in vitro using field stimulation of isolated left ventricular cardiomyocytes lacking either apelin or APJ. Both groups manifested less sarcomeric shortening and impaired velocity of contraction and relaxation with no difference in calcium transient. Taken together, these results demonstrate that endogenous apelin-APJ signaling plays a modest role in maintaining basal cardiac function in adult mice with a more substantive role during conditions of stress. In addition, an autocrine pathway seems to exist in myocardial cells, the ablation of which reduces cellular contraction without change in calcium transient. Finally, differences in the developmental phenotype between apelin and APJ null mice suggest the possibility of undiscovered APJ ligands or ligand-independent effects of APJ.

    View details for DOI 10.1152/ajpheart.00686.2009

    View details for Web of Science ID 000271143400045

    View details for PubMedID 19767528

    View details for PubMedCentralID PMC2781363

  • PTC124 targets genetic disorders caused by nonsense mutations NATURE Welch, E. M., Barton, E. R., Zhuo, J., Tomizawa, Y., Friesen, W. J., Trifillis, P., Paushkin, S., Patel, M., Trotta, C. R., Hwang, S., Wilde, R. G., Karp, G., Takasugi, J., Chen, G., Jones, S., Ren, H., Moon, Y., Corson, D., Turpoff, A. A., Campbell, J. A., Conn, M., Khan, A., Almstead, N. G., Hedrick, J., Mollin, A., Risher, N., Weetall, M., Yeh, S., Branstrom, A. A., Colacino, J. M., Babiak, J., Ju, W. D., Hirawat, S., Northcutt, V. J., Miller, L. L., Spatrick, P., He, F., Kawana, M., Feng, H., Jacobson, A., Peltz, S. W., Sweeney, H. 2007; 447 (7140): 87–U6

    Abstract

    Nonsense mutations promote premature translational termination and cause anywhere from 5-70% of the individual cases of most inherited diseases. Studies on nonsense-mediated cystic fibrosis have indicated that boosting specific protein synthesis from <1% to as little as 5% of normal levels may greatly reduce the severity or eliminate the principal manifestations of disease. To address the need for a drug capable of suppressing premature termination, we identified PTC124-a new chemical entity that selectively induces ribosomal readthrough of premature but not normal termination codons. PTC124 activity, optimized using nonsense-containing reporters, promoted dystrophin production in primary muscle cells from humans and mdx mice expressing dystrophin nonsense alleles, and rescued striated muscle function in mdx mice within 2-8 weeks of drug exposure. PTC124 was well tolerated in animals at plasma exposures substantially in excess of those required for nonsense suppression. The selectivity of PTC124 for premature termination codons, its well characterized activity profile, oral bioavailability and pharmacological properties indicate that this drug may have broad clinical potential for the treatment of a large group of genetic disorders with limited or no therapeutic options.

    View details for DOI 10.1038/nature05756

    View details for Web of Science ID 000246149300049

    View details for PubMedID 17450125

  • Systemic administration of L-arginine benefits mdx skeletal muscle function MUSCLE & NERVE Barton, E. R., Morris, L., Kawana, M., Bish, L. T., Toursel, T. 2005; 32 (6): 751–60

    Abstract

    A major consequence of muscular dystrophy is that increased membrane fragility leads to high calcium influx and results in muscle degeneration and myonecrosis. Prior reports have demonstrated that increased nitric oxide production via L-arginine treatment of normal and mdx mice resulted in increased expression of utrophin and increased activation of muscle satellite cells, which could ameliorate the dystrophic pathology. We delivered L-arginine to normal and mdx mice, and examined muscles for any functional changes associated with its administration. Treated mdx muscles were less susceptible to contraction-induced damage and exhibited a rightward shift of the force-frequency relationship. Immunoblotting revealed increases in utrophin and gamma-sarcoglycan in the treated muscles. There was also a decrease in Evans blue dye uptake, indicating a reduction in myonecrosis. However, there was no decrease in serum creatine kinase or the proportion of central nuclei, nor any improvement in specific force. Together, these results show that L-arginine treatment can be beneficial to mdx muscle function, perhaps through a combination of enhanced calcium handling and increased utrophin, thereby decreasing muscle degeneration.

    View details for PubMedID 16116642

  • gamma-sarcoglycan deficiency increases cell contractility, apoptosis and MAPK pathway activation but does not affect adhesion JOURNAL OF CELL SCIENCE Griffin, M. A., Feng, H. S., Tewari, M., Acosta, P., Kawana, M., Sweeney, H. L., Discher, D. E. 2005; 118 (7): 1405–16

    Abstract

    The functions of gamma-sarcoglycan (gammaSG) in normal myotubes are largely unknown, however gammaSG is known to assemble into a key membrane complex with dystroglycan and its deficiency is one known cause of limb-girdle muscular dystrophy. Previous findings of apoptosis from gammaSG-deficient mice are extended here to cell culture where apoptosis is seen to increase more than tenfold in gammaSG-deficient myotubes compared with normal cells. The deficient myotubes also exhibit an increased contractile prestress that results in greater shortening and widening when the cells are either lightly detached or self-detached. However, micropipette-forced peeling of single myotubes revealed no significant difference in cell adhesion. Consistent with a more contractile phenotype, acto-myosin striations were more prominent in gammaSG-deficient myotubes than in normal cells. An initial phosphoscreen of more than 12 signaling proteins revealed a number of differences between normal and gammaSG(-/-) muscle, both before and after stretching. MAPK-pathway proteins displayed the largest changes in activation, although significant phosphorylation also appeared for other proteins linked to hypertension. We conclude that gammaSG normally moderates contractile prestress in skeletal muscle, and we propose a role for gammaSG in membrane-based signaling of the effects of prestress and sarcomerogenesis.

    View details for PubMedID 15769854