1
|
Arjun K, Inbaraj G, Meghana A, Preethish-Kumar V, John AP, Polavarapu K, B S KP, Nandeesh BN, Nandan C, Kramer BW, Steinbusch HWM, Nalini A, Udupa K, Sathyaprabha TN. Cardiac dysregulation in Duchenne muscular dystrophy: An ECG analysis. J Electrocardiol 2025; 91:154015. [PMID: 40339309 DOI: 10.1016/j.jelectrocard.2025.154015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND AND OBJECTIVE Duchenne Muscular Dystrophy (DMD) is a progressive X-linked recessive disorder characterized by severe muscle degeneration and premature death, often due to cardiac complications. Despite the high prevalence of arrhythmogenic cardiomyopathy in DMD, the utility of Electrocardiogram (ECG) analysis in detecting subclinical cardiac dysregulation remains underexplored. This study aimed to investigate alterations in Lead II ECG parameters in children with DMD, potentially indicating an elevated risk of Sudden Cardiac Death (SCD). METHODS In this cross-sectional study, Lead II ECG recordings from 54 genetically confirmed DMD patients were compared against 31 age-matched healthy-controls. Parameters analyzed included PR interval, QRS duration, QT and QTc intervals, Tp-Te interval, and amplitudes of P, Q, R, S, and T waves. Analysis was conducted using LabChart Pro 8 software and the Hamilton-Tompkins QRS detection algorithm. Heart-rate-corrected QT interval (QTc) was calculated using Bazett's formula (QTc = QT/√RR). An independent samples t-test with a significance level of p < 0.05 was used for comparisons between groups. RESULTS The study revealed significant ECG alterations in the DMD group compared to controls, included a reduced PR interval, prolonged QRS and QT intervals, decreased QTc, and increased Tp-Te interval. Additionally, significant increases in P, Q, R wave amplitudes, and ST height were observed, indicative of atrial hypertrophy and potential ventricular arrhythmias. CONCLUSIONS Lead II ECG analysis in children with DMD demonstrates critical alterations suggestive of subclinical cardiac dysregulation, highlighting a potential non-invasive marker for early detection of cardiac involvement. These findings emphasize the importance of regular cardiac monitoring in DMD patients to mitigate SCD risk through timely interventions and underscore the need for further research into the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Krishnamurthy Arjun
- Department of Computer Science and Engineering (CSE), School of Engineering, Dayananda Sagar University, Bangalore, India
| | - Ganagarajan Inbaraj
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Adoor Meghana
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | - Anu P John
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Krishna Prasad B S
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - B N Nandeesh
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Chandregowda Nandan
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Boris W Kramer
- Department of Neonatology, Poznan University of Medical Sciences, Poland
| | - Harry W M Steinbusch
- Department of Cellular and Translational Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Kaviraja Udupa
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Talakad N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India.
| |
Collapse
|
2
|
Barthélémy I, Su JB, Cauchois X, Relaix F, Ghaleh B, Blot S. Ambulatory electrocardiographic longitudinal monitoring in a canine model for Duchenne muscular dystrophy identifies decreased very low frequency power as a hallmark of impaired heart rate variability. Sci Rep 2024; 14:8969. [PMID: 38637619 PMCID: PMC11026469 DOI: 10.1038/s41598-024-59196-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) patients exhibit a late left ventricular systolic dysfunction preceded by an occult phase, during which myocardial fibrosis progresses and some early functional impairments can be detected. These latter include electrocardiographic (ECG) and heart rate variability (HRV) abnormalities. This longitudinal study aimed at describing the sequence of ECG and HRV abnormalities, using Holter ECG in the GRMD (Golden retriever muscular dystrophy) dog model, known to develop a DMD-like disease, including cardiomyopathy. Most of the known ECG abnormalities described in DMD patients were also found in GRMD dogs, including increased heart rate, prolonged QT and shortened PR intervals, ventricular arrhythmias, and several of them could be detected months before the decrease of fractional shortening. The HRV was impaired like in DMD patients, one of the earliest evidenced abnormalities being a decrease in the very low frequency (VLF) component of the power spectrum. This decrease was correlated with the further reduction of fractional shortening. Such decreased VLF probably reflects impaired autonomic function and abnormal vasomotor tone. This study provides new insights into the knowledge of the GRMD dog model and DMD cardiomyopathy and emphasizes the interest to monitor the VLF power in DMD patients, still unexplored in this disease, whilst it is highly predictive of deleterious clinical events in many other pathological conditions.
Collapse
Affiliation(s)
- Inès Barthélémy
- "Biology of the Neuromuscular System" Team, U955 IMRB, INSERM, Univ Paris-Est Créteil, 94010, Créteil, France.
- École Nationale Vétérinaire d'Alfort, IMRB, 7 Avenue du Général de Gaulle, 94700, Maisons-Alfort, France.
| | - Jin Bo Su
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Xavier Cauchois
- "Biology of the Neuromuscular System" Team, U955 IMRB, INSERM, Univ Paris-Est Créteil, 94010, Créteil, France
- École Nationale Vétérinaire d'Alfort, IMRB, 7 Avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Frédéric Relaix
- "Biology of the Neuromuscular System" Team, U955 IMRB, INSERM, Univ Paris-Est Créteil, 94010, Créteil, France
- École Nationale Vétérinaire d'Alfort, IMRB, 7 Avenue du Général de Gaulle, 94700, Maisons-Alfort, France
| | - Bijan Ghaleh
- Inserm U955-IMRB, UPEC, Ecole Nationale Vétérinaire d'Alfort, Créteil, France
| | - Stéphane Blot
- "Biology of the Neuromuscular System" Team, U955 IMRB, INSERM, Univ Paris-Est Créteil, 94010, Créteil, France.
- École Nationale Vétérinaire d'Alfort, IMRB, 7 Avenue du Général de Gaulle, 94700, Maisons-Alfort, France.
| |
Collapse
|
3
|
Silva RMFLD, Monteze NM, Giannetti JG, Meira ZMA. Electrocardiographic and Autonomic Nervous System Changes after Changes in the Posture of Children and Adolescents with Duchenne Muscular Dystrophy. Arq Bras Cardiol 2024; 121:e20230483. [PMID: 38597534 DOI: 10.36660/abc.20230483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/13/2023] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a rare inherited neuromuscular disease. At first, cardiac involvement may be asymptomatic. Therefore, assessing patients using non-invasive methods can help detect any changes. OBJECTIVES Analyze the electrocardiogram (ECG) test and heart rate variability (HRV) of the DMD group and compare the information with that of the age-matched control group. METHODS A prospective study with 27 male patients with DMD (11.9 years old), who underwent clinical evaluation, ECG, echocardiogram, and Holter monitoring. ECG (200% increase) was assessed by two independent observers. HRV was measured over time (24 h) and in the frequency domain, in the supine and sitting positions. The healthy group consisted of nine patients (11.0 years old). A value of p < 0.05 was considered statistically significant. RESULTS The mean ejection fraction (EF) was 60% (34 to 71%). The Kappa coefficient for ECG measurements ranged from 0.64 to 1.00. An increase in the R/S ratio in V1 was observed in 25.9% of the subjects, pathological Q wave in 29.6%, and fragmented QRS in 22.2% in inferior/high lateral regions, with a negative correlation with EF (p = 0.006). There was low HRV, without the influence of any variable, including treatment. With the change in position, there was an increase in HR (p = 0.004), but there was no change in HRV. The LF/HF ratio was 2.7 in the DMD group and 0.7 in the control group (p = 0.002). CONCLUSIONS In DMD subjects, prominent R waves in V1 and changes in the inferior/high lateral regions occurred in almost 30% of the cases. Lower vagal tone was observed without the influence of the variables age, ejection fraction, QT dispersion, and treatment. Despite the increase in HR, there was no adequate HRV response to the change in position.
Collapse
Affiliation(s)
| | - Nathalia Mussi Monteze
- Universidade Federal de Minas Gerais - Faculdade de Medicina, Belo Horizonte, MG - Brasil
| | | | - Zilda Maria Alves Meira
- Hospital das Clínicas da Universidade Federal de Minas Gerais - Cardiologia Pediátrica, Belo Horizonte, MG - Brasil
| |
Collapse
|
4
|
Nakamura A, Matsumura T, Takeshima Y, Kuru S, Imazaki M, Nonomura H, Kaiya H. The Association Between Physical Activity/Heart Rate Variability Data Obtained Using a Wearable Device and Timed Motor Functional Tests in Patients with Duchenne Muscular Dystrophy: A Pilot Study. J Neuromuscul Dis 2024; 11:715-724. [PMID: 38607760 DOI: 10.3233/jnd-230142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) is a devastating X-linked muscle disease. Clinical evaluation of DMD uses patient-intensive motor function tests, and the recent development of wearable devices allows the collection of a variety of biometric information, including physical activity. Objective In this study, we examined differences in physical activity and heart rate variability (HRV) between patients with DMD and healthy subjects using a wearable device, and investigated any association between these parameters and motor function in patients with DMD. Methods Participants were 7 patients with DMD and 8 healthy males, whose physical activity and HRV were provided by a wearable device. These data were used to investigate the relationship between both physical activity and HRV parameters and timed motor functional tests [Time to stand from supine, 10-meter walking time (10MWT), North Star Ambulatory Assessment (NSAA), and 6-minute walking test (6MWT)] in patients with DMD. Results Results of 24-hours physical activity, fat burning, total number of steps and active distance, average step rate, average exercise intensity during walking, exercise, degree of forward lean during walking, maximum heart rate, normalized low frequency power (LF norm), and maximum exercise intensity in patients with DMD were lower than those in control subjects. Physical activity and HRV parameters did not correlate with the time to stand from supine. The 10MWT positively correlated with average heart rate, while NSAA negatively correlated with average heart rate, total frequency power (TF), and very low frequency power (VLF) during arousal. The 6MWT negatively correlated with ratio LF/high frequency power (HF). CONCLUSIONS Physical activity and HRV indices that differ from those of normal children and that correlate with motor function assessment may serve as digital biomarkers.
Collapse
Affiliation(s)
- Akinori Nakamura
- Department of Clinical Research, NHO Matsumoto Medical Center, Matsumoto, Japan
| | - Tsuyoshi Matsumura
- Department of Neurology, NHO Osaka Toneyama Medical Center, Toneyama, Osaka, Japan
| | - Yasuhiro Takeshima
- Department of Pediatrics, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Satoshi Kuru
- Department of Neurology, NHO Suzuka National Hospital, Suzuka, Japan
| | - Manami Imazaki
- Takeda Development Center Japan, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Hidenori Nonomura
- Takeda Development Center Japan, Takeda Pharmaceutical Company Limited, Osaka, Japan
| | - Hisanobu Kaiya
- Clinical Trials Accelerating Organization, Japan Muscular Dystrophy Association, Tokyo, Japan
| |
Collapse
|
5
|
Nayak A, S B A, Bardhan M, Rashmi R, Arunachal G, Prathyusha P, Nalini A, Sathyaprabha T, Udupa K. Evaluation of Cardiac, Autonomic Functions in Ambulant Patients with Duchenne Muscular Dystrophy. SN COMPREHENSIVE CLINICAL MEDICINE 2023; 5:138. [PMID: 37193318 PMCID: PMC10160717 DOI: 10.1007/s42399-023-01473-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 05/18/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder caused by dystrophin gene mutation resulting in muscle weakness, motor delays, difficulty in standing, and inability to walk by 12 years. As disease progresses, it leads to cardiac and respiratory failure. Evaluation of cardiac autonomic status and echocardiography in DMD patients at a young age can be a potential biomarker to assess disease progression. This study aimed to investigate the younger DMD population of 5-11years of age with mild to moderate cardiac involvement for early detection using non-invasive and cost-effective tools. Genetically confirmed male DMD patients, aged 5-11 years (n = 47), screened from the outpatient department of a tertiary neuroscience institution were subjected to heart rate variability and echocardiographic analysis, and values were correlated with their clinical variables. DMD patients showed a significantly higher difference in HR, interventricular septum, E m/s, and E-wave to A-wave (E/A) ratio than normal values (p < 0.001). Significantly higher HR indicates initial sinus tachycardia and decreased IVD (d), and increased E m/s and E/A ratio mark the onset of cardiac symptoms in DMD patients even though its chamber dimension remains normal and are associated with cardiac muscle fibrosis.
Collapse
Affiliation(s)
- Amritharekha Nayak
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Apoorva S B
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Mainak Bardhan
- Department of Neurology, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - R. Rashmi
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - G. Arunachal
- Department of Human Genetics, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - P.V. Prathyusha
- Department of Biostatistics, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health And Neurosciences (NIMHANS), Bengaluru, 560029 India
| | - T.N. Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| | - Kaviraja Udupa
- Department of Neurophysiology, National Institute of Mental Health And Neurosciences (NIMHANS), Hosur Road, Bengaluru, 560029 India
| |
Collapse
|
6
|
Inbaraj G, Arjun K, Meghana A, Preethish-Kumar V, John AP, Polavarapu K, Nashi S, Sekar D, Udupa K, Prathuysha PV, Prasad K, Bardhan M, Raju TR, Kramer BW, Nalini A, Sathyaprabha TN. Neuro-Cardio-Autonomic Modulations in Children with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2023; 10:227-238. [PMID: 36847014 DOI: 10.3233/jnd-221621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
BACKGROUND AND OBJECTIVE Duchenne muscular dystrophy (DMD) is a degenerative X-linked muscle disease. Death frequently results from complications in cardiopulmonary systems. Preclinical/early diagnosis of cardiac autonomic abnormalities may aid initiate cardioprotective therapy and enhance prognosis. METHODS A cross sectional, prospective study of 38 DMD boys compared with 37 age-matched healthy controls was conducted. Lead II electrocardiography and beat-to-beat blood pressure were recorded to assess heart rate variability (HRV), blood pressure variability (BPV), and baroreceptor sensitivity (BRS) in a standardized environment. Data were analysed and correlated with disease severity and genotype. RESULTS In the DMD group, the median age at assessment was 8 years [IQR 7-9 years], the median age at disease onset was 3 years [IQR, 2-6 years], and the mean duration of illness was 4 years [IQR, 2.5-5]. DNA sequencing showed deletions in 34/38 (89.5 %) and duplications in 4/38 (10.5%) patients. The median heart rate in DMD children was significantly higher [101.19 (Range, 94.71-108.49)] /min compared to controls [81 (Range, 76.2-92.76)] /min (p < 0.05). All the assessed HRV and BPV parameters were significantly impaired in DMD cases except for the coefficient of variance of systolic blood pressure. Further, BRS parameters were also significantly reduced in DMD, excluding alpha-LF. A positive correlation was found between alpha HF with age at onset and duration of illness. CONCLUSION This study demonstrates a distinct early impairment of neuro-cardio-autonomic regulation in DMD. Simple yet effective non-invasive techniques such as HRV, BPV, and BRS may help identify cardiac dysfunction in a pre-clinical state, paving the way for early cardio-protective therapies and limiting disease progression in DMD patients.
Collapse
Affiliation(s)
- Ganagarajan Inbaraj
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Krishnamurthy Arjun
- Department of CSE, School of Engineering, Dayananda Sagar University, Bangalore
| | - Adoor Meghana
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | | | - Anu P John
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Deepha Sekar
- Department of Molecular Genetics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Kaviraja Udupa
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Parthipulli V Prathuysha
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Krishna Prasad
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Mainak Bardhan
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Boris W Kramer
- Department of Paediatrics, School of Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bangalore, India
| | - Talakad N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bangalore, India
| |
Collapse
|
7
|
Woods WA, Harmon WG, Webb LW, Robinson GG, McCulloch MA. Emergency department care of patients with Duchenne muscular dystrophy. Am J Emerg Med 2022; 60:101-105. [PMID: 35933945 DOI: 10.1016/j.ajem.2022.07.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022] Open
Abstract
Patients with Duchenne muscular dystrophy are living longer and are increasingly seen in Emergency Departments. Though the most common cause of death remains progressive respiratory failure, increased life expectancies have unmasked the significance of progressive myocardial dysfunction, now associated with nearly 40% of mortalities in the DMD population. Cardiac complications such as arrhythmias and cardiomyopathy are becoming ever more widely recognized. Emergency physicians may encounter DMD patients with untreated, undiagnosed or worsening of known heart disease. This review will initially familiarize the emergency physician with the pathophysiology and lifetime trajectory of care for these patients before describing specific emergency department evaluation and treatment.
Collapse
Affiliation(s)
- William A Woods
- Department of Emergency Medicine, University of Virginia, Charlottesville, VA, United States of America; Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America.
| | - William G Harmon
- Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America
| | - Lauren W Webb
- Department of Emergency Medicine, University of Virginia, Charlottesville, VA, United States of America
| | - Grant G Robinson
- Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America
| | - Michael A McCulloch
- Department of Pediatrics, University of Virginia, Charlottesville, VA, United States of America
| |
Collapse
|
8
|
Tang L, Shao S, Wang C. Electrocardiographic features of children with Duchenne muscular dystrophy. Orphanet J Rare Dis 2022; 17:320. [PMID: 35987773 PMCID: PMC9392256 DOI: 10.1186/s13023-022-02473-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a clinically common X-linked recessive myopathy, which is caused by mutation of the gene encoding dystrophin on chromosome Xp21. The onset of heart injury in children with DMD is inconspicuous, and the prognosis is poor once it develops to the stage of heart failure. Cardiovascular complications remain an important cause of death in this patient population. At present, population and animal studies have suggested that Electrocardiogram (ECG) changes may be the initial manifestation of cardiac involvement in children with DMD. Relevant clinical studies have also confirmed that significant abnormal ECG changes already exist in DMD patients before cardiomegaly and/or LVEF decrease. With increases in age and decreases in cardiac function, the proportion of ECG abnormalities in DMD patients increase significantly. Some characteristic ECG changes, such as ST-segment changes, T wave inversion, Q wave at the inferolateral leads, LBBB and SDANN, have a certain correlation with the indexes of cardiac remodeling or impaired cardiac function in DMD patients, while VT and LBBB have demonstrated relatively good predictive value for the occurrence of long-term DCM and/or adverse cardiovascular events or even death in DMD patients. The present review discusses the electrocardiographic features in children with DMD.
Collapse
|
9
|
Li J, Wang K, Zhang Y, Qi T, Yuan J, Zhang L, Qiu H, Wang J, Yang HT, Dai Y, Song Y, Chang X. Therapeutic Exon Skipping Through a CRISPR-Guided Cytidine Deaminase Rescues Dystrophic Cardiomyopathy in Vivo. Circulation 2021; 144:1760-1776. [PMID: 34698513 DOI: 10.1161/circulationaha.121.054628] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Loss of dystrophin protein causes Duchenne muscular dystrophy (DMD), characterized by progressive degeneration of cardiac and skeletal muscles, and mortality in adolescence or young adulthood. Although cardiac failure has risen as the leading cause of mortality in patients with DMD, effective therapeutic interventions remain underdeveloped, in part, because of the lack of a suitable preclinical model. METHODS We analyzed a novel murine model of DMD created by introducing a 4-bp deletion into exon 4, one of the exons encoding the actin-binding domain 1 of dystrophin (referred to as DmdE4* mice). Echocardiography, microcomputed tomography, muscle force measurement, and histological analysis were performed to determine cardiac and skeletal muscle defects in these mice. Using this model, we examined the feasibility of using a cytidine base editor to install exon skipping and rescue dystrophic cardiomyopathy in vivo. AAV9-based CRISPR/Cas9-AID (eTAM) together with AAV9-sgRNA was injected into neonatal DmdE4* mice, which were analyzed 2 or 12 months after treatment to evaluate the extent of exon skipping, dystrophin restoration, and phenotypic improvements of cardiac and skeletal muscles. RESULTS DmdE4* mice recapitulated many aspects of human DMD, including shortened life span (by ≈50%), progressive cardiomyopathy, kyphosis, profound loss of muscle strength, and myocyte degeneration. A single-dose administration of AAV9-eTAM instituted >50% targeted exon skipping in the Dmd transcripts and restored up to 90% dystrophin in the heart. As a result, early ventricular remodeling was prevented and cardiac and skeletal muscle functions were improved, leading to an increased life span of the DmdE4* mice. Despite gradual decline of AAV vector and base editor expression, dystrophin restoration and pathophysiological rescue of muscular dystrophy were long lasted for at least 1 year. CONCLUSIONS Our study demonstrates the feasibility and efficacy to institute exon skipping through an enhanced TAM (eTAM) for therapeutic application(s).
Collapse
Affiliation(s)
- Jia Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Chinese Academy of Sciences, China. Joint Research Center of Hangzhou First Hospital Group and Westlake University, Zhejiang, China (J.L., K.W., Y.Z., T.Q., X.C.)
| | - Kaiying Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Chinese Academy of Sciences, China. Joint Research Center of Hangzhou First Hospital Group and Westlake University, Zhejiang, China (J.L., K.W., Y.Z., T.Q., X.C.)
| | - Yuchen Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Chinese Academy of Sciences, China. Joint Research Center of Hangzhou First Hospital Group and Westlake University, Zhejiang, China (J.L., K.W., Y.Z., T.Q., X.C.)
| | - Tuan Qi
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Shanghai Jiao Tong University School of Medicine (SJTUSM), China (T.Q.).,Chinese Academy of Sciences, China. Joint Research Center of Hangzhou First Hospital Group and Westlake University, Zhejiang, China (J.L., K.W., Y.Z., T.Q., X.C.).,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.)
| | - Juanjuan Yuan
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan City, Guangdong Province, China (J.Y., H.Q.)
| | - Lei Zhang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.)
| | - Han Qiu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan City, Guangdong Province, China (J.Y., H.Q.).,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (H.Q., J.W., H.-T.Y.).,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.)
| | - Jinxi Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health (J.W., H.-T.Y.).,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (H.Q., J.W., H.-T.Y.)
| | - Huang-Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology, Shanghai Institute of Nutrition and Health (J.W., H.-T.Y.).,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (H.Q., J.W., H.-T.Y.)
| | - Yi Dai
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China (Y.D.)
| | - Yan Song
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla (Y.S.)
| | - Xing Chang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.).,Chinese Academy of Sciences, China. Joint Research Center of Hangzhou First Hospital Group and Westlake University, Zhejiang, China (J.L., K.W., Y.Z., T.Q., X.C.).,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China (J.L., K.W., Y.Z., T.Q., L.Z., H.Q., X.C.)
| |
Collapse
|
10
|
Rodrigues MV, Stoco-Oliveira MC, Silva TDD, Ferreira C, Valente HB, Vanzella LM, Vanderlei LCM. Autonomic modulation at rest and in response to postural change in adolescents with Duchenne muscular dystrophy: a cross-sectional study. ARQUIVOS DE NEURO-PSIQUIATRIA 2021; 79:766-773. [PMID: 34669819 DOI: 10.1590/0004-282x-anp-2020-0458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 01/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Analysis of autonomic modulation after postural change may inform the prognosis and guide treatment in different populations. However, this has been insufficiently explored among adolescents with Duchenne muscular dystrophy (DMD). OBJECTIVE To investigate autonomic modulation at rest and in response to an active sitting test (AST) among adolescents with DMD. METHODS Fifty-nine adolescents were included in the study and divided into two groups: 1) DMD group: adolescents diagnosed with DMD; 2) control group (CG): healthy adolescents. Participants' weight and height were assessed. Lower limb function, motor limitations and functional abilities of the participants in the DMD group were classified using the Vignos scale, Egen classification and motor function measurement, respectively. The following variables were assessed before, during and after AST: systolic blood pressure (SBP), diastolic blood pressure (DBP), respiratory rate (f), oxygen saturation and heart rate (HR). To analyze the autonomic modulation, the HR was recorded beat-by-beat. Heart rate variability (HRV) indices were calculated in the time and frequency domains. RESULTS Differences in relation to groups were observed for all HRV indices, except LF/HF, oxygen saturation, HR and f (p < 0.05). Differences in relation to time and the interaction effect between group and time were observed for RMSSD, SD1, SD2, SD1/SD2, LFms2 and LFnu, HFun, SBP and DBP (p < 0.05). Differences in relation to time were also observed for the indice SDNN, FC and f (p < 0.05). CONCLUSIONS Performing the AST promoted reduced autonomic modulation and increased SBP, DBP and HR in adolescents with DMD.
Collapse
Affiliation(s)
- Mariana Viana Rodrigues
- Universidade Estadual Paulista, Faculdade de Ciências e Tecnologia, Presidente Prudente SP, Brazil
| | | | - Talita Dias da Silva
- Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo SP, Brazil
| | - Celso Ferreira
- Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo SP, Brazil
| | - Heloisa Balotari Valente
- Universidade Estadual Paulista, Faculdade de Ciências e Tecnologia, Presidente Prudente SP, Brazil
| | - Laís Manata Vanzella
- Universidade Estadual Paulista, Faculdade de Ciências e Tecnologia, Presidente Prudente SP, Brazil
| | | |
Collapse
|
11
|
Dias RM, Hoshi RA, Vanderlei LCM, Monteiro CBDM, Alvarez MPB, Crocetta TB, Grossklauss LF, Fernani DCGL, Dantas MTAP, Martins FPA, Garner DM, Abreu LC, Ferreira C, da Silva TD. Influence of Different Types of Corticosteroids on Heart Rate Variability of Individuals with Duchenne Muscular Dystrophy-A Pilot Cross Sectional Study. Life (Basel) 2021; 11:752. [PMID: 34440496 PMCID: PMC8398672 DOI: 10.3390/life11080752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 11/18/2022] Open
Abstract
Individuals with Duchenne Muscular Dystrophy (DMD) have an impairment of cardiac autonomic function categorized by parasympathetic reduction and sympathetic predominance. The objective of this study was to assess the cardiac autonomic modulation of individuals with DMD undergoing therapy with Prednisone/Prednisolone and Deflazacort and compare with individuals with DMD without the use of these medications and a typically developed control group. Methods: A cross-sectional study was completed, wherein 40 boys were evaluated. The four treatment groups were: Deflazacort; Prednisone/Prednisolone; no corticoid use; and typical development. Heart Rate Variability (HRV) was investigated via linear indices (Time Domain and Frequency Domain) and non-linear indices Results: The results of this study revealed that individuals with DMD undertaking pharmacotherapies with Prednisolone demonstrated HRV comparable to the Control Typically Developed (CTD) group. In contrast, individuals with DMD undergoing pharmacotherapies with Deflazacort achieved lower HRV, akin to individuals with DMD without any medications, as demonstrated in the metrics: RMSSD; LF (n.u.), HF (n.u.), LF/HF; SD1, α1, and α1/α2, and a significant effect for SD1/SD2; %DET and Ratio; Shannon Entropy, 0 V%, 2 LV% and 2 ULV%. Conclusions: Corticosteroids have the potential to affect the cardiac autonomic modulation in adolescents with DMD. The use of Prednisone/Prednisolone appears to promote improved responses in terms of sympathovagal activity as opposed to Deflazacort.
Collapse
Affiliation(s)
- Rodrigo Martins Dias
- Postgraduate Program in Medicine (Cardiology) at Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04024-002, Brazil; (C.F.); (T.D.d.S.)
| | | | | | - Carlos Bandeira de Mello Monteiro
- Postgraduate Program in Rehabilitation Sciences, Faculty of Medicine, University of São Paulo (FMUSP), São Paulo 05360-160, Brazil; (C.B.d.M.M.); (M.P.B.A.)
| | - Mayra Priscila Boscolo Alvarez
- Postgraduate Program in Rehabilitation Sciences, Faculty of Medicine, University of São Paulo (FMUSP), São Paulo 05360-160, Brazil; (C.B.d.M.M.); (M.P.B.A.)
- Department of Health Sciences, Anhanguera College- Campus of Jundiaí, Jundiaí 13209-355, Brazil
| | - Tânia Brusque Crocetta
- Laboratório de Psicologia do Esporte e do Exercício, Centro de Ciências da Saúde e do Esporte, Universidade do Estado de Santa Catarina, Florianópolis 88035-001, Brazil;
| | - Luis Fernando Grossklauss
- Department of Neurology/Neurosurgery, Neuropediatrist at the Federal University of São Paulo, São Paulo 04039-002, Brazil;
| | | | - Maria Tereza Artero Prado Dantas
- Department of Health Sciences, University of Western Paulista (UNOESTE), Presidente Prudente 19050-920, Brazil; (D.C.G.L.F.); (M.T.A.P.D.)
| | | | - David M. Garner
- Cardiorespiratory Research Group, Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Headington Campus, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK;
| | - Luiz Carlos Abreu
- Department of Integrated Health Education, Federal University of Espírito Santo (UFES), Vitória 29040-090, Brazil;
| | - Celso Ferreira
- Postgraduate Program in Medicine (Cardiology) at Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04024-002, Brazil; (C.F.); (T.D.d.S.)
| | - Talita Dias da Silva
- Postgraduate Program in Medicine (Cardiology) at Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), São Paulo 04024-002, Brazil; (C.F.); (T.D.d.S.)
- Postgraduate Program in Rehabilitation Sciences, Faculty of Medicine, University of São Paulo (FMUSP), São Paulo 05360-160, Brazil; (C.B.d.M.M.); (M.P.B.A.)
- Faculty of Medicine, University of Sao Paulo City (UNICID), São Paulo 03071-000, Brazil
| |
Collapse
|
12
|
Mekies LN, Regev D, Eisen B, Fernandez‐Gracia J, Baskin P, Ben Jehuda R, Shulman R, Reiter I, Palty R, Arad M, Gottlieb E, Binah O. Depressed β-adrenergic inotropic responsiveness and intracellular calcium handling abnormalities in Duchenne Muscular Dystrophy patients' induced pluripotent stem cell-derived cardiomyocytes. J Cell Mol Med 2021; 25:3922-3934. [PMID: 33619882 PMCID: PMC8051742 DOI: 10.1111/jcmm.16341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an X-linked disease affecting male and rarely adult heterozygous females, resulting in death by the late 20s to early 30s. Previous studies reported depressed left ventricular function in DMD patients which may result from deranged intracellular Ca2+ -handling. To decipher the mechanism(s) underlying the depressed LV function, we tested the hypothesis that iPSC-CMs generated from DMD patients feature blunted positive inotropic response to β-adrenergic stimulation. To test the hypothesis, [Ca2+ ]i transients and contractions were recorded from healthy and DMD-CMs. While in healthy CMs (HC) isoproterenol caused a prominent positive inotropic effect, DMD-CMs displayed a blunted inotropic response. Next, we tested the functionality of the sarcoplasmic reticulum (SR) by measuring caffeine-induced Ca2+ release. In contrast to HC, DMD-CMs exhibited reduced caffeine-induced Ca2+ signal amplitude and recovery time. In support of the depleted SR Ca2+ stores hypothesis, in DMD-CMs the negative inotropic effects of ryanodine and cyclopiazonic acid were smaller than in HC. RNA-seq analyses demonstrated that in DMD CMs the RNA-expression levels of specific subunits of the L-type calcium channel, the β1-adrenergic receptor (ADRβ1) and adenylate cyclase were down-regulated by 3.5-, 2.8- and 3-fold, respectively, which collectively contribute to the depressed β-adrenergic responsiveness.
Collapse
MESH Headings
- Adrenergic Agents/pharmacology
- Adult
- Calcium/metabolism
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Cell Differentiation
- Female
- Gene Expression Regulation
- Humans
- Induced Pluripotent Stem Cells/drug effects
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/pathology
- Male
- Middle Aged
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardial Contraction
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA-Seq
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Sarcoplasmic Reticulum/drug effects
- Sarcoplasmic Reticulum/metabolism
- Sarcoplasmic Reticulum/pathology
Collapse
Affiliation(s)
- Lucy N. Mekies
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Danielle Regev
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Binyamin Eisen
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Jonatan Fernandez‐Gracia
- Department of Cell Biology and Cancer ScienceRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Polina Baskin
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Ronen Ben Jehuda
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
- Faculty of Biotechnology and Food EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Rita Shulman
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Irina Reiter
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Raz Palty
- Department of BiochemistryRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Michael Arad
- Leviev Heart CenterSheba Medical CenterRamat GanIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Eyal Gottlieb
- Department of Cell Biology and Cancer ScienceRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Ofer Binah
- Department of PhysiologyBiophysics and Systems BiologyRappaport Faculty of MedicineTechnion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
13
|
Barraza-Flores P, Fontelonga TM, Wuebbles RD, Hermann HJ, Nunes AM, Kornegay JN, Burkin DJ. Laminin-111 protein therapy enhances muscle regeneration and repair in the GRMD dog model of Duchenne muscular dystrophy. Hum Mol Genet 2019; 28:2686-2695. [PMID: 31179490 PMCID: PMC6687953 DOI: 10.1093/hmg/ddz086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 03/22/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked disease affecting ~1 in 5000 males. DMD patients exhibit progressive muscle degeneration and weakness, leading to loss of ambulation and premature death from cardiopulmonary failure. We previously reported that mouse Laminin-111 (msLam-111) protein could reduce muscle pathology and improve muscle function in the mdx mouse model for DMD. In this study, we examined the ability of msLam-111 to prevent muscle disease progression in the golden retriever muscular dystrophy (GRMD) dog model of DMD. The msLam-111 protein was injected into the cranial tibial muscle compartment of GRMD dogs and muscle strength and pathology were assessed. The results showed that msLam-111 treatment increased muscle fiber regeneration and repair with improved muscle strength and reduced muscle fibrosis in the GRMD model. Together, these findings support the idea that Laminin-111 could serve as a novel protein therapy for the treatment of DMD.
Collapse
Affiliation(s)
- Pamela Barraza-Flores
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Tatiana M Fontelonga
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Ryan D Wuebbles
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Hailey J Hermann
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Andreia M Nunes
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Joe N Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
14
|
Taylor M, Jefferies J, Byrne B, Lima J, Ambale-Venkatesh B, Ostovaneh MR, Makkar R, Goldstein B, Smith RR, Fudge J, Malliaras K, Fedor B, Rudy J, Pogoda JM, Marbán L, Ascheim DD, Marbán E, Victor RG. Cardiac and skeletal muscle effects in the randomized HOPE-Duchenne trial. Neurology 2019; 92:e866-e878. [PMID: 30674601 PMCID: PMC6396968 DOI: 10.1212/wnl.0000000000006950] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To assess the feasibility, safety, and efficacy of intracoronary allogeneic cardiosphere-derived cells (CAP-1002) in patients with Duchenne muscular dystrophy (DMD). METHODS The Halt Cardiomyopathy Progression (HOPE)-Duchenne trial is a phase I/II, randomized, controlled, open-label trial (NCT02485938). Patients with DMD >12 years old, with substantial myocardial fibrosis, were randomized (1:1) to usual care (control) or global intracoronary infusion of CAP-1002 (75 million cells). Participants were enrolled at 3 US medical centers between January and August 2016 and followed for 12 months. An independent Data and Safety Monitoring Board provided safety oversight. Cardiac function and structure were assessed by MRI, and analyzed by a blinded core laboratory. Skeletal muscle function was assessed by performance of the upper limb (PUL). RESULTS Twenty-five eligible patients (mean age 17.8 years; 68% wheelchair-dependent) were randomized to CAP-1002 (n = 13) or control (n = 12). Incidence of treatment-emergent adverse events was similar between groups. Compared to baseline, MRI at 12 months revealed significant scar size reduction and improvement in inferior wall systolic thickening in CAP-1002 but not control patients. Mid-distal PUL improved at 12 months in 8 of 9 lower functioning CAP-1002 patients, and no controls (p = 0.007). CONCLUSIONS Intracoronary CAP-1002 in DMD appears safe and demonstrates signals of efficacy on both cardiac and upper limb function for up to 12 months. Thus, future clinical research on CAP-1002 treatment of DMD cardiac and skeletal myopathies is warranted. CLASSIFICATION OF EVIDENCE This phase I/II study provides Class II evidence that for patients with DMD, intracoronary CAP-1002 is feasible and appears safe and potentially effective.
Collapse
Affiliation(s)
- Michael Taylor
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece.
| | - John Jefferies
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Barry Byrne
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Joao Lima
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Bharath Ambale-Venkatesh
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Mohammad R Ostovaneh
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Raj Makkar
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Bryan Goldstein
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Rachel Ruckdeschel Smith
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - James Fudge
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Konstantinos Malliaras
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Brian Fedor
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Jeff Rudy
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Janice M Pogoda
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Linda Marbán
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Deborah D Ascheim
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Eduardo Marbán
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| | - Ronald G Victor
- From The Heart Institute (M.T., J.J., B.G.), Cincinnati Children's Hospital Medical Center, OH; Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center (B.B.), and Division of Pediatric Cardiology, Congenital Heart Center (J.F.), University of Florida, Gainesville; Department of Cardiology (J.L., B.A.-V., M.R.O.), Johns Hopkins University, Baltimore, MD; Smidt Heart Institute (R.M., E.M., R.G.V.), Cedars-Sinai Medical Center, Los Angeles, CA; Capricor Therapeutics (R.R.S., B.F., J.R., J.M.P., L.M., D.D.A.), Beverly Hills, CA; and Department of Cardiology (K.M.), Laikon Hospital, Athens, Greece
| |
Collapse
|
15
|
Eisen B, Ben Jehuda R, Cuttitta AJ, Mekies LN, Shemer Y, Baskin P, Reiter I, Willi L, Freimark D, Gherghiceanu M, Monserrat L, Scherr M, Hilfiker-Kleiner D, Arad M, Michele DE, Binah O. Electrophysiological abnormalities in induced pluripotent stem cell-derived cardiomyocytes generated from Duchenne muscular dystrophy patients. J Cell Mol Med 2019; 23:2125-2135. [PMID: 30618214 PMCID: PMC6378185 DOI: 10.1111/jcmm.14124] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 01/09/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X‐linked progressive muscle degenerative disease, caused by mutations in the dystrophin gene and resulting in death because of respiratory or cardiac failure. To investigate the cardiac cellular manifestation of DMD, we generated induced pluripotent stem cells (iPSCs) and iPSC‐derived cardiomyocytes (iPSC‐CMs) from two DMD patients: a male and female manifesting heterozygous carrier. Dystrophin mRNA and protein expression were analysed by qRT‐PCR, RNAseq, Western blot and immunofluorescence staining. For comprehensive electrophysiological analysis, current and voltage clamp were used to record transmembrane action potentials and ion currents, respectively. Microelectrode array was used to record extracellular electrograms. X‐inactive specific transcript (XIST) and dystrophin expression analyses revealed that female iPSCs underwent X chromosome reactivation (XCR) or erosion of X chromosome inactivation, which was maintained in female iPSC‐CMs displaying mixed X chromosome expression of wild type (WT) and mutated alleles. Both DMD female and male iPSC‐CMs presented low spontaneous firing rate, arrhythmias and prolonged action potential duration. DMD female iPSC‐CMs displayed increased beat rate variability (BRV). DMD male iPSC‐CMs manifested decreased If density, and DMD female and male iPSC‐CMs showed increased ICa,L density. Our findings demonstrate cellular mechanisms underlying electrophysiological abnormalities and cardiac arrhythmias in DMD.
Collapse
Affiliation(s)
- Binyamin Eisen
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ronen Ben Jehuda
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.,Department of Biotechnology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ashley J Cuttitta
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lucy N Mekies
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yuval Shemer
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Polina Baskin
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Irina Reiter
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Lubna Willi
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Dov Freimark
- Leviev Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel E Michele
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Ofer Binah
- Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
16
|
Pradnya D, Nalini A, Nagarathna R, Raju TR, Sendhilkumar R, Meghana A, Sathyaprabha TN. Effect of Yoga as an Add-on Therapy in the Modulation of Heart Rate Variability in Children with Duchenne Muscular Dystrophy. Int J Yoga 2019; 12:55-61. [PMID: 30692784 PMCID: PMC6329227 DOI: 10.4103/ijoy.ijoy_12_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Duchene muscular dystrophy (DMD) is a progressive muscular disorder. Cardiac disorder is the second-most common cause of death in children with DMD, with 10%–20% of them dying of cardiac failure. Heart rate variability (HRV) is shown to be a predictor of cardio-autonomic function. Physiotherapy (PT) is advised for these children as a regular treatment for maintaining their functional status. The effect of yogic practices on the cardio-autonomic functions has been demonstrated in various neurological conditions and may prove beneficial in DMD. Materials and Methods: In this study, 124 patients with DMD were randomized to PT alone or PT with yoga intervention. Home-based PT and yoga were advised. Adherence was serially assessed at a follow-up interval of 3 months. Error-free, electrocardiogram was recorded in all patients at rest in the supine position. HRV parameters were computed in time and frequency domains. HRV was recorded at baseline and at an interval of 3 months up to 1 year. Repeated-measures ANOVA was used to analyze longitudinal follow-up and least significant difference for post hoc analysis and P < 0.05 was considered statistically significant. Results: In our study, with PT protocol, standard deviation of NN, root of square mean of successive NN, total power, low frequency, high-frequency normalized units (HFnu), and sympathovagal balance improved at varying time points and the improvement lasted up for 6–9 months, whereas PT and yoga protocol showed an improvement in HFnu during the last 3 months of the study period and all the other parameters were stable up to 1 year. Thus, it is evident that both the groups improved cardiac functions in DMD. However, no significant difference was noted in the changes observed between the groups. Conclusion: The intense PT and PT with yoga, particularly home-based program, is indeed beneficial as a therapeutic strategy in DMD children to maintain and/or to sustain HRV in DMD.
Collapse
Affiliation(s)
- Dhargave Pradnya
- Physiotherapy Center, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | | | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Ragupathy Sendhilkumar
- Physiotherapy Center, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Adoor Meghana
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Talakad N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
17
|
Heart Rate Variability and Cardiopulmonary Dysfunction in Patients with Duchenne Muscular Dystrophy: A Systematic Review. Pediatr Cardiol 2018; 39:869-883. [PMID: 29696428 DOI: 10.1007/s00246-018-1881-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/04/2018] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic recessive disorder with progressive muscle weakness. Despite the general muscle wasting, degeneration and necrosis of cardiomyocytes have been the main causes of morbidity and death in individuals with DMD. Cardiac failure is generally preceded by disturbances in heart rate variability (HRV), and non-invasive measurement of the autonomic nervous system has been an important tool to predict adverse cardiovascular events. Hence, the application of HRV to study autonomic modulation in DMD individuals, and the establishment of correlations between HRV and heart/lung diseases, age, and mortality will have the potential to improve quality of life and life expectancy of individuals with DMD. In order to evaluate the state of the art in this field, we conducted a systematic search in Medline/PubMed and BVS (virtual library in health) databases. We selected 8 studies using pre-defined criteria and meta-analysis revealed decreased parasympathetic activity and increased sympathetic predominance in individuals with DMD as major observations. Moreover, there is a strong association between diminished HRV and myocardial fibrosis with DMD. These patterns are evident in patients at early-stage DMD and become more prominent as disease severity and age increase. Thus, data minning clearly indicates that HRV assessment can be used as a predictor for sudden death in individuals with DMD. The use of the HRV, which is inexpensive, ubiquitously available in clinics and hospitals, and a non-invasive analysis tool, can save lives and decrease the morbity in DMD by alerting care givers to consider autonomic nervous system intervention.
Collapse
|
18
|
Botsva N, Naishtetik I, Khimion L, Chernetchenko D. Predictors of aging based on the analysis of heart rate variability. PACING AND CLINICAL ELECTROPHYSIOLOGY: PACE 2017; 40:1269-1278. [PMID: 28983984 DOI: 10.1111/pace.13180] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/31/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND The current significant progress in the use of heart rate variability in the solution of many diagnostic and therapeutic problems is determined by the availability of standardized methods of measurement and physiological interpretation of heart rate variability indices on the one hand and the high technological level of state-of-the-art electronic measuring equipment that is used for automatic registration and computer processing of cardio-signals. METHODS A retrospective analysis of anonymized cardio screening results of 22,433 adult residents of 565 settlements (cities and villages) across all 20 administrative districts of the Khmelnytskyi Region (Ukraine) was conducted to find a statistically significant connection between individual heart rate variability parameters and the age of people. RESULTS Primary statistical analysis and visualization showed a correlation between the selected heart rate variability parameters and the age and sex of the examined persons. The study found values of the predicted age slightly over estimation versus the actual age for very young test subjects and below estimation for elderly subjects. CONCLUSION The use of neural network computations and the modification of the algorithm through the construction of individual training samples for different age intervals, and the creation of individual ensembles of classification neural networks, therefore achieved a prediction of the age of examined persons based on the values of their time and frequency domain heart rate variability indices, with 87% accuracy for women and 85% accuracy for men in the 66-85 years age interval and at least 85% for age groups across the entire sample.
Collapse
Affiliation(s)
- Nataliia Botsva
- Oles Honchar Dnipropetrovsk National University, 20 Kazakova Str., Dnipro, 49010, Ukraine
| | - Iryna Naishtetik
- Academy of the Postgraduate Education named after P.L. Schupik, Dorogozhytska Str., Kyiv, 04112, Ukraine
| | - Ludmyla Khimion
- Academy of the Postgraduate Education named after P.L. Schupik, Dorogozhytska Str., Kyiv, 04112, Ukraine
| | - Dmitriy Chernetchenko
- Oles Honchar Dnipropetrovsk National University, 20 Kazakova Str., Dnipro, 49010, Ukraine
| |
Collapse
|
19
|
D'Amario D, Amodeo A, Adorisio R, Tiziano FD, Leone AM, Perri G, Bruno P, Massetti M, Ferlini A, Pane M, Niccoli G, Porto I, D'Angelo GA, Borovac JA, Mercuri E, Crea F. A current approach to heart failure in Duchenne muscular dystrophy. Heart 2017; 103:1770-1779. [PMID: 28668906 DOI: 10.1136/heartjnl-2017-311269] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/10/2017] [Accepted: 05/15/2017] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic, progressive neuromuscular condition that is marked by the long-term muscle deterioration with significant implications of pulmonary and cardiac dysfunction. As such, end-stage heart failure (HF) in DMD is increasingly becoming the main cause of death in this population. The early detection of cardiomyopathy is often challenging, due to a long subclinical phase of ventricular dysfunction and difficulties in assessment of cardiovascular symptomatology in these patients who usually loose ambulation during the early adolescence. However, an early diagnosis of cardiovascular disease in patients with DMD is decisive since it allows a timely initiation of cardioprotective therapies that can mitigate HF symptoms and delay detrimental heart muscle remodelling. Echocardiography and ECG are standardly used for screening and detection of cardiovascular abnormalities in these patients, although these tools are not always adequate to detect an early, clinically asymptomatic phases of disease progression. In this regard, cardiovascular magnetic resonance (CMR) with late gadolinium enhancement is emerging as a promising method for the detection of early cardiac involvement in patients with DMD. The early detection of cardiac dysfunction allows the therapeutic institution of various classes of drugs such as corticosteroids, beta-blockers, ACE inhibitors, antimineralocorticoid diuretics and novel pharmacological and surgical solutions in the multimodal and multidisciplinary care for this group of patients. This review will focus on these challenges and available options for HF in patients with DMD.
Collapse
Affiliation(s)
- Domenico D'Amario
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Antonio Amodeo
- Department of Paediatric Cardiology and Cardiac Surgery, Bambino Gesù Hospital, Rome, Italy
| | - Rachele Adorisio
- Department of Paediatric Cardiology and Cardiac Surgery, Bambino Gesù Hospital, Rome, Italy
| | | | - Antonio Maria Leone
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Gianluigi Perri
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy.,Department of Paediatric Cardiology and Cardiac Surgery, Bambino Gesù Hospital, Rome, Italy
| | - Piergiorgio Bruno
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Marika Pane
- Department of Neurology, Catholic University of the Sacred Heart, Rome, Italy
| | - Giampaolo Niccoli
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Italo Porto
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | - Gianluca A D'Angelo
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Eugenio Mercuri
- Department of Neurology, Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Medicine, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
20
|
Alvarez MPB, da Silva TD, Favero FM, Valenti VE, Raimundo RD, Vanderlei LCM, Garner DM, Monteiro CBDM. Autonomic Modulation in Duchenne Muscular Dystrophy during a Computer Task: A Prospective Control Trial. PLoS One 2017; 12:e0169633. [PMID: 28118369 PMCID: PMC5261738 DOI: 10.1371/journal.pone.0169633] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 12/20/2016] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Duchenne Muscular Dystrophy (DMD) is characterized by progressive muscle weakness that can lead to disability. Owing to functional difficulties faced by individuals with DMD, the use of assistive technology is essential to provide or facilitate functional abilities. In DMD, cardiac autonomic dysfunction has been reported in addition to musculoskeletal impairment. Consequently, the objective was to investigate acute cardiac autonomic responses, by Heart Rate Variability (HRV), during computer tasks in subjects with DMD. METHOD HRV was assessed by linear and nonlinear methods, using the heart rate monitor Polar RS800CX chest strap Electrocardiographic measuring device. Then, 45 subjects were included in the group with DMD and 45 in the healthy Typical Development (TD) control group. They were assessed for twenty minutes at rest sitting, and five minutes after undergoing a task on the computer. RESULTS Individuals with DMD had a statistically significant lower parasympathetic cardiac modulation at rest when compared to the control group, which further declined when undergoing the tasks on the computer. CONCLUSION DMD patients presented decreased HRV and exhibited greater intensity of cardiac autonomic responses during computer tasks characterized by vagal withdrawal when compared to the healthy TD control subjects.
Collapse
Affiliation(s)
- Mayra Priscila Boscolo Alvarez
- Physical Therapy, Speech and Occupational Therapy Department, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Talita Dias da Silva
- Federal University of São Paulo, Paulista School of Medicine, São Paulo, SP, Brazil
| | - Francis Meire Favero
- Federal University of São Paulo, Department of Neurology/Neurosurgery, Paulista School of Medicine, São Paulo, SP, Brazil
- * E-mail:
| | - Vitor Engrácia Valenti
- Autonomic Nervous System Center Study, Speech Therapy Department Faculty of Sciences, Paulista State University (UNESP), Marília, SP, Brazil
| | - Rodrigo Daminello Raimundo
- Laboratory Design and Scientific Writing, Department of Community Health, ABC Medical School, Santo André, SP, Brazil
| | | | - David M. Garner
- Cardiorespiratory Research Group, Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, United Kingdom
| | - Carlos Bandeira de Mello Monteiro
- Physical Therapy, Speech and Occupational Therapy Department, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
- School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
21
|
Sujan MU, Rao MR, Kisan R, Abhishekh HA, Nalini A, Raju TR, Sathyaprabha TN. Influence of hydrotherapy on clinical and cardiac autonomic function in migraine patients. J Neurosci Rural Pract 2016; 7:109-13. [PMID: 26933356 PMCID: PMC4750306 DOI: 10.4103/0976-3147.165389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background: Migraine is associated with autonomic symptoms. The growing body of literature suggests that the dysfunctional autonomic nervous system might play a pivotal role in the pathogenesis of migraine. Thermal therapies have been hypothesized to modulate these changes and alleviate pain. However, data regarding the efficacy of hydrotherapy in migraine remain scant. We evaluated the effect of add on hydrotherapy procedure (a hot arm and foot bath with ice massage to head) in migraine patients. Methods: Forty chronic migraine patients fulfilling the International Classification of Headache Disorders II criteria were recruited from the neurology outpatient clinic. Patients were randomized to receive either hydrotherapy plus conventional pharmacological care (n = 20) or conventional medication only (n = 20). Hydrotherapy group received treatment with hot arm and foot bath (103°F to 110°F) and ice massage to head daily for 20 min for 45 days. Patients were assessed using headache impact test (HIT), visual analog scale for pain and cardiac autonomic function by heart rate variability (HRV) before and after intervention period. Results: There was a significant decrease in HIT score, frequency, and intensity of headaches following treatment in both the groups. However, it was more evident in add on hydrotherapy group compared to pharmacological treatment alone group. There was also significant improvement in the HRV parameters. In particular, there was a significant decrease in heart rate (P = 0.017), increase in high frequency (HF) (P = 0.014) and decrease in low frequency/HF ratio (P = 0.004) in add on hydrotherapy group. Conclusion: Our study shows that add on hydrotherapy enhanced the vagal tone in addition to reducing the frequency and intensity of headaches in migraine patients.
Collapse
Affiliation(s)
- M U Sujan
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - M Raghavendra Rao
- Department of Complementary and Alternative Medicine, Health Care Global Enterprises Private Ltd., Bengaluru, Karnataka, India
| | - Ravikiran Kisan
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Hulegar A Abhishekh
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - T N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
22
|
Abstract
Several lines of investigations have shown the deleterious effect of an alcohol on the autonomic nervous system. Recent evidence shows that infants exposed to alcohol during the antenatal period displayed aberration in the cardiac autonomic function after the birth. However, there is dearth of literature on the long term influence of antenatal alcohol exposure. In this study we measured the cardiac autonomic functions in children who were exposed to alcohol in the antenatal period and compared them with non-exposed control children. Twenty eight children (age: 9±2 years) in the antenatal alcohol exposed group and age, gender matched 30 non exposed healthy volunteers as a control (age: 10±2 years) were recruited. Electrocardiogram was recorded in all subjects at rest in the supine position. HRV parameters were analyzed in the time and frequency domains using customized software. The average heart rate was similar between both the groups. There was no statistical significant difference in the time domain measures between the groups. However, the low frequency power, normalized units and low frequency to high frequency ratio were significantly higher in the antenatal alcohol exposed children compared to the controls. This suggests sympathetic predominance in children who were exposed to alcohol in the antenatal period. In this study we provide evidence for the deleterious long lasting effect of antenatal exposure of alcohol on cardiac autonomic regulation. Further prospective studies are needed to confirm the causal relationship between antenatal alcohol exposure and autonomic dysregulation.
Collapse
|