1
|
Esteso P, Auerbach SR, Bansal N, Harris R, Soslow JH, Birnbaum BF, Conway J, Cripe LH, Nandi D, Hayes E, Gambetta KE, Hall EK, Hsu DT, Kaufman BD, Rosenthal D, Kirmani S, Ploutz MS, Lal AK, Peng DM, Villa CR, Shugh S, Wittlieb-Weber CA, Shih R. Cardiac treatment for Duchenne muscular dystrophy: consensus recommendations from the ACTION muscular dystrophy committee. Cardiol Young 2025; 35:770-775. [PMID: 40012319 DOI: 10.1017/s1047951125000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Duchenne muscular dystrophy is a devastating neuromuscular disorder characterized by the loss of dystrophin, inevitably leading to cardiomyopathy. Despite publications on prophylaxis and treatment with cardiac medications to mitigate cardiomyopathy progression, gaps remain in the specifics of medication initiation and optimization. METHOD This document is an expert opinion statement, addressing a critical gap in cardiac care for Duchenne muscular dystrophy. It provides thorough recommendations for the initiation and titration of cardiac medications based on disease progression and patient response. Recommendations are derived from the expertise of the Advance Cardiac Therapies Improving Outcomes Network and are informed by established guidelines from the American Heart Association, American College of Cardiology, and Duchenne Muscular Dystrophy Care Considerations. These expert-derived recommendations aim to navigate the complexities of Duchenne muscular dystrophy-related cardiac care. RESULTS Comprehensive recommendations for initiation, titration, and optimization of critical cardiac medications are provided to address Duchenne muscular dystrophy-associated cardiomyopathy. DISCUSSION The management of Duchenne muscular dystrophy requires a multidisciplinary approach. However, the diversity of healthcare providers involved in Duchenne muscular dystrophy can result in variations in cardiac care, complicating treatment standardization and patient outcomes. The aim of this report is to provide a roadmap for managing Duchenne muscular dystrophy-associated cardiomyopathy, by elucidating timing and dosage nuances crucial for optimal therapeutic efficacy, ultimately improving cardiac outcomes, and improving the quality of life for individuals with Duchenne muscular dystrophy. CONCLUSION This document seeks to establish a standardized framework for cardiac care in Duchenne muscular dystrophy, aiming to improve cardiac prognosis.
Collapse
Affiliation(s)
- Paul Esteso
- Cardiology, Boston Children's Hospital, Boston, MA, USA
| | | | - Neha Bansal
- Pediatric Cardiology, Mount Sinai Kravis Children's Hospital, New York, NY, USA
| | - Rachel Harris
- Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| | - Jonathan H Soslow
- Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, TN, USA
| | | | - Jennifer Conway
- Cardiology, Stollery Children's Hospital, Edmonton, AB, Canada
| | - Linda H Cripe
- Cardiology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Deipanjan Nandi
- Cardiology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Emily Hayes
- Cardiology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Katheryn E Gambetta
- Cardiology, Ann & Robert Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - E Kevin Hall
- Pediatric Cardiology, Yale Children's Hospital, New Heaven, CT, USA
| | - Daphne T Hsu
- Cardiology, The Children's Hospital at Montefiore, Bronx, NY, USA
| | - Beth D Kaufman
- Pediatric Cardiology, Lucille Packard Children's Hospital, Palo Alto, CA, USA
| | - David Rosenthal
- Pediatric Cardiology, Lucille Packard Children's Hospital, Palo Alto, CA, USA
| | - Sonya Kirmani
- Cardiology, American Family Children's Hospital at the University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle S Ploutz
- Pediatric Cardiology, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Ashwin K Lal
- Pediatric Cardiology, Primary Children's Hospital, Salt Lake City, UT, USA
| | - David M Peng
- Cardiology, C.S Mott Children's Hospital, Ann Arbor, MI, USA
| | - Chet R Villa
- Cardiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Svetlana Shugh
- Pediatric Cardiology, Joe DiMaggio Children's Hospital, Hollywood, FL, USA
| | | | - Renata Shih
- Child Health Research Institute at University of Florida Shands Children's Hospital, Gainesville, FL, USA
| |
Collapse
|
2
|
Marion-Knudsen R, Lindberg LA, Jespersen T, Saljic A. Quantitative histologic assessment of atrial fibrillation-associated fibrosis in animal models: A systematic review. Heart Rhythm 2025:S1547-5271(25)02102-2. [PMID: 40058516 DOI: 10.1016/j.hrthm.2025.03.1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia, and cardiac fibrosis is a major component in driving its progressive nature. Quantitative histologic assessment of fibrosis in animal models is crucial for understanding AF, but current published studies present various methodologies that limit comparison. This systematic review examines 195 AF studies across multiple animal models (mice, rats, goats, dogs, pigs, and horses) to summarize (1) quantified fibrosis results and (2) methodologies for histologic fibrosis assessment; and (3) evaluate antifibrotic therapies used in these studies. The fibrosis quantified across the studies ranged from 0.34%-60.2% depending on the animal, intervention model, and quantification method. The findings underscore the need for a standardized fibrosis quantification protocol in AF research, enabling comparison across studies and offering greater insight into potential pharmacologic interventions.
Collapse
Affiliation(s)
- Rikke Marion-Knudsen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lucas Alexander Lindberg
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Jespersen
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Laboratory of Cardiac Physiology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Routh BL, Tripathi R, Giuliano E, Lujin P, Sinha PR, Mohan RR. Anti-fibrotic effects of lisinopril (ACE inhibitor) and fasudil (ROCK inhibitor) in combination for canine corneal fibrosis in vitro. Vet Ophthalmol 2024. [PMID: 39592228 DOI: 10.1111/vop.13304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/02/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Corneal fibrosis is a leading cause of blindness in mammalian species and may result in compromised performance in sports and daily functions. This study evaluated the safety and anti-fibrotic effects of the FDA-approved drugs, angiotensin-converting enzyme inhibitor (ACE-I) lisinopril and rho-kinase inhibitor (ROCK-I) fasudil, alone and in combination, on the canine cornea using an established in vitro model. METHODS To test the safety and efficacy of lisinopril and fasudil, primary canine corneal fibroblasts (CCFs) generated from donor corneas of healthy dogs (n = 20) were used. A series of dose-dependent and time-dependent assays with lisinopril (1-50 μM) and fasudil (1-10 nM) were performed. qRT-PCR, immunofluorescence (IF) staining, cell viability assay, cell proliferation assay, LIVE/DEAD viability/cytotoxicity assay, TUNEL assay, and total cell count were performed. RESULTS A 25-μM lisinopril and 3-nM fasudil dose were safe, nontoxic, and optimal for therapeutic evaluations in vitro. Treatments of lisinopril or fasudil, alone or in-combination, to CCFs grown in the presence of TGF-β1 (5 ng/mL) showed inhibition of myofibroblast formation based on phase-contrast microscopy. The qRT-PCR and IF studies showed a significant decrease in expression of profibrotic markers, including α-smooth muscle actin (α-SMA; p < .0001), fibronectin (FN; p = .0002), tenascin C (TNC; p < .0001), Collagen I (Col-I; p < .0001), Collagen IIIA1 (Co-IIIA1; p < .0001), and Collagen IV (Co-lV; p < .0001). CONCLUSION An ophthalmic formulation consisting of lisinopril and fasudil may offer a safe and effective method to treat canine corneal fibrosis. Additional studies evaluating safety and efficacy of this formulation in vivo are warranted.
Collapse
Affiliation(s)
- Brayden L Routh
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ratnakar Tripathi
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
| | - Elizabeth Giuliano
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
| | - Payton Lujin
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
| | - Prashant R Sinha
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
| | - Rajiv R Mohan
- Department of Veterinary Medicine and Surgery, One-Health One-Medicine Ophthalmology and Vision Research Center, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
- Mason Eye Institute, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
4
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
5
|
Russo V, Antonini G, Massa R, Casali C, Mauriello A, Martino AM, Marconi R, Garibaldi M, Franciosa P, Zecchin M, Gaudio C, D’Andrea A, Strano S. Comprehensive Cardiovascular Management of Myotonic Dystrophy Type 1 Patients: A Report from the Italian Neuro-Cardiology Network. J Cardiovasc Dev Dis 2024; 11:63. [PMID: 38392277 PMCID: PMC10889677 DOI: 10.3390/jcdd11020063] [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: 01/01/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024] Open
Abstract
Myotonic dystrophy is a hereditary disorder with systemic involvement. The Italian Neuro-Cardiology Network-"Rete delle Neurocardiologie" (INCN-RNC) is a unique collaborative experience involving neurology units combined with cardio-arrhythmology units. The INCN facilitates the creation of integrated neuro-cardiac teams in Neuromuscular Disease Centers for the management of cardiovascular involvement in the treatment of myotonic dystrophy type 1 (MD1).
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”—“Monaldi” Hospital, 80126 Naples, Italy;
| | - Giovanni Antonini
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, “Sant’Andrea” Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (G.A.); (M.G.)
| | - Roberto Massa
- Neuromuscular Diseases Unit, Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Carlo Casali
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00196 Rome, Italy;
| | - Alfredo Mauriello
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”—“Monaldi” Hospital, 80126 Naples, Italy;
- Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | | | - Roberto Marconi
- Unit of Neurology, Cardio-Thoracic-Neuro-Vascular Department, “Misericordia” Hospital, 58100 Grosseto, Italy;
| | - Matteo Garibaldi
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, “Sant’Andrea” Hospital, Via di Grottarossa, 1035-1039, 00189 Rome, Italy; (G.A.); (M.G.)
| | - Pasquale Franciosa
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| | - Massimo Zecchin
- Cardiothoracovascular Department, “Cattinara” Hospital, ASUGI and University of Trieste, 34149 Trieste, Italy;
| | - Carlo Gaudio
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| | - Antonello D’Andrea
- Department of Cardiology, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | - Stefano Strano
- Department of Internal, Anesthesiological and Cardiovascular Clinical Sciences, Sapienza University of Rome, 00196 Rome, Italy; (P.F.); (C.G.); (S.S.)
| |
Collapse
|
6
|
Kashyap N, Nikhanj A, Labib D, Prosia E, Rivest S, Flewitt J, Pfeffer G, Bakal JA, Siddiqi ZA, Coulden RA, Thompson R, White JA, Oudit GY. Prognostic Utility of Cardiovascular Magnetic Resonance-Based Phenotyping in Patients With Muscular Dystrophy. J Am Heart Assoc 2023; 12:e030229. [PMID: 37929714 PMCID: PMC10727409 DOI: 10.1161/jaha.123.030229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
Background The prognostic utility of cardiovascular magnetic resonance imaging, including strain analysis and tissue characterization, has not been comprehensively investigated in adult patients with muscular dystrophy. Methods and Results We prospectively enrolled 148 patients with dystrophinopathies (including heterozygotes), limb-girdle muscular dystrophy, and type 1 myotonic dystrophy (median age, 36.0 [interquartile range, 23.0-50.0] years; 51 [34.5%] women) over 7.7 years in addition to an age- and sex-matched healthy control cohort (n=50). Cardiovascular magnetic resonance markers, including 3-dimensional strain and fibrosis, were assessed for their respective association with major adverse cardiac events. Our results showed that markers of contractile performance were reduced across all muscular dystrophy groups. In particular, the dystrophinopathies cohort experienced reduced left ventricular (LV) ejection fraction and high burden of replacement fibrosis. Patients with type 1 myotonic dystrophy showed a 26.8% relative reduction in LV mass with corresponding reduction in chamber volumes. Eighty-two major adverse cardiac events occurred over a median follow-up of 5.2 years. Although LV ejection fraction was significantly associated with major adverse cardiac events (adjusted hazard ratio [aHR], 3.0 [95% CI, 1.4-6.4]) after adjusting for covariates, peak 3-dimensional strain amplitude demonstrated greater predictive value (minimum principal amplitude: aHR, 5.5 [95% CI, 2.5-11.9]; maximum principal amplitude: aHR, 3.3 [95% CI, 1.6-6.8]; circumferential amplitude: aHR, 3.4 [95% CI, 1.6-7.2]; longitudinal amplitude: aHR, 3.4 [95% CI, 1.7-6.9]; and radial strain amplitude: aHR, 3.0 [95% CI, 1.4-6.1]). Minimum principal strain yielded incremental prognostic value beyond LV ejection fraction for association with major adverse cardiac events (change in χ2=13.8; P<0.001). Conclusions Cardiac dysfunction is observed across all muscular dystrophy subtypes; however, the subtypes demonstrate distinct phenotypic profiles. Myocardial deformation analysis highlights unique markers of principal strain that improve risk assessment over other strain markers, LV ejection fraction, and late gadolinium enhancement in this vulnerable patient population.
Collapse
Affiliation(s)
- Niharika Kashyap
- Division of CardiologyUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medicine, Faculty of Medicine and DentistryMazankowski Alberta Heart Institute, University of AlbertaEdmontonAlbertaCanada
| | - Anish Nikhanj
- Division of CardiologyUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medicine, Faculty of Medicine and DentistryMazankowski Alberta Heart Institute, University of AlbertaEdmontonAlbertaCanada
| | - Dina Labib
- Department of Cardiac SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Libin Cardiovascular Institute of Alberta, University of CalgaryCalgaryAlbertaCanada
| | - Easter Prosia
- Department of Cardiac SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Libin Cardiovascular Institute of Alberta, University of CalgaryCalgaryAlbertaCanada
| | - Sandra Rivest
- Department of Cardiac SciencesUniversity of AlbertaEdmontonAlbertaCanada
| | - Jacqueline Flewitt
- Libin Cardiovascular Institute of Alberta, University of CalgaryCalgaryAlbertaCanada
| | - Gerald Pfeffer
- Department of Clinical NeurosciencesUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, University of CalgaryCalgaryAlbertaCanada
| | - Jeffrey A. Bakal
- Alberta Strategy for Patient Oriented Research UnitUniversity of CalgaryCalgaryAlbertaCanada
- Provincial Research Data ServicesUniversity of AlbertaEdmontonAlbertaCanada
| | - Zaeem A. Siddiqi
- Division of Neurology, Department of Medicine, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Richard A. Coulden
- Department of Radiology and Diagnostic ImagingUniversity of Alberta HospitalEdmontonAlbertaCanada
| | - Richard Thompson
- Division of CardiologyUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medicine, Faculty of Medicine and DentistryMazankowski Alberta Heart Institute, University of AlbertaEdmontonAlbertaCanada
| | - James A. White
- Department of Cardiac SciencesUniversity of AlbertaEdmontonAlbertaCanada
- Libin Cardiovascular Institute of Alberta, University of CalgaryCalgaryAlbertaCanada
| | - Gavin Y. Oudit
- Division of CardiologyUniversity of AlbertaEdmontonAlbertaCanada
- Department of Medicine, Faculty of Medicine and DentistryMazankowski Alberta Heart Institute, University of AlbertaEdmontonAlbertaCanada
| |
Collapse
|
7
|
Mogharehabed F, Czubryt MP. The role of fibrosis in the pathophysiology of muscular dystrophy. Am J Physiol Cell Physiol 2023; 325:C1326-C1335. [PMID: 37781738 DOI: 10.1152/ajpcell.00196.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Muscular dystrophy exerts significant and dramatic impacts on affected patients, including progressive muscle wasting leading to lung and heart failure, and results in severely curtailed lifespan. Although the focus for many years has been on the dysfunction induced by the loss of function of dystrophin or related components of the striated muscle costamere, recent studies have demonstrated that accompanying pathologies, particularly muscle fibrosis, also contribute adversely to patient outcomes. A significant body of research has now shown that therapeutically targeting these accompanying pathologies via their underlying molecular mechanisms may provide novel approaches to patient management that can complement the current standard of care. In this review, we discuss the interplay between muscle fibrosis and muscular dystrophy pathology. A better understanding of these processes will contribute to improved patient care options, restoration of muscle function, and reduced patient morbidity and mortality.
Collapse
Affiliation(s)
- Farnaz Mogharehabed
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
8
|
Russo V, Capolongo A, Bottino R, Carbone A, Palladino A, Liccardo B, Nigro G, Marchel M, Golino P, D’Andrea A. Echocardiographic Features of Cardiac Involvement in Myotonic Dystrophy 1: Prevalence and Prognostic Value. J Clin Med 2023; 12:1947. [PMID: 36902735 PMCID: PMC10004242 DOI: 10.3390/jcm12051947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults. Cardiac involvement is reported in 80% of cases and includes conduction disturbances, arrhythmias, subclinical diastolic and systolic dysfunction in the early stage of the disease; in contrast, severe ventricular systolic dysfunction occurs in the late stage of the disease. Echocardiography is recommended at the time of diagnosis with periodic revaluation in DM1 patients, regardless of the presence or absence of symptoms. Data regarding the echocardiographic findings in DM1 patients are few and conflicting. This narrative review aimed to describe the echocardiographic features of DM1 patients and their prognostic role as predictors of cardiac arrhythmias and sudden death.
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Antonio Capolongo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Roberta Bottino
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Andreina Carbone
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Alberto Palladino
- Cardiomyology and Genetic Section, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Biagio Liccardo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Gerardo Nigro
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Michał Marchel
- 1st Department of Cardiology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Paolo Golino
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania “Luigi Vanvitelli”, Monaldi Hospital, 80121 Naples, Italy
| | - Antonello D’Andrea
- Department of Cardiology, Umberto I Hospital, 84014 Nocera Inferiore, Italy
| |
Collapse
|
9
|
Groh WJ, Bhakta D, Tomaselli GF, Aleong RG, Teixeira RA, Amato A, Asirvatham SJ, Cha YM, Corrado D, Duboc D, Goldberger ZD, Horie M, Hornyak JE, Jefferies JL, Kääb S, Kalman JM, Kertesz NJ, Lakdawala NK, Lambiase PD, Lubitz SA, McMillan HJ, McNally EM, Milone M, Namboodiri N, Nazarian S, Patton KK, Russo V, Sacher F, Santangeli P, Shen WK, Sobral Filho DC, Stambler BS, Stöllberger C, Wahbi K, Wehrens XHT, Weiner MM, Wheeler MT, Zeppenfeld K. 2022 HRS expert consensus statement on evaluation and management of arrhythmic risk in neuromuscular disorders. Heart Rhythm 2022; 19:e61-e120. [PMID: 35500790 DOI: 10.1016/j.hrthm.2022.04.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/04/2022]
Abstract
This international multidisciplinary document is intended to guide electrophysiologists, cardiologists, other clinicians, and health care professionals in caring for patients with arrhythmic complications of neuromuscular disorders (NMDs). The document presents an overview of arrhythmias in NMDs followed by detailed sections on specific disorders: Duchenne muscular dystrophy, Becker muscular dystrophy, and limb-girdle muscular dystrophy type 2; myotonic dystrophy type 1 and type 2; Emery-Dreifuss muscular dystrophy and limb-girdle muscular dystrophy type 1B; facioscapulohumeral muscular dystrophy; and mitochondrial myopathies, including Friedreich ataxia and Kearns-Sayre syndrome, with an emphasis on managing arrhythmic cardiac manifestations. End-of-life management of arrhythmias in patients with NMDs is also covered. The document sections were drafted by the writing committee members according to their area of expertise. The recommendations represent the consensus opinion of the expert writing group, graded by class of recommendation and level of evidence utilizing defined criteria. The recommendations were made available for public comment; the document underwent review by the Heart Rhythm Society Scientific and Clinical Documents Committee and external review and endorsement by the partner and collaborating societies. Changes were incorporated based on these reviews. By using a breadth of accumulated available evidence, the document is designed to provide practical and actionable clinical information and recommendations for the diagnosis and management of arrhythmias and thus improve the care of patients with NMDs.
Collapse
Affiliation(s)
- William J Groh
- Ralph H. Johnson VA Medical Center and Medical University of South Carolina, Charleston, South Carolina
| | - Deepak Bhakta
- Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | - Anthony Amato
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Domenico Corrado
- Department of Cardiac, Thoracic, and Vascular Sciences, University of Padova, Padova, Italy
| | - Denis Duboc
- Cardiology Department, Hôpital Cochin, AP-HP, Université de Paris, Paris, France
| | - Zachary D Goldberger
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Minoru Horie
- Shiga University of Medical Sciences, Otsu, Japan
| | | | | | - Stefan Kääb
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Jonathan M Kalman
- Royal Melbourne Hospital and University of Melbourne, Melbourne, Victoria, Australia
| | | | - Neal K Lakdawala
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pier D Lambiase
- Barts Heart Centre, St Bartholomew's Hospital, University College London, and St Bartholomew's Hospital London, London, United Kingdom
| | | | - Hugh J McMillan
- Montreal Children's Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | - Narayanan Namboodiri
- Sree Chitra Institute for Medical Sciences and Technology, Thiruvananthapuram, India
| | | | | | | | - Frederic Sacher
- Bordeaux University Hospital, LIRYC Institute, Bordeaux, France
| | | | | | | | | | - Claudia Stöllberger
- Second Medical Department with Cardiology and Intensive Care Medicine, Klinik Landstraße, Vienna, Austria
| | - Karim Wahbi
- Cardiology Department, Hôpital Cochin, AP-HP, Université de Paris, Paris, France
| | | | | | | | | |
Collapse
|
10
|
Oliveira-Santos A, Dagda M, Burkin DJ. Sunitinib inhibits STAT3 phosphorylation in cardiac muscle and prevents cardiomyopathy in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2022; 31:2358-2369. [PMID: 35157045 PMCID: PMC9307308 DOI: 10.1093/hmg/ddac042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 11/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked genetic disorder affecting approximately 1 in 5000 male births worldwide. DMD is caused by mutations in the dystrophin gene. Dystrophin is essential for maintaining muscle cell membrane integrity and stability by linking the cytoskeleton to the extracellular matrix, which protects myofibers from contraction-induced damage. Loss of dystrophin leads to mechanically induced skeletal and cardiac muscle damage. Although the disease is not evident in DMD patients at birth, muscular dystrophy rapidly progresses and results in respiratory and cardiac muscle failure as early as the teenage years. Premature death in DMD patients is due to cardiac arrhythmias and left ventricular dysfunction. Currently, there is no effective treatment for DMD-related cardiac failure. Recently, we have shown that a Food and Drug Administration-approved small molecule, sunitinib, a multi-targeted tyrosine kinase inhibitor can mitigate skeletal muscle disease through an increase in myogenic capacity, cell membrane integrity, and improvement of skeletal muscle function via regulation of STAT3-related signaling pathway. Chronic activation of STAT3 has been shown to promote cardiac hypertrophy and failure. In this study, we examined the effects of long-term sunitinib treatment on cardiac pathology and function. Our results showed sunitinib treatment reduced STAT3 phosphorylation in the heart muscle of mdx mice, improved cardiac electrical function, increased cardiac output and stroke volume, decreased ventricular hypertrophy, reduced cardiomyocytes membrane damage, fibrotic tissue deposition and slightly decreased cardiac inflammation. Together, our studies support the idea that sunitinib could serve as a novel treatment to slow cardiomyopathy progression in DMD. One Sentence Summary In this study, we determined if sunitinib, a Food and Drug Administration-approved drug, could reduce the pathology and improve cardiac function in an animal model for DMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| |
Collapse
|
11
|
Preserved Left Ventricular Function despite Myocardial Fibrosis and Myopathy in the Dystrophin-Deficient D2.B10-Dmdmdx/J Mouse. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5362115. [PMID: 35340200 PMCID: PMC8942668 DOI: 10.1155/2022/5362115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 11/18/2022]
Abstract
Duchenne muscular dystrophy involves an absence of dystrophin, a cytoskeletal protein which supports cell structural integrity and scaffolding for signalling molecules in myocytes. Affected individuals experience progressive muscle degeneration that leads to irreversible loss of ambulation and respiratory diaphragm function. Although clinical management has greatly advanced, heart failure due to myocardial cell loss and fibrosis remains the major cause of death. We examined cardiac morphology and function in D2.B10-Dmdmdx/J (D2-mdx) mice, a relatively new mouse model of muscular dystrophy, which we compared to their wild-type background DBA/2J mice (DBA/2). We also tested whether drug treatment with a specific blocker of mitochondrial permeability transition pore opening (Debio-025), or ACE inhibition (Perindopril), had any effect on dystrophy-related cardiomyopathy. D2-mdx mice were treated for six weeks with Vehicle control, Debio-025 (20 mg/kg/day), Perindopril (2 mg/kg/day), or a combination (n = 8/group). At 18 weeks, compared to DBA/2, D2-mdx hearts displayed greater ventricular collagen, lower cell density, greater cell diameter, and greater protein expression levels of IL-6, TLR4, BAX/Bcl2, caspase-3, PGC-1α, and notably monoamine oxidases A and B. Remarkably, these adaptations in D2-mdx mice were associated with preserved resting left ventricular function similar to DBA/2 mice. Compared to vehicle, although Perindopril partly attenuated the increase in heart weight and collagen at 18 weeks, the drug treatments had no marked impact on dystrophic cardiomyopathy.
Collapse
|
12
|
He X, Liu J, Gu F, Chen J, Lu YW, Ding J, Guo H, Nie M, Kataoka M, Lin Z, Hu X, Chen H, Liao X, Dong Y, Min W, Deng ZL, Pu WT, Huang ZP, Wang DZ. Cardiac CIP protein regulates dystrophic cardiomyopathy. Mol Ther 2022; 30:898-914. [PMID: 34400329 PMCID: PMC8822131 DOI: 10.1016/j.ymthe.2021.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/24/2021] [Accepted: 08/08/2021] [Indexed: 02/04/2023] Open
Abstract
Heart failure is a leading cause of fatality in Duchenne muscular dystrophy (DMD) patients. Previously, we discovered that cardiac and skeletal-muscle-enriched CIP proteins play important roles in cardiac function. Here, we report that CIP, a striated muscle-specific protein, participates in the regulation of dystrophic cardiomyopathy. Using a mouse model of human DMD, we found that deletion of CIP leads to dilated cardiomyopathy and heart failure in young, non-syndromic mdx mice. Conversely, transgenic overexpression of CIP reduces pathological dystrophic cardiomyopathy in old, syndromic mdx mice. Genome-wide transcriptome analyses reveal that molecular pathways involving fibrogenesis and oxidative stress are affected in CIP-mediated dystrophic cardiomyopathy. Mechanistically, we found that CIP interacts with dystrophin and calcineurin (CnA) to suppress the CnA-Nuclear Factor of Activated T cells (NFAT) pathway, which results in decreased expression of Nox4, a key component of the oxidative stress pathway. Overexpression of Nox4 accelerates the development of dystrophic cardiomyopathy in mdx mice. Our study indicates CIP is a modifier of dystrophic cardiomyopathy and a potential therapeutic target for this devastating disease.
Collapse
Affiliation(s)
- Xin He
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Fei Gu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jinghai Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jian Ding
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Haipeng Guo
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Biology, Nanjing Normal University, Nanjing, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhong-Liang Deng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
13
|
Cardiac Complications of Neuromuscular Disorders. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Stalens C, Motté L, Béhin A, Ben Yaou R, Leturcq F, Bassez G, Laforêt P, Fontaine B, Ederhy S, Masingue M, Saadi M, Louis SL, Berber N, Stojkovic T, Duboc D, Wahbi K. Improved Cardiac Outcomes by Early Treatment with Angiotensin-Converting Enzyme Inhibitors in Becker Muscular Dystrophy. J Neuromuscul Dis 2021; 8:495-502. [PMID: 33814458 PMCID: PMC8385526 DOI: 10.3233/jnd-200620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: The latest practice guidelines from the American College of Cardiology/American Heart Association recommend the prescription of an ACE-i for patients presenting with non-ischemic cardiomyopathy when left ventricular ejection fraction (LVEF) falls below 40%. Objective: To determine if the initiation of treatment with an angiotensin-converting enzyme inhibitor (ACE-i) earlier than recommended by practice guidelines issued by professional societies improves the long-term cardiac outcomes of patients presenting with Becker muscular dystrophy (MD) cardiomyopathy. Methods: From a multicenter registry of Becker MD, we selected retrospectively patients presenting between January 1990 and April 2019 with a LVEF ≥40 and ≤49%. We used a propensity score analysis to compare the risk of a) hospitalization for management of heart failure (HF), and b) a decrease in LVEF to <35% in patients who received an ACE-i when LVEF fell below 40% (conventional treatment), versus below 50% (early treatment). Results: From the 183 patients entered in our registry, we identified 85 whose LVEF was between 40 and 49%, 51 of whom received early and 34 received conventional ACE-i treatment. Among patients with early versus conventional treatments, 2 (3.9%) versus 4 (11.8%) were hospitalized for management of HF [hazard ratio (HR) 0.151; 95% confidence interval (CI) 0.028 to 0.822; p = 0.029], and 9 (17.6%) versus 10 (29.4%) had a decrease in LVEF below 35% (HR 0.290; 95% CI 0.121 to 0.694; p = 0.005). Conclusions: The long-term cardiac outcome of patients presenting with Becker MD was significantly better when treatment with ACE-i was introduced after a decrease in LVEF below 50%, instead of below 40% as recommended in the current practice guidelines issued by professional societies.
Collapse
Affiliation(s)
- Caroline Stalens
- Medical Affairs Department, AFM-Téléthon, Evry, France.,INSERM Unit 970, Paris Cardiovascular Research Centre (PARCC), Paris, France
| | - Leslie Motté
- AP-HP, Cochin Hospital, Cardiology Department, Paris, France
| | - Anthony Béhin
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Rabah Ben Yaou
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - France Leturcq
- AP-HP, Cochin Hospital, Department of Genetics and Molecular Biology, Paris, France
| | - Guillaume Bassez
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Pascal Laforêt
- Neurology Department, CHU Paris IdF Ouest-Hôpital Raymond Poincaré, Garches, France
| | - Bertrand Fontaine
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Stéphane Ederhy
- AP-HP, Saint Antoine Hospital, Cardiology Department, Paris, France
| | - Marion Masingue
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Malika Saadi
- AP-HP, Cochin Hospital, Cardiology Department, Paris, France
| | - Sarah Leonard Louis
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Nawal Berber
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Tanya Stojkovic
- AP-HP, Pitié-Salpêtrière Hospital, Reference Center for Muscle Diseases Paris-Est, Myology Institute, Paris, France
| | - Denis Duboc
- AP-HP, Cochin Hospital, Cardiology Department, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Karim Wahbi
- AP-HP, Cochin Hospital, Cardiology Department, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Inserm, UMRS, Paris, France
| |
Collapse
|
15
|
Chen K, Guan Y, Ma Y, Quan D, Zhang J, Liu X, Wu S, Lv L, Zhang G. Modulation of transforming growth factor-beta signaling pathway mediates the effects of Kangxian Formula on cardiac remodeling. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113922. [PMID: 33588010 DOI: 10.1016/j.jep.2021.113922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 01/30/2021] [Accepted: 02/10/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kangxian formula (KXF) is a traditional Chinese medicine which shows effective outcomes in treating cardiac remodeling induced by hypertension. However, the exact effects and the mechanisms involved remain obscure. AIM OF THE STUDY In this study, we aimed to identify the therapeutic role of KXF in vivo and in vitro, and investigate the mechanism of KXF on hypertension induced cardiac remodeling. MATERIALS AND METHODS After quality control of KXF using fingerprint, blood pressure, cardiac structure/function indexes, and degree of myocardial collagen were measured in vivo. Moreover, the proliferation, migration, and fibroblast-to-myofibroblast transformation (FMT) of cardiac fibroblasts (CFBs) were determined. Using gene chip, the related mechanisms of KXF treatment on cardiac remodeling were identified and further validated by western blot and polymerase chain reaction. RESULTS A stable quality control standard of KXF was established in this study. KXF administration ameliorated systolic/diastolic blood pressure, cardiac damages, and cardiac fibrosis in vivo. The proliferation, migration, and FMT of CFBs were also inhibited by the treatment of KXF medicated serum. Furthermore, KXF reduced the protein level of transforming growth factor-beta (TGF-β) receptors Ⅰ, Ⅱ, Tak1, p38, Smad2/3, and Smad4 and the expression of mRNA, which are the hub proteins in the TGF-β signaling pathway. CONCLUSION Our findings suggest that KXF attenuates cardiac remodeling by improving cardiac damages, attenuating cardiac fibrosis, and inhibiting the activity of CFBs. In addition, KXF ameliorates cardiac remodeling partially through modulating the TGF-β signaling pathway. These data provide insights and mechanisms into the wide application of KXF in clinical practice.
Collapse
Affiliation(s)
- Kai Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China; Hong Kong University-Shenzhen Hospital, Shenzhen, China
| | - Yiqing Guan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Yunci Ma
- Southern Medical University Nanfang Hospital, Guangzhou, PR China
| | - Dongling Quan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, PR China
| | - Jingru Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, PR China
| | - Xin Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Shaoyu Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, PR China
| | - Lin Lv
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, PR China.
| | - Guohua Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
16
|
Russo V, Papa AA, Lioncino M, Rago A, Di Fraia F, Palladino A, Politano L, Golino P, Nigro G. Prevalence of atrial fibrillation in myotonic dystrophy type 1: A systematic review. Neuromuscul Disord 2021; 31:281-290. [PMID: 33573883 DOI: 10.1016/j.nmd.2021.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/22/2020] [Accepted: 01/02/2021] [Indexed: 12/26/2022]
Abstract
Cardiac involvement is recorded in about 80% of patients affected by myotonic dystrophy type 1 (DM1). The prevalence of cardiac conduction abnormalities is well described. Data regarding the prevalence of atrial fibrillation (AF) are still conflicting. The primary objective of this review was to assess the prevalence of AF in DM1. The secondary aim was to examine the association of clinical features with AF, to detect predisposing and/or influencing prognosis factors. A systematic search was developed in MEDLINE, EMBASE, Cochrane Register of Controlled Trials and Web of Science databases, to identify original reports between January 1, 2002 and January 30, 2020, assessing the prevalence of AF in DM1 population. Retrospective/prospective cohort studies and case series describing the prevalence of atrial fibrillation evaluated by periodic electrocardiogram (ECG) and/or ECG Holter 24 h, external loop recording (ELR) and implantable devices interrogation in DM1 patients were included. Case reports, simple reviews, commentaries and editorials were excluded. Thirteen reports fulfilled eligibility criteria and were included in our systematic review. According to the results from all the evaluated studies, the mean prevalence of AF in DM1 patients was 10.9% (n = 404) in 3677 DM1 patients. Male sex, conduction defects, echocardiographic findings of prolonged atrial electromechanical delay seem to be strongly associated with atrial fibrillation, representing factors favoring its onset. DM1 patients who develop AF seem to have a higher risk of cardiovascular and non-cardiovascular death. Further studies are needed to assess the prevalence of AF in DM1 patients and to investigate ECG abnormalities and other clinical features associated with this condition.
Collapse
Affiliation(s)
- Vincenzo Russo
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| | - Andrea Antonio Papa
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy.
| | - Michele Lioncino
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| | - Anna Rago
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| | - Francesco Di Fraia
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| | - Alberto Palladino
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paolo Golino
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| | - Gerardo Nigro
- Chair of Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli" Monaldi Hospital, Naples, Italy
| |
Collapse
|
17
|
Palmiero G, Florio MT, Rubino M, Nesti M, Marchel M, Russo V. Cardiac Resynchronization Therapy in Patients with Heart Failure: What is New? Heart Fail Clin 2021; 17:289-301. [PMID: 33673953 DOI: 10.1016/j.hfc.2021.01.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cardiac resynchronization therapy (CRT) is an established treatment of patients with medically refractory, mild-to-severe systolic heart failure (HF), impaired left ventricular function, and wide QRS complex. The pathologic activation sequence observed in patients with abnormal QRS duration and morphology results in a dyssynchronous ventricular activation and contraction leading to cardiac remodeling, worsening systolic and diastolic function, and progressive HF. In this article, the authors aim to explore the current CRT literature, focusing their attentions on the promising innovation in this field.
Collapse
Affiliation(s)
- Giuseppe Palmiero
- Department of Cardiology, AORN Ospedali dei Colli - Monaldi Hospital, Naples, Italy; Inherited and Rare Cardiovascular Diseases Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Maria Teresa Florio
- Division of Internal Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marta Rubino
- Inherited and Rare Cardiovascular Diseases Unit, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Martina Nesti
- Cardiovascular and Neurology Department, Ospedale San Donato, Arezzo, Italy
| | - Michal Marchel
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Vincenzo Russo
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
18
|
Szabó PL, Ebner J, Koenig X, Hamza O, Watzinger S, Trojanek S, Abraham D, Todt H, Kubista H, Schicker K, Remy S, Anegon I, Kiss A, Podesser BK, Hilber K. Cardiovascular phenotype of the Dmdmdx rat - a suitable animal model for Duchenne muscular dystrophy. Dis Model Mech 2021; 14:14/2/dmm047704. [PMID: 33619211 PMCID: PMC7927653 DOI: 10.1242/dmm.047704] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/23/2020] [Indexed: 12/22/2022] Open
Abstract
Besides skeletal muscle abnormalities, Duchenne muscular dystrophy (DMD) patients present with dilated cardiomyopathy development, which considerably contributes to morbidity and mortality. Because the mechanisms responsible for the cardiac complications in the context of DMD are largely unknown, evidence-based therapy approaches are still lacking. This has increased the need for basic research efforts into animal models for DMD. Here, we characterized in detail the cardiovascular abnormalities of Dmdmdx rats, with the aim of determining the suitability of this recently established dystrophin-deficient small animal as a model for DMD. Various methods were applied to compare cardiovascular properties between wild-type and Dmdmdx rats, and to characterize the Dmdmdx cardiomyopathy. These methods comprised echocardiography, invasive assessment of left ventricular hemodynamics, examination of adverse remodeling and endothelial cell inflammation, and evaluation of vascular function, employing wire myography. Finally, intracellular Ca2+ transient measurements, and recordings of currents through L-type Ca2+ channels were performed in isolated single ventricular cardiomyocytes. We found that, similar to respective observations in DMD patients, the hearts of Dmdmdx rats show significantly impaired cardiac function, fibrosis and inflammation, consistent with the development of a dilated cardiomyopathy. Moreover, in Dmdmdx rats, vascular endothelial function is impaired, which may relate to inflammation and oxidative stress, and Ca2+ handling in Dmdmdx cardiomyocytes is abnormal. These findings indicate that Dmdmdx rats represent a promising small-animal model to elucidate mechanisms of cardiomyopathy development in the dystrophic heart, and to test mechanism-based therapies aiming to combat cardiovascular complications in DMD. Summary: We characterized the cardiovascular abnormalities of Dmdmdx rats, demonstrating that Dmdmdx rats show similar cardiac and vascular endothelial function impairments to Duchenne muscular dystrophy patients, representing a model of the dystrophic heart.
Collapse
Affiliation(s)
- Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Vienna 1090, Austria
| | - Janine Ebner
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Xaver Koenig
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Ouafa Hamza
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Vienna 1090, Austria
| | - Simon Watzinger
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Vienna 1090, Austria
| | - Sandra Trojanek
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Hannes Todt
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Helmut Kubista
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Klaus Schicker
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Séverine Remy
- INSERM, Center for Research in Transplantation and Immunology, UMR 1064, Nantes Université, F-44000 Nantes, France
| | - Ignacio Anegon
- INSERM, Center for Research in Transplantation and Immunology, UMR 1064, Nantes Université, F-44000 Nantes, France
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Vienna 1090, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research, Medical University of Vienna, Vienna 1090, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology and Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| |
Collapse
|
19
|
Oreto L, Vita GL, Mandraffino G, Carerj S, Calabrò MP, Manganaro R, Cusmà-Piccione M, Todaro MC, Sframeli M, Cinquegrani M, Toscano A, Vita G, Messina S, Zito C. Impaired myocardial strain in early stage of Duchenne muscular dystrophy: its relation with age and motor performance. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2020; 39:191-199. [PMID: 33458574 PMCID: PMC7783425 DOI: 10.36185/2532-1900-022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 01/02/2023]
Abstract
Duchenne muscular dystrophy (DMD) is complicated by an early and progressive left ventricular (LV) dysfunction. Despite the reduction of ejection fraction (EF) usually manifests in the second decade, subtle alterations in LV mechanics can be detected earlier. Longitudinal and circumferential LV deformation, evaluated by speckle tracking echocardiography (STE), are considered sensitive markers of early dysfunction. We retrospectively examined clinical and echocardiographic data of 32 DMD children with preserved LV function. According to the median age, patients were then divided into younger and older than 9 years, and compared to 24 age-matched healthy subjects. Six-minute-walk test (6MWT), North Star Ambulatory Assessment (NSAA), and a comprehensive cardiac evaluation were performed. Although EF was within the normal range, DMD patients had significantly lower values than healthy controls, and the same occurred for the remaining conventional systolic and diastolic indices. Global longitudinal strain (GLS) was reduced in all patients (older and younger, both p < 0.001). Global circumferential strain (GCS) was reduced only in older patients (< 0.001). Both GLS and GCS worsened with age in DMD patients (GLS p = 0.005; GCS p = 0.024). GLS was significantly worse in the apical segments and in the postero-lateral wall. GCS in the antero-septal, anterior and antero-lateral segments was significantly reduced in older patients, with a prevalent involvement of the sole septal wall in the younger boys. 6MWT appeared to be correlated inversely to GLS and directly to EF. A longitudinal evaluation should be scheduled in DMD boys to assess the global cardiac performance over time and to evaluate the impact of therapies.
Collapse
Affiliation(s)
- Lilia Oreto
- Mediterranean Pediatric Cardiologic Centre, S. Vincenzo Hospital, Taormina - "Bambin Gesù", Rome, Italy.,Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Gian Luca Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina University Hospital, Messina, Italy
| | - Giuseppe Mandraffino
- Internal Medicine Unit, Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Scipione Carerj
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maria Pia Calabrò
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age, University of Messina, Italy
| | - Roberta Manganaro
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maurizio Cusmà-Piccione
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maria Chiara Todaro
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Cardiology Unit, Papardo Hospital, Messina, Italy
| | - Maria Sframeli
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina University Hospital, Messina, Italy
| | - Maria Cinquegrani
- Internal Medicine Unit, Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Antonio Toscano
- Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Giuseppe Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina University Hospital, Messina, Italy.,Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Sonia Messina
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina University Hospital, Messina, Italy.,Unit of Neurology and Neuromuscular Disorders, Department of Clinical and Experimental Medicine, University of Messina, Italy
| | - Concetta Zito
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
20
|
Di Toro A, Giuliani L, Serio A, Arbustini E. Diagnostic Criteria of Left Ventricular Dysfunction in Patients With Myotonic Dystrophy Type 1. J Card Fail 2020; 26:857-859. [DOI: 10.1016/j.cardfail.2019.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/05/2019] [Accepted: 09/13/2019] [Indexed: 11/30/2022]
|
21
|
Paldino A, De Angelis G, Dal Ferro M, Faganello G, Porcari A, Barbati G, Korcova R, Gentile P, Artico J, Cannatà A, Gigli M, Pinamonti B, Merlo M, Sinagra G. High prevalence of subtle systolic and diastolic dysfunction in genotype-positive phenotype-negative relatives of dilated cardiomyopathy patients. Int J Cardiol 2020; 324:108-114. [PMID: 32949639 DOI: 10.1016/j.ijcard.2020.09.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/13/2020] [Accepted: 09/10/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The early diagnosis of genetically determined dilated cardiomyopathy (DCM) could improve the prognosis in mutation carriers. Left ventricular global longitudinal strain (LV GLS) and peak left atrial longitudinal strain (PALS) are promising techniques for the detection of subtle systolic and diastolic dysfunction. We sought to evaluate the prevalence of subtle systolic and diastolic dysfunction by LV GLS and PALS in a cohort of genotype-positive phenotype-negative (GPFN) DCM relatives. METHODS AND RESULTS In this retrospective study, we analyzed echocardiograms of forty-one GPFN relatives of DCM patients. They were compared with age and sex matched healthy individuals (control group). Reduced LV GLS and PALS were defined as >18% and <23.1%, respectively. GPFN relatives (37 ± 14 years, 48.8% male) and controls were similar according to standard echocardiographic measurements. Conversely, LV GLS was -18.8 ± 2.7% in the GPFN group vs. -24.0 ± 1.8% in the control group (p < 0.001). Twenty subjects (48.8%) in the GPFN group and no subjects in the control group had a reduced LV GLS. PALS was 29.2 ± 6.7% in the GPFN group vs. 40.8 ± 8.5% in the control group (p < 0.001). Seven subjects (18.4%) in the GPFN group and one (2%) in the control group had a reduced PALS. A cohort of 17 genotype-negative phenotype-negative relatives showed higher values of LV GLS compared to GPFN. CONCLUSIONS Despite standard echocardiographic parameters are within the normal range, LV GLS and PALS are lower in GPFN relatives of DCM patients when compared to healthy individuals, suggesting a consistent proportion of subtle systolic and diastolic dysfunction in this population.
Collapse
Affiliation(s)
- Alessia Paldino
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Giulia De Angelis
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Matteo Dal Ferro
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Giorgio Faganello
- Cardiovascular Center, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Aldostefano Porcari
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Giulia Barbati
- Biostatistics Unit, Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Renata Korcova
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Piero Gentile
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Jessica Artico
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Antonio Cannatà
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Marta Gigli
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Bruno Pinamonti
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Marco Merlo
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy.
| | - Gianfranco Sinagra
- Cardiothoracic Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| |
Collapse
|
22
|
Abstract
Cardiac fibrosis is associated with non-ischemic dilated cardiomyopathy, increasing its morbidity and mortality. Cardiac fibroblast is the keystone of fibrogenesis, being activated by numerous cellular and humoral factors. Macrophages, CD4+ and CD8+ T cells, mast cells, and endothelial cells stimulate fibrogenesis directly by activating cardiac fibroblasts and indirectly by synthetizing various profibrotic molecules. The synthesis of type 1 and type 3 collagen, fibronectin, and α-smooth muscle actin is rendered by various mechanisms like transforming growth factor-beta/small mothers against decapentaplegic pathway, renin angiotensin system, and estrogens, which in turn alter the extracellular matrix. Investigating the underlying mechanisms will allow the development of diagnostic and prognostic tools and discover novel specific therapies. Serum biomarkers aid in the diagnosis and tracking of cardiac fibrosis progression. The diagnostic gold standard is cardiac magnetic resonance with gadolinium administration that allows quantification of cardiac fibrosis either by late gadolinium enhancement assessment or by T1 mapping. Therefore, the goal is to stop and even reverse cardiac fibrosis by developing specific therapies that directly target fibrogenesis, in addition to the drugs used to treat heart failure. Cardiac resynchronization therapy had shown to revert myocardial remodeling and to reduce cardiac fibrosis. The purpose of this review is to provide an overview of currently available data.
Collapse
|
23
|
Valle-Tenney R, Rebolledo D, Acuña MJ, Brandan E. HIF-hypoxia signaling in skeletal muscle physiology and fibrosis. J Cell Commun Signal 2020; 14:147-158. [PMID: 32088838 DOI: 10.1007/s12079-020-00553-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia refers to the decrease in oxygen tension in the tissues, and the central effector of the hypoxic response is the transcription factor Hypoxia-Inducible Factor α (HIF1-α). Transient hypoxia in acute events, such as exercising or regeneration after damage, play an important role in skeletal muscle physiology and homeostasis. However, sustained activation of hypoxic signaling is a feature of skeletal muscle injury and disease, which can be a consequence of chronic damage but can also increase the severity of the pathology and worsen its outcome. Here, we review evidence that supports the idea that hypoxia and HIF-1α can contribute to the establishment of fibrosis in skeletal muscle through its crosstalk with other profibrotic factors, such as Transforming growth factor β (TGF-β), the induction of profibrotic cytokines expression, as is the case of Connective Tissue Growth Factor (CTGF/CCN2), or being the target of the Renin-angiotensin system (RAS).
Collapse
Affiliation(s)
- Roger Valle-Tenney
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile. .,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Fundación Ciencia & Vida, Santiago, Chile. .,Department Cell and Molecular Biology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Bernasconi P, Carboni N, Ricci G, Siciliano G, Politano L, Maggi L, Mongini T, Vercelli L, Rodolico C, Biagini E, Boriani G, Ruggiero L, Santoro L, Schena E, Prencipe S, Evangelisti C, Pegoraro E, Morandi L, Columbaro M, Lanzuolo C, Sabatelli P, Cavalcante P, Cappelletti C, Bonne G, Muchir A, Lattanzi G. Elevated TGF β2 serum levels in Emery-Dreifuss Muscular Dystrophy: Implications for myocyte and tenocyte differentiation and fibrogenic processes. Nucleus 2019; 9:292-304. [PMID: 29693488 PMCID: PMC5973167 DOI: 10.1080/19491034.2018.1467722] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Among rare diseases caused by mutations in LMNA gene, Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B are characterized by muscle weakness and wasting, joint contractures, cardiomyopathy with conduction system disorders. Circulating biomarkers for these pathologies have not been identified. Here, we analyzed the secretome of a cohort of patients affected by these muscular laminopathies in the attempt to identify a common signature. Multiplex cytokine assay showed that transforming growth factor beta 2 (TGF β2) and interleukin 17 serum levels are consistently elevated in the vast majority of examined patients, while interleukin 6 and basic fibroblast growth factor are altered in subgroups of patients. Levels of TGF β2 are also increased in fibroblast and myoblast cultures established from patient biopsies as well as in serum from mice bearing the H222P Lmna mutation causing Emery-Dreifuss Muscular Dystrophy in humans. Both patient serum and fibroblast conditioned media activated a TGF β2-dependent fibrogenic program in normal human myoblasts and tenocytes and inhibited myoblast differentiation. Consistent with these results, a TGF β2 neutralizing antibody avoided fibrogenic marker activation and myogenesis impairment. Cell intrinsic TGF β2-dependent mechanisms were also determined in laminopathic cells, where TGF β2 activated AKT/mTOR phosphorylation. These data show that TGF β2 contributes to the pathogenesis of Emery-Dreifuss Muscular Dystrophy type 2 and Limb-Girdle muscular Dystrophy 1B and can be considered a potential biomarker of those diseases. Further, the evidence of TGF β2 pathogenetic effects in tenocytes provides the first mechanistic insight into occurrence of joint contractures in muscular laminopathies.
Collapse
Affiliation(s)
- Pia Bernasconi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Nicola Carboni
- b Neurology Department , Hospital San Francesco of Nuoro , Nuoro , Italy
| | - Giulia Ricci
- c Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Gabriele Siciliano
- c Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Luisa Politano
- d Cardiomyology and Medical Genetics, Department of Experimental Medicine , Campania University "Luigi Vanvitelli" (former denomination: Second University of Naples) , Italy
| | - Lorenzo Maggi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Tiziana Mongini
- e Department of Neurosciences "Rita Levi Montalcini" , University of Turin , Turin , Italy
| | - Liliana Vercelli
- e Department of Neurosciences "Rita Levi Montalcini" , University of Turin , Turin , Italy
| | - Carmelo Rodolico
- f Institute of Applied Sciences and Intelligent Systems "ISASI Edoardo Caianello", National Research Council of Italy , Messina , Italy
| | - Elena Biagini
- g Istituto di Cardiologia, Università di Bologna, Policlinico S.Orsola-Malpighi , Bologna , Italy
| | - Giuseppe Boriani
- h Cardiology Division, Department of Diagnostics , Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Policlinico di Modena , Modena , Italy
| | - Lucia Ruggiero
- i Department of Neurosciences , Odontostomatological and Reproductive Sciences, University of Naples "Federico II" , Naples , Italy
| | - Lucio Santoro
- i Department of Neurosciences , Odontostomatological and Reproductive Sciences, University of Naples "Federico II" , Naples , Italy
| | - Elisa Schena
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Sabino Prencipe
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Camilla Evangelisti
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Elena Pegoraro
- l Department of Neurosciences , Neuromuscular Center, University of Padova , Padova , Italy
| | - Lucia Morandi
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Marta Columbaro
- k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Chiara Lanzuolo
- m Istituto Nazionale di Genetica Molecolare "Romeo and Enrica Invernizzi" , Milan , Italy.,n Institute of Cell Biology and Neurobiology, IRCCS Santa Lucia Foundation , Rome , Italy
| | - Patrizia Sabatelli
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| | - Paola Cavalcante
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Cristina Cappelletti
- a Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit , Foundation IRCCS Neurological Institute "Carlo Besta" , Milan , Italy
| | - Gisèle Bonne
- o Sorbonne Universités , UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière , Paris Cedex 13, France
| | - Antoine Muchir
- o Sorbonne Universités , UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Institut de Myologie, G.H. Pitié Salpêtrière , Paris Cedex 13, France
| | - Giovanna Lattanzi
- j Institute of Molecular Genetics (IGM)-CNR, Unit of Bologna , Bologna , Italy.,k Laboratory of Musculoskeletal Cell Biology , Rizzoli Orthopaedic Institute , Bologna , Italy
| |
Collapse
|
25
|
Meyers TA, Townsend D. Cardiac Pathophysiology and the Future of Cardiac Therapies in Duchenne Muscular Dystrophy. Int J Mol Sci 2019; 20:E4098. [PMID: 31443395 PMCID: PMC6747383 DOI: 10.3390/ijms20174098] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/12/2019] [Accepted: 08/19/2019] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating disease featuring skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. Historically, respiratory failure has been the leading cause of mortality in DMD, but recent improvements in symptomatic respiratory management have extended the life expectancy of DMD patients. With increased longevity, the clinical relevance of heart disease in DMD is growing, as virtually all DMD patients over 18 year of age display signs of cardiomyopathy. This review will focus on the pathophysiological basis of DMD in the heart and discuss the therapeutic approaches currently in use and those in development to treat dystrophic cardiomyopathy. The first section will describe the aspects of the DMD that result in the loss of cardiac tissue and accumulation of fibrosis. The second section will discuss cardiac small molecule therapies currently used to treat heart disease in DMD, with a focus on the evidence supporting the use of each drug in dystrophic patients. The final section will outline the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, or repair. There are several new and promising therapeutic approaches that may protect the dystrophic heart, but their limitations suggest that future management of dystrophic cardiomyopathy may benefit from combining gene-targeted therapies with small molecule therapies. Understanding the mechanistic basis of dystrophic heart disease and the effects of current and emerging therapies will be critical for their success in the treatment of patients with DMD.
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
26
|
Renin-angiotensin system in osteoarthritis: A new potential therapy. Int Immunopharmacol 2019; 75:105796. [PMID: 31408841 DOI: 10.1016/j.intimp.2019.105796] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/26/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is one of the most common chronic joint diseases. However, the mechanism remains unclear. The traditional renin-angiotensin system (RAS) is an important system for regulating homeostasis and controlling balance. In recent years, RAS-related components have played an important role in the occurrence of OA. The purpose of this review is to summarize the research results of RAS-related components that are associated with OA. This study systematically searched e-medical databases such as PubMed, Embase, Medline, and Web of Science. The search targets included English publications describing the effects of RAS-related components in OA, including the role of renin, angiotensin-converting enzyme (ACE), Angiotensin II (Ang II), and angiotensin receptor (ATR). Additionally, this study summarizes the potential pathways for RAS-related components to intervene in OA. This study found that RAS-related components including renin, ACE, Ang II, AT1R and AT2R are involved in inflammation and chondrocyte hypertrophy in OA. RAS is involved in signaling pathways including the NF-κB, JNK, VEGFR/Tie-2, and the Axna2/Axna2R axis ones, which may be potential targets for the treatment of OA. Although there are few studies on RAS in the field of OA, the pathogenic effect of RAS-related components is still an important topic in OA treatment, and great progress may be made in this aspect in future studies.
Collapse
|
27
|
Meyers TA, Heitzman JA, Krebsbach AM, Aufdembrink LM, Hughes R, Bartolomucci A, Townsend D. Acute AT 1R blockade prevents isoproterenol-induced injury in mdx hearts. J Mol Cell Cardiol 2019; 128:51-61. [PMID: 30664850 DOI: 10.1016/j.yjmcc.2019.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 12/31/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked disease characterized by skeletal muscle degeneration and a significant cardiomyopathy secondary to cardiomyocyte damage and myocardial loss. The molecular basis of DMD lies in the absence of the protein dystrophin, which plays critical roles in mechanical membrane integrity and protein localization at the sarcolemma. A popular mouse model of DMD is the mdx mouse, which lacks dystrophin and displays mild cardiac and skeletal pathology that can be exacerbated to advance the disease state. In clinical and pre-clinical studies of DMD, angiotensin signaling pathways have emerged as therapeutic targets due to their adverse influence on muscle remodeling and oxidative stress. Here we aim to establish a physiologically relevant cardiac injury model in the mdx mouse, and determine whether acute blockade of the angiotensin II type 1 receptor (AT1R) may be utilized for prevention of dystrophic injury. METHODS AND RESULTS A single IP injection of isoproterenol (Iso, 10 mg/kg) was used to induce cardiac stress and injury in mdx and wild type (C57Bl/10) mice. Mice were euthanized 8 h, 30 h, 1 week, or 1 month following the injection, and hearts were harvested for injury evaluation. At 8 and 30 h post-injury, mdx hearts showed 2.2-fold greater serum cTnI content and 3-fold more extensive injury than wild type hearts. Analysis of hearts 1 week and 1 month after injury revealed significantly higher fibrosis in mdx hearts, with a more robust and longer-lasting immune response compared to wild type hearts. In the 30-hour group, losartan treatment initiated 1 h before Iso injection protected dystrophic hearts from cardiac damage, reducing mdx acute injury area by 2.8-fold, without any significant effect on injury in wild type hearts. However, both wild type and dystrophic hearts showed a 2-fold reduction in the magnitude of the macrophage response to injury 30 h after Iso with losartan. CONCLUSIONS This work demonstrates that acute blockade of AT1R has the potential for robust injury prevention in a model of Iso-induced dystrophic heart injury. In addition to selectively limiting dystrophic cardiac damage, blocking AT1R may serve to limit the inflammatory nature of the immune response to injury in all hearts. Our findings strongly suggest that earlier adoption of angiotensin receptor blockers in DMD patients could limit myocardial damage and subsequent cardiomyopathy.
Collapse
MESH Headings
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Cardiomyopathies/drug therapy
- Cardiomyopathies/genetics
- Cardiomyopathies/pathology
- Dystrophin/genetics
- Heart/drug effects
- Heart/physiopathology
- Humans
- Isoproterenol/pharmacology
- Losartan/pharmacology
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Receptor, Angiotensin, Type 1/genetics
- Sarcolemma/metabolism
- Sarcolemma/pathology
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jackie A Heitzman
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Aimee M Krebsbach
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Lauren M Aufdembrink
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert Hughes
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
28
|
Corey SJ, Jha J, McCart EA, Rittase WB, George J, Mattapallil JJ, Mehta H, Ognoon M, Bylicky MA, Summers TA, Day RM. Captopril mitigates splenomegaly and myelofibrosis in the Gata1 low murine model of myelofibrosis. J Cell Mol Med 2018; 22:4274-4282. [PMID: 29971909 PMCID: PMC6111823 DOI: 10.1111/jcmm.13710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/05/2018] [Indexed: 01/06/2023] Open
Abstract
Allogeneic stem cell transplantation is currently the only curative therapy for primary myelofibrosis (MF), while the JAK2 inhibitor, ruxolitinib. Has been approved only for palliation. Other therapies are desperately needed to reverse life-threatening MF. However, the cell(s) and cytokine(s) that promote MF remain unclear. Several reports have demonstrated that captopril, an inhibitor of angiotensin-converting enzyme that blocks the production of angiotensin II (Ang II), mitigates fibrosis in heart, lung, skin and kidney. Here, we show that captopril can mitigate the development of MF in the Gata1low mouse model of primary MF. Gata1low mice were treated with 79 mg/kg/d captopril in the drinking water from 10 to 12 months of age. At 13 months of age, bone marrows were examined for fibrosis, megakaryocytosis and collagen expression; spleens were examined for megakaryocytosis, splenomegaly and collagen expression. Treatment of Gata1low mice with captopril in the drinking water was associated with normalization of the bone marrow cellularity; reduced reticulin fibres, splenomegaly and megakaryocytosis; and decreased collagen expression. Our findings suggest that treating with the ACE inhibitors captopril has a significant benefit in overcoming pathological changes associated with MF.
Collapse
Affiliation(s)
- Seth J. Corey
- Division of Pediatric Hematology, Oncology & Stem Cell TransplantationThe Massey Cancer Center at Virginia Commonwealth UniversityRichmondVAUSA
| | - Jyoti Jha
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Elizabeth A. McCart
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - William B. Rittase
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Jeffy George
- Department of MicrobiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Joseph J. Mattapallil
- Department of MicrobiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Hrishikesh Mehta
- Division of Pediatric Hematology, Oncology & Stem Cell TransplantationThe Massey Cancer Center at Virginia Commonwealth UniversityRichmondVAUSA
| | - Mungunsukh Ognoon
- Department of AnesthesiologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Michelle A. Bylicky
- Neuroscience Graduate ProgramUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Thomas A. Summers
- Department of PathologyUniformed Services University of the Health SciencesBethesdaMDUSA
| | - Regina M. Day
- Department of Pharmacology and Molecular TherapeuticsUniformed Services University of the Health SciencesBethesdaMDUSA
| |
Collapse
|
29
|
Taylor MD. Editorial commentary: Preserving myocardium in muscular dystrophy patients using ACE inhibition. Trends Cardiovasc Med 2018; 28:338-339. [PMID: 29574046 DOI: 10.1016/j.tcm.2018.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 10/18/2022]
Affiliation(s)
- Michael D Taylor
- The Heart Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue Cincinnati, OH 45229.
| |
Collapse
|