1
|
Ouaddouh Y, Bouyaddid S, Bazid Z, Ismaili N, El Ouafi N. Early onset development of hypertrophic cardiomyopathy in less than 1 year in a patient with familial Friedrich's ataxia: Case report. Radiol Case Rep 2025; 20:3016-3020. [PMID: 40224233 PMCID: PMC11993149 DOI: 10.1016/j.radcr.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 04/15/2025] Open
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disease characterized by progressive ataxia, dysarthria, sensory loss. While neurological symptoms are prominent, cardiac manifestations significantly contribute to mortality. Cardiomyopathy in Friedreich's disease results from mitochondrial dysfunction, loss of contractile proteins and an accumulation of fibrosis in heart. To better characterize the severity of cardiac involvement, the MICONOS study group developed a classification system categorizing FRDA cardiomyopathy as "no," "mild," "intermediate," "severe." We report an uncommon case of early-onset development of hypertrophic cardiomyopathy (HCM) in a 25-year-old female diagnosed with Friedreich's ataxia (FRDA) at age 12. Through annual cardiac evaluations, no signs of cardiac disease were noted. Until presenting with dyspnea and palpitations. Clinical examination revealed truncal ataxia and dysarthria, but no signs of heart failure. However, a transthoracic echocardiography demonstrated nonobstructive hypertrophic cardiomyopathy with a maximal wall thickness of 20 mm, incomplete anterior systolic motion of the mitral valve, a significant development in less than 1 year after last normal cardiac assessment. Left ventricular systolic function was preserved (ejection fraction 50%). She was prescribed bisoprolol and dapagliflozin, with significant improvement at her latest checkup. Family screening revealed HCM in her 30 year female sibling, who also has FRDA. No cardiac abnormalities were detected in her younger brother or parents. Friedreich's hypertrophic cardiomyopathy has been reported as the most significant cause of mortality, especially among younger patients with early onset disease manifestations.
Collapse
Affiliation(s)
- Yasmine Ouaddouh
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Cardiology, Mohammed VI University Hospital/Mohammed I University Oujda Morocco
| | - Salma Bouyaddid
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Cardiology, Mohammed VI University Hospital/Mohammed I University Oujda Morocco
| | - Zakaria Bazid
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Cardiology, Mohammed VI University Hospital/Mohammed I University Oujda Morocco
- Laboratory of Epidemiology, Clinical Research and Public Health, Faculty of Medicine and Pharmacy, Oujda, Morocco
| | - Nabila Ismaili
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Cardiology, Mohammed VI University Hospital/Mohammed I University Oujda Morocco
- Laboratory of Epidemiology, Clinical Research and Public Health, Faculty of Medicine and Pharmacy, Oujda, Morocco
| | - Noha El Ouafi
- Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
- Department of Cardiology, Mohammed VI University Hospital/Mohammed I University Oujda Morocco
- Laboratory of Epidemiology, Clinical Research and Public Health, Faculty of Medicine and Pharmacy, Oujda, Morocco
| |
Collapse
|
2
|
Cardoso I, Melo M, Brás P, Viegas JM, Almeida I, Nunes S, Custódio I, Trigo C, Laranjo S, Graça R, Cruz Ferreira R, Oliveira M, Aguiar Rosa S, Antunes D. The contribution of genetics to the understanding and management of cardiomyopathies: Part 2. Rev Port Cardiol 2025; 44:321-329. [PMID: 40154602 DOI: 10.1016/j.repc.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 05/19/2024] [Accepted: 11/06/2024] [Indexed: 04/01/2025] Open
Abstract
Cardiomyopathies may present as a manifestation of various inherited syndromes. Recognizing the rarity and diagnostic challenges of syndromic and metabolic cardiomyopathies is crucial, as their identification holds significant implications for targeted treatment and enables the use of specific risk stratification tools. Genetics has assumed a pivotal role in clarifying the pathophysiology of cardiomyopathies, facilitating molecular diagnosis, and enabling effective family screening. The advent of next-generation sequencing has revolutionized genetic testing, enabling cost-effective, high-throughput analyses, facilitating the diagnosis of these rare conditions, and allowing the provision of specific management and therapeutics.
Collapse
Affiliation(s)
- Isabel Cardoso
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal.
| | - Mafalda Melo
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Pedro Brás
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - José Miguel Viegas
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Inês Almeida
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Sofia Nunes
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Inês Custódio
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal
| | - Conceição Trigo
- Serviço de Cardiologia Pediátrica, Centro de Referência de Cardiopatias Congénitas do CHULC, Hospital de Santa Marta, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network Guard-Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Sérgio Laranjo
- Serviço de Cardiologia Pediátrica, Centro de Referência de Cardiopatias Congénitas do CHULC, Hospital de Santa Marta, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network Guard-Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Rafael Graça
- GenoMed - Diagnósticos de Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Rui Cruz Ferreira
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Mário Oliveira
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Centro Clínico Académico de Lisboa, Lisboa, Portugal; Arrhythmology, Pacing and Electrophysiology Unit, Cardiology Service, CHLC, Lisbon, Portugal; Institute of Physiology and CCUL, Faculty of Medicine of Lisbon, Portugal; Cardiology Center, Hospital CUF Tejo, Lisbon, Portugal
| | - Sílvia Aguiar Rosa
- Serviço de Cardiologia, Centro de Referência de Cardiopatias Miocardiopatias do Centro Hospitalar Universitário de Lisboa Central (CHULC), Santa Marta Hospital, CHULC, EPE, Lisboa, Portugal; Member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Centro Clínico Académico de Lisboa, Lisboa, Portugal; CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, Lisboa, Portugal; Centro Clínico Académico de Lisboa, Lisboa, Portugal
| | - Diana Antunes
- Medical Genetics Department, Dona Estefânia Hospital, Central Lisbon Hospital and University Centre, Lisbon, Portugal; GenoMed - Diagnósticos de Medicina Molecular, Instituto de Medicina Molecular, Lisbon, Portugal
| |
Collapse
|
3
|
Ben Zichri- David S, Shkuri L, Ast T. Pulling back the mitochondria's iron curtain. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:6. [PMID: 40052109 PMCID: PMC11879881 DOI: 10.1038/s44324-024-00045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/09/2024] [Indexed: 03/09/2025]
Abstract
Mitochondrial functionality and cellular iron homeostasis are closely intertwined. Mitochondria are biosynthetic hubs for essential iron cofactors such as iron-sulfur (Fe-S) clusters and heme. These cofactors, in turn, enable key mitochondrial pathways, such as energy and metabolite production. Mishandling of mitochondrial iron is associated with a spectrum of human pathologies ranging from rare genetic disorders to common conditions. Here, we review mitochondrial iron utilization and its intersection with disease.
Collapse
Affiliation(s)
| | - Liraz Shkuri
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| | - Tslil Ast
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| |
Collapse
|
4
|
Naghipour S, Corben LA, Hulme AJ, Dottori M, Delatycki MB, Lees JG, Lim SY. Omaveloxolone for the Treatment of Friedreich Ataxia: Efficacy, Safety, and Future Perspectives. Mov Disord 2025; 40:226-230. [PMID: 39559924 DOI: 10.1002/mds.30070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Affiliation(s)
- Saba Naghipour
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Louise A Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, Melbourne University, Parkville, Victoria, Australia
| | - Amy J Hulme
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | - Mirella Dottori
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | - Martin B Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, Melbourne University, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Parkville, Victoria, Australia
| | - Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Department of Medicine and Surgery, University of Melbourne, Parkville, Victoria, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Clayton, Victoria, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Department of Medicine and Surgery, University of Melbourne, Parkville, Victoria, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Clayton, Victoria, Australia
- National Heart Research Institute, National Heart Center, Singapore
| |
Collapse
|
5
|
Hayıroğlu Mİ, Kalenderoğlu K, Gürkan K. Tachycardiomyopathy Treated With Ablation by Using 3D Mapping System in a Patient With Friedreich Ataxia. Pacing Clin Electrophysiol 2025; 48:227-229. [PMID: 39717928 DOI: 10.1111/pace.15125] [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] [Received: 10/28/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024]
Abstract
This case report presents the management of tachycardiomyopathy (TCM) in a patient with Friedreich ataxia, a hereditary disorder characterized by progressive neurodegeneration and associated cardiac complications. The patient exhibited severe tachycardia-induced cardiac dysfunction, complicating the clinical picture due to the overlapping neurological symptoms of Friedreich ataxia. Utilizing a 3D mapping system, catheter ablation was performed to accurately identify and target the arrhythmogenic foci contributing to the patient's TCM. The procedure resulted in significant symptom relief and improvement in cardiac function, underscoring the potential benefits of advanced electrophysiological techniques in managing complex cases. This report highlights the importance of a multidisciplinary approach in diagnosing and treating cardiac manifestations in patients with Friedreich ataxia, as well as the efficacy of 3D mapping technology in guiding successful ablation therapies.
Collapse
Affiliation(s)
- Mert İlker Hayıroğlu
- Department of Cardiology, Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Koray Kalenderoğlu
- Department of Cardiology, Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Kadir Gürkan
- Department of Cardiology, Dr. Siyami Ersek Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
6
|
Zahir H, Murai M, Wu L, Valentine M, Hynes SM. Effect of a Supratherapeutic Dose of Omaveloxolone on the Corrected QT Interval in Healthy Participants: A Randomized, Double-Blind, Placebo- and Active-Controlled, Three-Way Crossover Study. Clin Transl Sci 2025; 18:e70139. [PMID: 39976332 PMCID: PMC11840845 DOI: 10.1111/cts.70139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 02/21/2025] Open
Abstract
Omaveloxolone is approved for the treatment of Friedreich ataxia (FA) in patients aged ≥ 16 years at a dose of 150 mg once daily. This double-blind, randomized, placebo- and active-controlled, three-way crossover, thorough corrected QT interval (QTc) study (NCT05927649) evaluated the effect of supratherapeutic omaveloxolone exposure on QTc to exclude a clinically significant prolongation (defined as > 10 ms). Healthy adults were randomized to one of six sequences of three single oral doses (omaveloxolone 450 mg, placebo, or moxifloxacin 400 mg [open-label positive control]) administered with an FDA high-fat meal. Serial pharmacokinetic blood sampling and time-matched electrocardiogram assessments were performed. The primary endpoint was placebo-corrected change from baseline in QTcF (ΔΔQTcF) following omaveloxolone administration. Secondary endpoints included pharmacokinetic parameters of omaveloxolone and its major plasma metabolites (M17 and M22) and safety. All 30 enrolled participants completed the study. The mean omaveloxolone Cmax was 319 ng/mL in this study (4.5-fold the mean steady-state Cmax [71.5 ng/mL] with the approved dose). The mean QTcF intervals were < 450 ms, and mean changes from baseline were < 10 ms at all timepoints following all doses. The upper limit of the 90% CIs of ΔΔQTcF following omaveloxolone administration was < 10 ms at all timepoints. At the Cmax of omaveloxolone, M17, and M22, alone or combined, the upper limits of the 90% CIs of the model-predicted ΔΔQTcF were all < 10 ms. No safety concerns were identified. Supratherapeutic omaveloxolone exposure that covers the worst-case clinical exposure did not cause a clinically significant QTc prolongation and was generally well tolerated.
Collapse
Affiliation(s)
| | | | - Lucy Wu
- Biogen Inc.CambridgeMassachusettsUSA
| | | | | |
Collapse
|
7
|
Janáky M, Braunitzer G. Syndromic Retinitis Pigmentosa: A Narrative Review. Vision (Basel) 2025; 9:7. [PMID: 39846623 PMCID: PMC11755594 DOI: 10.3390/vision9010007] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
Retinitis pigmentosa (RP) encompasses inherited retinal dystrophies, appearing either as an isolated eye condition or as part of a broader systemic syndrome, known as syndromic RP. In these cases, RP includes systemic symptoms impacting other organs, complicating diagnosis and management. This review highlights key systemic syndromes linked with RP, such as Usher, Bardet-Biedl, and Alström syndromes, focusing on genetic mutations, inheritance, and clinical symptoms. These insights support clinicians in recognizing syndromic RP early. Ocular signs like nystagmus and congenital cataracts may indicate systemic disease, prompting genetic testing. Conversely, systemic symptoms may necessitate eye exams, even if vision symptoms are absent. Understanding the systemic aspects of these syndromes emphasizes the need for multidisciplinary collaboration among ophthalmologists, pediatricians, and other specialists to optimize patient care. The review also addresses emerging genetic therapies aimed at both visual and systemic symptoms, though more extensive studies are required to confirm their effectiveness. Overall, by detailing the genetic and clinical profiles of syndromic RP, this review seeks to aid healthcare professionals in diagnosing and managing these complex conditions more effectively, enhancing patient outcomes through timely, specialized intervention.
Collapse
Affiliation(s)
- Márta Janáky
- Department of Ophthalmology, Szent-Györgyi Albert Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Gábor Braunitzer
- Sztárai Institute, University of Tokaj, 3950 Sárospatak, Hungary;
| |
Collapse
|
8
|
Figueroa F, Salinas L, Thai PN, Montgomery CB, Chiamvimonvat N, Cortopassi G, Dedkova EN. Poincaré plot analysis of electrocardiogram uncovers beneficial effects of omaveloxolone in a mouse model of Friedreich's ataxia. Heart Rhythm 2025:S1547-5271(25)00001-3. [PMID: 39788175 DOI: 10.1016/j.hrthm.2024.12.041] [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: 06/17/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Friedreich's ataxia (FA) is a rare inherited neuromuscular disorder whereby most patients die of lethal cardiomyopathy and arrhythmias. Mechanisms leading to arrhythmic events in patients with FA are poorly understood. OBJECTIVE This study aimed to examine cardiac electrical signal propagation in a mouse model of FA with severe cardiomyopathy and to evaluate effects of omaveloxolone (OMAV), the first Food and Drug Administration-approved therapy. METHODS Cardiac-specific MCK-Cre frataxin knockout (FXN-cKO) mice were used to mimic FA cardiomyopathy. In vivo surface electrocardiogram (ECG) recordings, Western blotting, quantitative real-time polymerase chain reaction analysis, and histochemistry were performed. RESULTS Characteristics like long QT syndrome, interatrial block, and ST-segment abnormalities in patients with FA were identified in FXN-cKO mice. FXN-cKO mice exhibited sexual dimorphism in electrical signal propagation and cardiac structural integrity. Untreated FA males showed increased ventricular propagation intervals, whereas females exhibited delayed atrial propagation. OMAV showed no significant therapeutic effect on average ECG time intervals but improved chamber-specific waveforms when aggregated frequency distributions were analyzed. The J wave was absent in FXN-cKO male mice but reappeared with OMAV treatment. Poincaré plots revealed disparate idiopathic arrhythmias with multi-clustering events in individual mice with high incidence in FXN-cKO males. OMAV treatment reduced multi-clustering events to a single cluster; however, autonomic nervous system dysfunction still remained. CONCLUSION Our study revealed significant electrical propagation disturbances and sexual dimorphism in FXN-cKO mice with severe cardiomyopathy. Poincaré plots identified irregularities in heart rhythm and autonomic nervous system dysfunction. OMAV improved heart function by stabilizing early repolarization and reducing disparate arrhythmias. This work stresses sex-specific ECG interpretations and alternative mathematical approaches for drug testing in FA models.
Collapse
Affiliation(s)
- Francisco Figueroa
- Department of Molecular Biosciences, University of California, Davis, California
| | - Lili Salinas
- Department of Molecular Biosciences, University of California, Davis, California
| | - Phung N Thai
- Department of Internal Medicine, Cardiovascular Medicine, University of California, Davis, California
| | - Claire B Montgomery
- Department of Molecular Biosciences, University of California, Davis, California
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Cardiovascular Medicine, University of California, Davis, California; Department of Veterans Affairs, Northern California Health Care System, Mather, California
| | - Gino Cortopassi
- Department of Molecular Biosciences, University of California, Davis, California
| | - Elena N Dedkova
- Department of Molecular Biosciences, University of California, Davis, California; Department of Basic Sciences, California Northstate University, Elk Grove, California.
| |
Collapse
|
9
|
Kwa FAA, Kendal E. Precision medicine and Friedreich ataxia: promoting equity, beneficence, and informed consent for novel gene therapies. Int J Equity Health 2024; 23:230. [PMID: 39516866 PMCID: PMC11545357 DOI: 10.1186/s12939-024-02318-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Friedreich Ataxia (FA) is an incurable neurodegenerative disease with systemic consequences affecting vital organs including those of the central and peripheral nervous systems. This article will use FA as an example to explore some of the practical and ethical issues emerging in precision medicine for rare diseases. It will first describe the existing management strategies available for FA patients, before considering the potential impact of gene therapy trials on the prevention and treatment of disease symptoms. Finally, ethical considerations will be discussed, including equity of access and managing resource allocation dilemmas; balancing benefits, burdens and harms; and gaining informed consent for novel treatments.
Collapse
Affiliation(s)
- Faith A A Kwa
- School of Health Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Evie Kendal
- School of Health Sciences, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
10
|
Gera P, Oliveira VD, Frishman WH, Aronow WS. Cardiac Manifestations of Hemochromatosis. Cardiol Rev 2024:00045415-990000000-00313. [PMID: 39145627 DOI: 10.1097/crd.0000000000000762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Cardiac hemochromatosis, a consequence of primary or secondary iron-overload conditions, poses a threat to patient health, leading to cardiomyopathy and heart failure. This review aims to compile comprehensive information on cardiac hemochromatosis, elucidating its pathophysiology, clinical presentation, diagnosis, and management strategies. Primary and secondary hemochromatosis, genetic and acquired forms, can result in cardiotoxicity by means of iron dysregulation. Diagnostic tools, including biochemical markers, electrocardiography, echocardiography, and magnetic resonance imaging (MRI), are utilized for early detection as well as long-term monitoring post-treatment. For treatment options, phlebotomy is the standard, but for some patients (such as those with anemia), chelation therapy is an alternative option. Other potential therapies include erythrocytapheresis, calcium channel blockers, and hepcidin-targeted approaches, for which more research is needed to understand cardiac function benefits. With the onset of cardiac symptoms, patient health rapidly deteriorates. Thus, timely intervention to mitigate associated morbidity and mortality by means of screening can promote and prolong patient survival.
Collapse
Affiliation(s)
- Priyanka Gera
- From the Departments of Cardiology and Medicine, New York Medical College, Westchester Medical Center, Valhalla, NY
| | | | | | | |
Collapse
|
11
|
Menon PJ, Yi TX, Moran S, Walsh RA, Murphy SM, Bogdanova-Mihaylova P. Health-Related Quality of Life in Patients with Inherited Ataxia in Ireland. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1328-1337. [PMID: 38010570 DOI: 10.1007/s12311-023-01640-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Inherited cerebellar ataxias (CA) are heterogeneous progressive neurological conditions associated with significant functional limitations. This study aimed to assess the implications of inherited CA on patients' self-reported quality of life (QoL) and impairments in work and activities. 129 individuals with ataxia responded to a survey focused on QoL. Health-related QoL was measured using the RAND 36-Item Short Form Survey. An adaptation of the validated Work Productivity and Activity Impairment questionnaire was used to assess the effect of health on work productivity and ability to perform activities over the past week. Nine percent of respondents were currently employed. Individuals with inherited ataxia experienced significant activity impairment, and 75% required professional or informal care. Health-related quality of life (HRQoL) was significantly worse in all areas for the individuals with inherited ataxia compared with Irish population normative values. Participants with Friedreich's ataxia (n = 56) demonstrated worse physical functioning then those with undetermined ataxia (n = 55). Female gender, younger age at symptom onset, current employment, retirement due to age or ataxia, and living in a long-term care facility were associated with higher sub-scores in different domains of HRQoL, while disease duration correlated with worse physical functioning sub-scores. This study is the first cross-sectional study on HRQoL in patients with inherited ataxia in Ireland. It highlights high rates of unemployment, difficulty with daily activities and physical functioning limitations, which is worse than comparative international studies. Given the limited therapeutic options currently available, optimising HRQoL is an important aspect of managing ataxia.
Collapse
Affiliation(s)
- Poornima Jayadev Menon
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland.
- School of Postgraduate Studies, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Tan Xin Yi
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sharon Moran
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
| | - Richard A Walsh
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | - Sinéad M Murphy
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
12
|
Agdamag AC, Nandar PP, Tang WHW. Advanced Heart Failure Therapies in Neuromuscular Diseases. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2024; 26:255-270. [PMID: 39777119 PMCID: PMC11706575 DOI: 10.1007/s11936-024-01046-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 01/11/2025]
Abstract
Purpose of Review The main objective of this review article is to discuss the prevalence, utilization, and outcomes associated with advanced heart failure therapies among patients with neuromuscular disorders. Recent Findings Neuromuscular disorders often have multisystem involvement with a high prevalence of cardiovascular pathology. With the improvement in management of respiratory related complications, heart failure is now the leading cause of mortality in this patient population. Advanced heart failure therapies with durable left ventricular assist devices and heart transplantation have proven to be feasible and safe treatment options in selected patients. Summary Management of neuromuscular disease involves multidisciplinary team involvement given the systemic nature of the disease. Early recognition and close monitoring of these patients will allow for timely initiation of advanced heart failure therapies that can lead to successful outcomes.
Collapse
Affiliation(s)
- Arianne Clare Agdamag
- Section of Advanced Heart Failure and Transplantation Medicine, Department of Cardiovascular Medicine, Miller Family Heart, Vascular and Thoracic Institute, Robert and Suzanne Tomsich, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Phoo Pwint Nandar
- Section of Advanced Heart Failure, Department of Medicine, MetroHealth Medical Center, Cleveland, OH 44109, USA
| | - W. H. Wilson Tang
- Section of Advanced Heart Failure and Transplantation Medicine, Department of Cardiovascular Medicine, Miller Family Heart, Vascular and Thoracic Institute, Robert and Suzanne Tomsich, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
13
|
Perry CE, Halawani SM, Mukherjee S, Ngaba LV, Lieu M, Lee WD, Davis JG, Adzika GK, Bebenek AN, Bazianos DD, Chen B, Mercado-Ayon E, Flatley LP, Suryawanshi AP, Ho I, Rabinowitz JD, Serai SD, Biko DM, Tamaroff J, DeDio A, Wade K, Lin KY, Livingston DJ, McCormack SE, Lynch DR, Baur JA. NAD+ precursors prolong survival and improve cardiac phenotypes in a mouse model of Friedreich's Ataxia. JCI Insight 2024; 9:e177152. [PMID: 39171530 PMCID: PMC11343603 DOI: 10.1172/jci.insight.177152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/12/2024] [Indexed: 08/23/2024] Open
Abstract
Friedreich's ataxia (FRDA) is a progressive disorder caused by insufficient expression of frataxin, which plays a critical role in assembly of iron-sulfur centers in mitochondria. Individuals are cognitively normal but display a loss of motor coordination and cardiac abnormalities. Many ultimately develop heart failure. Administration of nicotinamide adenine dinucleotide-positive (NAD+) precursors has shown promise in human mitochondrial myopathy and rodent models of heart failure, including mice lacking frataxin in cardiomyocytes. We studied mice with systemic knockdown of frataxin (shFxn), which display motor deficits and early mortality with cardiac hypertrophy. Hearts in these mice do not "fail" per se but become hyperdynamic with small chamber sizes. Data from an ongoing natural history study indicate that hyperdynamic hearts are observed in young individuals with FRDA, suggesting that the mouse model could reflect early pathology. Administering nicotinamide mononucleotide or riboside to shFxn mice increases survival, modestly improves cardiac hypertrophy, and limits increases in ejection fraction. Mechanistically, most of the transcriptional and metabolic changes induced by frataxin knockdown are insensitive to NAD+ precursor administration, but glutathione levels are increased, suggesting improved antioxidant capacity. Overall, our findings indicate that NAD+ precursors are modestly cardioprotective in this model of FRDA and warrant further investigation.
Collapse
Affiliation(s)
- Caroline E. Perry
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarah M. Halawani
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sarmistha Mukherjee
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lucie V. Ngaba
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Melissa Lieu
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Won Dong Lee
- Department of Chemistry, Princeton University, Princeton, New Jersey, USA
| | - James G. Davis
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gabriel K. Adzika
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alyssa N. Bebenek
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel D. Bazianos
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Beishan Chen
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Mercado-Ayon
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Liam P. Flatley
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Arjun P. Suryawanshi
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isabelle Ho
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Suraj D. Serai
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology and
| | - David M. Biko
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Radiology and
| | - Jaclyn Tamaroff
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Pediatric Endocrinology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Anna DeDio
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kristin Wade
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kimberly Y. Lin
- Division of Pediatric Cardiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Shana E. McCormack
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David R. Lynch
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph A. Baur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Shen MM, Rummey C, Lynch DR. Phenotypic variation of FXN compound heterozygotes in a Friedreich ataxia cohort. Ann Clin Transl Neurol 2024; 11:1110-1121. [PMID: 38396238 PMCID: PMC11093247 DOI: 10.1002/acn3.52027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Most individuals with Friedreich ataxia (FRDA) have homozygous GAA triplet repeat expansions in the FXN gene, correlating with a typical phenotype of ataxia and cardiomyopathy. A minority are compound heterozygotes carrying a GAA expansion on one allele and a mutation on the other. The study aim was to examine phenotypic variation among compound heterozygotes. METHODS Data on FXN mutations were obtained from the Friedreich Ataxia Clinical Outcome Measures Study (FA-COMS). We compared clinical features in a single-site FA-COMS cohort of 51 compound heterozygous and 358 homozygous patients, including quantitative measures of cardiac, neurologic, and visual disease progression. RESULTS Non-GAA repeat mutations were associated with reduced cardiac disease, and patients with minimal/no function mutations otherwise had a typical FRDA phenotype but with significantly more severe progression. The partial function mutation group was characterized by relative sparing of bulbar and upper limb function, as well as particularly low cardiac involvement. Other clinical features in this group, including optic atrophy and diabetes mellitus, varied widely depending on the specific type of partial function mutation. INTERPRETATION These data support that the typical FRDA phenotype is driven by frataxin deficiency, especially severe in compound heterozygotes with minimal/no function mutations, whereas the heterogeneous presentations of those with partial function mutations may indicate other contributing factors to FRDA pathogenesis.
Collapse
Affiliation(s)
- Megan M. Shen
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| | | | - David R. Lynch
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
Johnston KJA, Cote AC, Hicks E, Johnson J, Huckins LM. Genetically Regulated Gene Expression in the Brain Associated With Chronic Pain: Relationships With Clinical Traits and Potential for Drug Repurposing. Biol Psychiatry 2024; 95:745-761. [PMID: 37678542 PMCID: PMC10924073 DOI: 10.1016/j.biopsych.2023.08.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 07/20/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Chronic pain is a common, poorly understood condition. Genetic studies including genome-wide association studies have identified many relevant variants, which have yet to be translated into full understanding of chronic pain. Transcriptome-wide association studies using transcriptomic imputation methods such as S-PrediXcan can help bridge this genotype-phenotype gap. METHODS We carried out transcriptomic imputation using S-PrediXcan to identify genetically regulated gene expression associated with multisite chronic pain in 13 brain tissues and whole blood. Then, we imputed genetically regulated gene expression for over 31,000 Mount Sinai BioMe participants and performed a phenome-wide association study to investigate clinical relationships in chronic pain-associated gene expression changes. RESULTS We identified 95 experiment-wide significant gene-tissue associations (p < 7.97 × 10-7), including 36 unique genes and an additional 134 gene-tissue associations reaching within-tissue significance, including 53 additional unique genes. Of the 89 unique genes in total, 59 were novel for multisite chronic pain and 18 are established drug targets. Chronic pain genetically regulated gene expression for 10 unique genes was significantly associated with cardiac dysrhythmia, metabolic syndrome, disc disorders/dorsopathies, joint/ligament sprain, anemias, and neurologic disorder phecodes. Phenome-wide association study analyses adjusting for mean pain score showed that associations were not driven by mean pain score. CONCLUSIONS We carried out the largest transcriptomic imputation study of any chronic pain trait to date. Results highlight potential causal genes in chronic pain development and tissue and direction of effect. Several gene results were also drug targets. Phenome-wide association study results showed significant associations for phecodes including cardiac dysrhythmia and metabolic syndrome, thereby indicating potential shared mechanisms.
Collapse
Affiliation(s)
- Keira J A Johnston
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| | - Alanna C Cote
- Pamela Sklar Division of Psychiatric Genetics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Emily Hicks
- Pamela Sklar Division of Psychiatric Genetics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jessica Johnson
- School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Laura M Huckins
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
16
|
Rojsajjakul T, Selvan N, De B, Rosenberg JB, Kaminsky SM, Sondhi D, Janki P, Crystal RG, Mesaros C, Khanna R, Blair IA. Expression and processing of mature human frataxin after gene therapy in mice. Sci Rep 2024; 14:8391. [PMID: 38600238 PMCID: PMC11006666 DOI: 10.1038/s41598-024-59060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/06/2024] [Indexed: 04/12/2024] Open
Abstract
Friedreich's ataxia is a degenerative and progressive multisystem disorder caused by mutations in the highly conserved frataxin (FXN) gene that results in FXN protein deficiency and mitochondrial dysfunction. While gene therapy approaches are promising, consistent induction of therapeutic FXN protein expression that is sub-toxic has proven challenging, and numerous therapeutic approaches are being tested in animal models. FXN (hFXN in humans, mFXN in mice) is proteolytically modified in mitochondria to produce mature FXN. However, unlike endogenous hFXN, endogenous mFXN is further processed into N-terminally truncated, extra-mitochondrial mFXN forms of unknown function. This study assessed mature exogenous hFXN expression levels in the heart and liver of C57Bl/6 mice 7-10 months after intravenous administration of a recombinant adeno-associated virus encoding hFXN (AAVrh.10hFXN) and examined the potential for hFXN truncation in mice. AAVrh.10hFXN induced dose-dependent expression of hFXN in the heart and liver. Interestingly, hFXN was processed into truncated forms, but found at lower levels than mature hFXN. However, the truncations were at different positions than mFXN. AAVrh.10hFXN induced mature hFXN expression in mouse heart and liver at levels that approximated endogenous mFXN levels. These results suggest that AAVrh.10hFXN can likely induce expression of therapeutic levels of mature hFXN in mice.
Collapse
Affiliation(s)
- Teerapat Rojsajjakul
- Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, Penn/CHOP Friedreich's Ataxia Center of Excellence, Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Bishnu De
- Department of Genetic Medicine, Weill Cornell College of Medicine, New York, NY, USA
| | - Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell College of Medicine, New York, NY, USA
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell College of Medicine, New York, NY, USA
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell College of Medicine, New York, NY, USA
| | | | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell College of Medicine, New York, NY, USA
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, Penn/CHOP Friedreich's Ataxia Center of Excellence, Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, Penn/CHOP Friedreich's Ataxia Center of Excellence, Center of Excellence in Environmental Toxicology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Chang JC, Ryan MR, Stark MC, Liu S, Purushothaman P, Bolan F, Johnson CA, Champe M, Meng H, Lawlor MW, Halawani S, Ngaba LV, Lynch DR, Davis C, Gonzalo-Gil E, Lutz C, Urbinati F, Medicherla B, Fonck C. AAV8 gene therapy reverses cardiac pathology and prevents early mortality in a mouse model of Friedreich's ataxia. Mol Ther Methods Clin Dev 2024; 32:101193. [PMID: 38352270 PMCID: PMC10862410 DOI: 10.1016/j.omtm.2024.101193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Friedreich's ataxia (FRDA) is an autosomal-recessive disorder primarily attributed to biallelic GAA repeat expansions that reduce expression of the mitochondrial protein frataxin (FXN). FRDA is characterized by progressive neurodegeneration, with many patients developing cardiomyopathy that progresses to heart failure and death. The potential to reverse or prevent progression of the cardiac phenotype of FRDA was investigated in a mouse model of FRDA, using an adeno-associated viral vector (AAV8) containing the coding sequence of the FXN gene. The Fxnflox/null::MCK-Cre conditional knockout mouse (FXN-MCK) has an FXN gene ablation that prevents FXN expression in cardiac and skeletal muscle, leading to cardiac insufficiency, weight loss, and morbidity. FXN-MCK mice received a single intravenous injection of an AAV8 vector containing human (hFXN) or mouse (mFXN) FXN genes under the control of a phosphoglycerate kinase promoter. Compared to vehicle-treated FXN-MCK control mice, AAV-treated FXN-MCK mice displayed increases in body weight, reversal of cardiac deficits, and increases in survival without apparent toxicity in the heart or liver for up to 12 weeks postdose. FXN protein expression in heart tissue was detected in a dose-dependent manner, exhibiting wide distribution throughout the heart similar to wild type, but more speckled. These results support an AAV8-based approach to treat FRDA-associated cardiomyopathy.
Collapse
Affiliation(s)
- Joshua C. Chang
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Molly R. Ryan
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Marie C. Stark
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Su Liu
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | | | - Fria Bolan
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | | | - Mark Champe
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Hui Meng
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
- Medical College of Wisconsin, Department of Pathology and Laboratory Medicine, Milwaukee, WI 53226, USA
| | - Michael W. Lawlor
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
- Medical College of Wisconsin, Department of Pathology and Laboratory Medicine, Milwaukee, WI 53226, USA
| | - Sarah Halawani
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lucie V. Ngaba
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R. Lynch
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | - Fabrizia Urbinati
- Formerly of Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Bala Medicherla
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Carlos Fonck
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
18
|
Czornobil R, Abou-Assali O, Remily-Wood E, Lynch DR, Noujaim SF, Chidipi B. The Cardiac Calcium Handling Machinery is Remodeled in Friedreich's Ataxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566141. [PMID: 38014032 PMCID: PMC10680642 DOI: 10.1101/2023.11.09.566141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background Friedreich's ataxia (FA) is an inherited neurodegenerative disorder that causes progressive nervous system damage resulting in impaired muscle coordination. FA is the most common autosomal recessive form of ataxia and is caused by an expansion of the DNA triplet guanine-adenine-adenine (GAA) in the first intron of the Frataxin gene (FXN), located on chromosome 9q13. In the unaffected population, the number of GAA repeats ranges from 6 to 27 repetitions. In FA patients, GAA repeat expansions range from 44 to 1,700 repeats which decreases frataxin protein expression. Frataxin is a mitochondrial protein essential for various cellular functions, including iron metabolism. Reduced frataxin expression is thought to negatively affect mitochondrial iron metabolism, leading to increased oxidative damage. Although FA is considered a neurodegenerative disorder, FA patients display heart disease that includes hypertrophy, heart failure, arrhythmias, conduction abnormalities, and cardiac fibrosis. Objective In this work, we investigated whether abnormal Ca 2+ handling machinery is the molecular mechanism that perpetuates cardiac dysfunction in FA. Methods We used the frataxin knock-out (FXN-KO) mouse model of FA as well as human heart samples from donors with FA and from unaffected donors. ECG and echocardiography were used to assess cardiac function in the mice. Expression of calcium handling machinery proteins was assessed with proteomics and western blot. In left ventricular myocytes from FXN-KO and FXN-WT mice, the IonOptix system was used for calcium imaging, the seahorse assay was utilized to measure oxygen consumption rate (OCR), and confocal imaging was used to quantify the mitochondrial membrane potential (Δψm) and reactive oxygen species (ROS). Results We found that major contractile proteins, including SERCA2a and Ryr2, were downregulated in human left ventricular samples from deceased donors with FA compared to unaffected donors, similar to the downregulation of these proteins in the left ventricular tissue from FXN-KO compared to FXN-WT. On the ECG, the RR, PR, QRS, and QTc were significantly longer in the FXN-KO mice compared to FXN-WT. The ejection fraction and fractional shortening were significantly decreased and left ventricular wall thickness and diameter were significantly increased in the FXN-KO mice versus FXN-WT. The mitochondrial membrane potential Δψm was depolarized, ROS levels were elevated, and OCR was decreased in ventricular myocytes from FXN-KO versus FXN-WT. Conclusion The development of left ventricular contractile dysfunction in FA is associated with reduced expression of calcium handling proteins and mitochondrial dysfunction.
Collapse
|
19
|
Kioutchoukova IP, Foster DT, Thakkar RN, Foreman MA, Burgess BJ, Toms RM, Molina Valero EE, Lucke-Wold B. Neurologic orphan diseases: Emerging innovations and role for genetic treatments. World J Exp Med 2023; 13:59-74. [PMID: 37767543 PMCID: PMC10520757 DOI: 10.5493/wjem.v13.i4.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/16/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023] Open
Abstract
Orphan diseases are rare diseases that affect less than 200000 individuals within the United States. Most orphan diseases are of neurologic and genetic origin. With the current advances in technology, more funding has been devoted to developing therapeutic agents for patients with these conditions. In our review, we highlight emerging options for patients with neurologic orphan diseases, specifically including diseases resulting in muscular deterioration, epilepsy, seizures, neurodegenerative movement disorders, inhibited cognitive development, neuron deterioration, and tumors. After extensive literature review, gene therapy offers a promising route for the treatment of neurologic orphan diseases. The use of clustered regularly interspaced palindromic repeats/Cas9 has demonstrated positive results in experiments investigating its role in several diseases. Additionally, the use of adeno-associated viral vectors has shown improvement in survival, motor function, and developmental milestones, while also demonstrating reversal of sensory ataxia and cardiomyopathy in Friedreich ataxia patients. Antisense oligonucleotides have also been used in some neurologic orphan diseases with positive outcomes. Mammalian target of rapamycin inhibitors are currently being investigated and have reduced abnormal cell growth, proliferation, and angiogenesis. Emerging innovations and the role of genetic treatments open a new window of opportunity for the treatment of neurologic orphan diseases.
Collapse
Affiliation(s)
| | - Devon T Foster
- Florida International University Herbert Wertheim College of Medicine, Florida International University Herbert Wertheim College of Medicine, Miami, FL 33199, United States
| | - Rajvi N Thakkar
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Marco A Foreman
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Brandon J Burgess
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | - Rebecca M Toms
- College of Medicine, University of Florida, Gainesville, FL 32611, United States
| | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
20
|
Munoz-Zuluaga C, Gertz M, Yost-Bido M, Greco A, Gorman N, Chen A, Kooner V, Rosenberg JB, De BP, Kaminsky SM, Borczuk A, Ricart Arbona RJ, Martin HR, Monette S, Khanna R, Barth JA, Crystal RG, Sondhi D. Identification of Safe and Effective Intravenous Dose of AAVrh.10hFXN to Treat the Cardiac Manifestations of Friedreich's Ataxia. Hum Gene Ther 2023; 34:605-615. [PMID: 37166361 PMCID: PMC10354731 DOI: 10.1089/hum.2023.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/16/2023] [Indexed: 05/12/2023] Open
Abstract
Friedreich's ataxia (FA) is a life-threatening autosomal recessive disorder characterized by neurological and cardiac dysfunction. Arrhythmias and heart failure are the main cause of premature death. From prior studies in murine models of FA, adeno-associated virus encoding the normal human frataxin gene (AAVrh.10hFXN) effectively treated the cardiac manifestations of the disease. However, the therapeutic dose window is limited by high level of human frataxin (hFXN) gene expression associated with toxicity. As a therapeutic goal, since FA heterozygotes have no clinical manifestations of FA, we estimated the level of frataxin (FXN) necessary to convert the heart of a homozygote to that of a heterozygote. In noncardiac cells, FA heterozygotes have 30-80% of normal FXN levels (17.7-47.2 ng/mg, average 32.5 ng/mg) and FA homozygotes 2-30% normal levels (1.2-17.7 ng/mg, average 9.4 ng/mg). Therefore, an AAV vector would need to augment endogenous in an FA homozygote by >8.3 ng/mg. To determine the required dose of AAVrh.10hFXN, we administered 1.8 × 1011, 5.7 × 1011, or 1.8 × 1012 gc/kg of AAVrh.10hFXN intravenously (IV) to muscle creatine kinase (mck)-Cre conditional knockout Fxn mice, a cardiac and skeletal FXN knockout model. The minimally effective dose was 5.7 × 1011 gc/kg, resulting in cardiac hFXN levels of 6.1 ± 4.2 ng/mg and a mild (p < 0.01 compared with phosphate-buffered saline controls) improvement in mortality. A dose of 1.8 × 1012 gc/kg resulted in cardiac hFXN levels of 33.7 ± 6.4 ng/mg, a significant improvement in ejection fraction and fractional shortening (p < 0.05, both comparisons) and a 21.5% improvement in mortality (p < 0.001). To determine if the significantly effective dose of 1.8 × 1012 gc/kg could achieve human FA heterozygote levels in a large animal, this dose was administered IV to nonhuman primates. After 12 weeks, the vector-expressed FXN in the heart was 17.8 ± 4.9 ng/mg, comparable to the target human levels. These data identify both minimally and significantly effective therapeutic doses that are clinically relevant for the treatment of the cardiac manifestations of FA.
Collapse
Affiliation(s)
| | - Monica Gertz
- Department of Genetic Medicine, New York, New York, USA
| | | | | | | | - Alvin Chen
- Department of Genetic Medicine, New York, New York, USA
| | - Vikrum Kooner
- Department of Genetic Medicine, New York, New York, USA
| | | | - Bishnu P. De
- Department of Genetic Medicine, New York, New York, USA
| | | | - Alain Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, New York, USA
| | - Rodolfo J. Ricart Arbona
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Heather R. Martin
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Sebastien Monette
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | | | | | | | - Dolan Sondhi
- Department of Genetic Medicine, New York, New York, USA
| |
Collapse
|
21
|
Chen PY, Qin L, Simons M. TGFβ signaling pathways in human health and disease. Front Mol Biosci 2023; 10:1113061. [PMID: 37325472 PMCID: PMC10267471 DOI: 10.3389/fmolb.2023.1113061] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
Transforming growth factor beta (TGFβ) is named for the function it was originally discovered to perform-transformation of normal cells into aggressively growing malignant cells. It became apparent after more than 30 years of research, however, that TGFβ is a multifaceted molecule with a myriad of different activities. TGFβs are widely expressed with almost every cell in the human body producing one or another TGFβ family member and expressing its receptors. Importantly, specific effects of this growth factor family differ in different cell types and under different physiologic and pathologic conditions. One of the more important and critical TGFβ activities is the regulation of cell fate, especially in the vasculature, that will be the focus of this review.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
22
|
Lew SY, Mohd Hisam NS, Phang MWL, Syed Abdul Rahman SN, Poh RYY, Lim SH, Kamaruzzaman MA, Chau SC, Tsui KC, Lim LW, Wong KH. Adenosine Improves Mitochondrial Function and Biogenesis in Friedreich's Ataxia Fibroblasts Following L-Buthionine Sulfoximine-Induced Oxidative Stress. BIOLOGY 2023; 12:biology12040559. [PMID: 37106759 PMCID: PMC10136261 DOI: 10.3390/biology12040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
Adenosine is a nucleoside that is widely distributed in the central nervous system and acts as a central excitatory and inhibitory neurotransmitter in the brain. The protective role of adenosine in different pathological conditions and neurodegenerative diseases is mainly mediated by adenosine receptors. However, its potential role in mitigating the deleterious effects of oxidative stress in Friedreich's ataxia (FRDA) remains poorly understood. We aimed to investigate the protective effects of adenosine against mitochondrial dysfunction and impaired mitochondrial biogenesis in L-buthionine sulfoximine (BSO)-induced oxidative stress in dermal fibroblasts derived from an FRDA patient. The FRDA fibroblasts were pre-treated with adenosine for 2 h, followed by 12.50 mM BSO to induce oxidative stress. Cells in medium without any treatments or pre-treated with 5 µM idebenone served as the negative and positive controls, respectively. Cell viability, mitochondrial membrane potential (MMP), aconitase activity, adenosine triphosphate (ATP) level, mitochondrial biogenesis, and associated gene expressions were assessed. We observed disruption of mitochondrial function and biogenesis and alteration in gene expression patterns in BSO-treated FRDA fibroblasts. Pre-treatment with adenosine ranging from 0-600 µM restored MMP, promoted ATP production and mitochondrial biogenesis, and modulated the expression of key metabolic genes, namely nuclear respiratory factor 1 (NRF1), transcription factor A, mitochondrial (TFAM), and NFE2-like bZIP transcription factor 2 (NFE2L2). Our study demonstrated that adenosine targeted mitochondrial defects in FRDA, contributing to improved mitochondrial function and biogenesis, leading to cellular iron homeostasis. Therefore, we suggest a possible therapeutic role for adenosine in FRDA.
Collapse
Affiliation(s)
- Sze Yuen Lew
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | | | - Michael Weng Lok Phang
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | | | - Rozaida Yuen Ying Poh
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siew Huah Lim
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Mohd Amir Kamaruzzaman
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, Kuala Lumpur 56000, Malaysia
| | - Sze Chun Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ka Chun Tsui
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kah Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Angulo MB, Bertalovitz A, Argenziano MA, Yang J, Patel A, Zesiewicz T, McDonald TV. Frataxin deficiency alters gene expression in Friedreich ataxia derived IPSC-neurons and cardiomyocytes. Mol Genet Genomic Med 2022; 11:e2093. [PMID: 36369844 PMCID: PMC9834160 DOI: 10.1002/mgg3.2093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/16/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Friedreich's ataxia (FRDA) is an autosomal recessive disease, whereby homozygous inheritance of an expanded GAA trinucleotide repeat expansion in the first intron of the FXN gene leads to transcriptional repression of the encoded protein frataxin. FRDA is a progressive neurodegenerative disorder, but the primary cause of death is heart disease which occurs in 60% of the patients. Several functions of frataxin have been proposed, but none of them fully explain why its deficiency causes the FRDA phenotypes nor why the most affected cell types are neurons and cardiomyocytes. METHODS To investigate, we generated iPSC-derived neurons (iNs) and cardiomyocytes (iCMs) from an FRDA patient and upregulated FXN expression via lentivirus without altering genomic GAA repeats at the FXN locus. RESULTS RNA-seq and differential gene expression enrichment analyses demonstrated that frataxin deficiency affected the expression of glycolytic pathway genes in neurons and extracellular matrix pathway genes in cardiomyocytes. Genes in these pathways were differentially expressed when compared to a control and restored to control levels when FRDA cells were supplemented with frataxin. CONCLUSIONS These results offer novel insight into specific roles of frataxin deficiency pathogenesis in neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Mariana B. Angulo
- Heart Institute, Morsani College of Medicine, University of South FloridaTampaFloridaUSA,Department of Molecular Pharmacology & PhysiologyMorsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Alexander Bertalovitz
- Heart Institute, Morsani College of Medicine, University of South FloridaTampaFloridaUSA,Department of Medicine (Cardiology)Morsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Mariana A. Argenziano
- Heart Institute, Morsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Jiajia Yang
- Heart Institute, Morsani College of Medicine, University of South FloridaTampaFloridaUSA,Department of Molecular Pharmacology & PhysiologyMorsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Aarti Patel
- Department of Medicine (Cardiology)Morsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Theresa Zesiewicz
- Department of NeurologyMorsani College of Medicine, University of South FloridaTampaFloridaUSA
| | - Thomas V. McDonald
- Heart Institute, Morsani College of Medicine, University of South FloridaTampaFloridaUSA,Department of Molecular Pharmacology & PhysiologyMorsani College of Medicine, University of South FloridaTampaFloridaUSA,Department of Medicine (Cardiology)Morsani College of Medicine, University of South FloridaTampaFloridaUSA
| |
Collapse
|
24
|
Keita M, McIntyre K, Rodden LN, Schadt K, Lynch DR. Friedreich ataxia: clinical features and new developments. Neurodegener Dis Manag 2022; 12:267-283. [PMID: 35766110 PMCID: PMC9517959 DOI: 10.2217/nmt-2022-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Friedreich's ataxia (FRDA), a neurodegenerative disease characterized by ataxia and other neurological features, affects 1 in 50,000-100,000 individuals in the USA. However, FRDA also includes cardiac, orthopedic and endocrine dysfunction, giving rise to many secondary disease characteristics. The multifaceted approach for clinical care has necessitated the development of disease-specific clinical care guidelines. New developments in FRDA include the advancement of clinical drug trials targeting the NRF2 pathway and frataxin restoration. Additionally, a novel understanding of gene silencing in FRDA, reflecting a variegated silencing pattern, will have applications to current and future therapeutic interventions. Finally, new perspectives on the neuroanatomy of FRDA and its developmental features will refine the time course and anatomical targeting of novel approaches.
Collapse
Affiliation(s)
- Medina Keita
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kellie McIntyre
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Layne N Rodden
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kim Schadt
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Poole J, Ray D. The Role of Circadian Clock Genes in Critical Illness: The Potential Role of Translational Clock Gene Therapies for Targeting Inflammation, Mitochondrial Function, and Muscle Mass in Intensive Care. J Biol Rhythms 2022; 37:385-402. [PMID: 35880253 PMCID: PMC9326790 DOI: 10.1177/07487304221092727] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Earth's 24-h planetary rotation, with predictable light and heat cycles, has driven profound evolutionary adaptation, with prominent impacts on physiological mechanisms important for surviving critical illness. Pathways of interest include inflammation, mitochondrial function, energy metabolism, hypoxic signaling, apoptosis, and defenses against reactive oxygen species. Regulation of these by the cellular circadian clock (BMAL-1 and its network) has an important influence on pulmonary inflammation; ventilator-associated lung injury; septic shock; brain injury, including vasospasm; and overall mortality in both animals and humans. Whether it is cytokines, the inflammasome, or mitochondrial biogenesis, circadian medicine represents exciting opportunities for translational therapy in intensive care, which is currently lacking. Circadian medicine also represents a link to metabolic determinants of outcome, such as diabetes and cardiovascular disease. More than ever, we are appreciating the problem of circadian desynchrony in intensive care. This review explores the rationale and evidence for the importance of the circadian clock in surviving critical illness.
Collapse
Affiliation(s)
- Joanna Poole
- Anaesthetics and Critical Care, Gloucestershire Royal Hospital, Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - David Ray
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Tong WH, Ollivierre H, Noguchi A, Ghosh MC, Springer DA, Rouault TA. Hyperactivation of mTOR and AKT in a cardiac hypertrophy animal model of Friedreich ataxia. Heliyon 2022; 8:e10371. [PMID: 36061025 PMCID: PMC9433723 DOI: 10.1016/j.heliyon.2022.e10371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/28/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiomyopathy is a primary cause of death in Friedreich ataxia (FRDA) patients with defective iron-sulfur cluster (ISC) biogenesis due to loss of functional frataxin and in rare patients with functional loss of other ISC biogenesis factors. The mechanistic target of rapamycin (mTOR) and AKT signaling cascades that coordinate eukaryotic cell growth and metabolism with environmental inputs, including nutrients and growth factors, are crucial regulators of cardiovascular growth and homeostasis. We observed increased phosphorylation of AKT and dysregulation of multiple downstream effectors of mTORC1, including S6K1, S6, ULK1 and 4EBP1, in a cardiac/skeletal muscle specific FRDA conditional knockout (cKO) mouse model and in human cell lines depleted of ISC biogenesis factors. Knockdown of several mitochondrial metabolic proteins that are downstream targets of ISC biogenesis, including lipoyl synthase and subunit B of succinate dehydrogenase, also resulted in activation of mTOR and AKT signaling, suggesting that mTOR and AKT hyperactivations are part of the metabolic stress response to ISC deficiencies. Administration of rapamycin, a specific inhibitor of mTOR signaling, enhanced the survival of the Fxn cKO mice, providing proof of concept for the potential of mTOR inhibition to ameliorate cardiac disease in patients with defective ISC biogenesis. However, AKT phosphorylation remained high in rapamycin-treated Fxn cKO hearts, suggesting that parallel mTOR and AKT inhibition might be necessary to further improve the lifespan and healthspan of ISC deficient individuals.
Collapse
Affiliation(s)
- Wing-Hang Tong
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
| | - Hayden Ollivierre
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
| | - Audrey Noguchi
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, Bethesda, MD 20892, United States
| | - Manik C. Ghosh
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, Bethesda, MD 20892, United States
| | - Tracey A. Rouault
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
- Corresponding author.
| |
Collapse
|
27
|
Payne RM. Cardiovascular Research in Friedreich Ataxia: Unmet Needs and Opportunities. JACC Basic Transl Sci 2022; 7:1267-1283. [PMID: 36644283 PMCID: PMC9831864 DOI: 10.1016/j.jacbts.2022.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 01/18/2023]
Abstract
Friedreich Ataxia (FRDA) is an autosomal recessive disease in which a mitochondrial protein, frataxin, is severely decreased in its expression. In addition to progressive ataxia, patients with FRDA often develop a cardiomyopathy that can be hypertrophic. This cardiomyopathy is unlike the sarcomeric hypertrophic cardiomyopathies in that the hypertrophy is associated with massive mitochondrial proliferation within the cardiomyocyte rather than contractile protein overexpression. This is associated with atrial arrhythmias, apoptosis, and fibrosis over time, and patients often develop heart failure leading to premature death. The differences between this mitochondrial cardiomyopathy and the more common contractile protein hypertrophic cardiomyopathies can be a source of misunderstanding in the management of these patients. Although imaging studies have revealed much about the structure and function of the heart in this disease, we still lack an understanding of many important clinical and fundamental molecular events that determine outcome of the heart in FRDA. This review will describe the current basic and clinical understanding of the FRDA heart, and most importantly, identify major gaps in our knowledge that represent new directions and opportunities for research.
Collapse
Affiliation(s)
- R. Mark Payne
- Address for correspondence: Dr R. Mark Payne, Division of Pediatric Cardiology, Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, R4 302b, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
28
|
Recessive cerebellar and afferent ataxias - clinical challenges and future directions. Nat Rev Neurol 2022; 18:257-272. [PMID: 35332317 DOI: 10.1038/s41582-022-00634-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 02/07/2023]
Abstract
Cerebellar and afferent ataxias present with a characteristic gait disorder that reflects cerebellar motor dysfunction and sensory loss. These disorders are a diagnostic challenge for clinicians because of the large number of acquired and inherited diseases that cause cerebellar and sensory neuron damage. Among such conditions that are recessively inherited, Friedreich ataxia and RFC1-associated cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) include the characteristic clinical, neuropathological and imaging features of ganglionopathies, a distinctive non-length-dependent type of sensory involvement. In this Review, we discuss the typical and atypical phenotypes of Friedreich ataxia and CANVAS, along with the features of other recessive ataxias that present with a ganglionopathy or polyneuropathy, with an emphasis on recently described clinical features, natural history and genotype-phenotype correlations. We review the main developments in understanding the complex pathology that affects the sensory neurons and cerebellum, which seem to be most vulnerable to disorders that affect mitochondrial function and DNA repair mechanisms. Finally, we discuss disease-modifying therapeutic advances in Friedreich ataxia, highlighting the most promising candidate molecules and lessons learned from previous clinical trials.
Collapse
|
29
|
Moisa SM, Miron IC, Tarca E, Trandafir L, Lupu VV, Lupu A, Rusu TE. Non-Cardiac Cause of Death in Selected Group Children with Cardiac Pathology: A Retrospective Single Institute Study. CHILDREN 2022; 9:children9030335. [PMID: 35327707 PMCID: PMC8946943 DOI: 10.3390/children9030335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/05/2022]
Abstract
Background: Pediatricians and pediatric surgeons often face children with cardiomegaly and dilatative or hypertrophic cardiomyopathies presenting with or without symptoms. Some of these patients have already been diagnosed and received medication, and some present with completely unrelated pathologies. Methods: We performed a 4-year retrospective study on the causes and mechanisms of death of children with cardiac pathology who died outside the cardiology clinic of our hospital by studying the hospital charts and necropsy reports. All children who were in this situation in our hospital were included. Results: Most children in our study group were infants (81.82%), most were boys (81.82%), and in most cases, the cause or mechanism of death was unrelated to their heart condition, whether it had already been diagnosed or not (one case probably died as a result of a malignant ventricular arrhythmia). Additionally, 27.27% of children died as a consequence of bronchopneumonia, the same percentage died as a consequence of an acquired non-pulmonary disease or after surgery, and 18.18% died as a consequence of congenital malformations. Conclusions: Cardiac disease needs to be thoroughly investigated using multiple tools for all children presenting with heart failure symptoms, those with heart murmurs, and children scheduled for surgery of any type. The intensive care specialist and surgeon need to be aware of any heart pathology before non-cardiac surgical interventions.
Collapse
Affiliation(s)
- Stefana Maria Moisa
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
| | - Ingrith Crenguta Miron
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
| | - Elena Tarca
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
| | - Laura Trandafir
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
| | - Vasile Valeriu Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
- Correspondence: (V.V.L.); (A.L.)
| | - Ancuta Lupu
- Pediatrics Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (S.M.M.); (I.C.M.); (E.T.); (L.T.)
- Correspondence: (V.V.L.); (A.L.)
| | | |
Collapse
|
30
|
Quatrana A, Morini E, Tiano F, Vancheri C, Panarello L, Romano S, Marcotulli C, Casali C, Mariotti C, Mongelli A, Fichera M, Rufini A, Condò I, Novelli G, Testi R, Amati F, Malisan F. Hsa-miR223-3p circulating level is upregulated in Friedreich's ataxia and inversely associated with HCLS1 associated protein X-1, HAX-1. Hum Mol Genet 2022; 31:2010-2022. [PMID: 35015850 DOI: 10.1093/hmg/ddac005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 11/12/2022] Open
Abstract
Frataxin (FXN) deficiency is responsible for Friedreich's ataxia (FRDA) in which, besides the characteristic features of spinocerebellar ataxia, two thirds of patients develop hypertrophic cardiomyopathy that often progresses to heart failure and premature death. Different mechanisms might underlie FRDA pathogenesis. Among them, the role of miRNAs deserves investigations. We carried out a miRNA PCR-array analysis of plasma samples of early-, intermediate- and late-onset FRDA groups, defining a set of 30 differentially expressed miRNAs. Hsa-miR223-3p is the only miRNA shared between the three patient groups and appears upregulated in all of them. The upregulation of hsa-miR223-3p was further validated in all enrolled patients (n = 37, Fc = +2.3; p < 0.0001). Using a Receiver Operating Characteristic (ROC) curve analysis, we quantified the predictive value of circulating hsa-miR223-3p for FRDA, obtaining an AUC (Area Under the ROC Curve) value of 0.835 (p < 0.0001) for all patients. Interestingly, we found a significant positive correlation between hsa-miR223-3p expression and cardiac parameters in typical FRDA patients (onset < 25 years). Moreover, a significant negative correlation between hsa-miR223-3p expression and HAX-1 (HCLS1 associated protein X-1) at mRNA and protein level was observed in all FRDA patients. In silico analyses suggested HAX-1 as a target gene of hsa-miR223-3p. Accordingly, we report that HAX-1 is negatively regulated by hsa-miR223-3p in cardiomyocytes (AC16) and neurons (SH-SY5Y), which are critically affected cell types in FRDA. This study describes for the first time the association between hsa-miR223-3p and HAX-1 expression in FRDA, thus supporting a potential role of this microRNA as non-invasive epigenetic biomarker for FRDA.
Collapse
Affiliation(s)
- Andrea Quatrana
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Elena Morini
- Section of Medical Genetics, Dept. of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Francesca Tiano
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy.,Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Chiara Vancheri
- Section of Medical Genetics, Dept. of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Luca Panarello
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Silvia Romano
- Neurosciences, Mental Health and Sensory Organs (NESMOS) Department, Faculty of Medicine and Psychology, Sapienza University, 00189 Rome, Italy
| | | | - Carlo Casali
- Dept. of Medical Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University of Rome, 04100 Latina, Italy
| | - Caterina Mariotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Alessia Mongelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Mario Fichera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Alessandra Rufini
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy.,Fratagene Therapeutics Srl, Rome, 00144 Rome, Italy.,Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Ivano Condò
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giuseppe Novelli
- Section of Medical Genetics, Dept. of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy.,Neuromed Institute, IRCCS, 86077 Pozzilli, Italy
| | - Roberto Testi
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy.,Fratagene Therapeutics Srl, Rome, 00144 Rome, Italy
| | - Francesca Amati
- Section of Medical Genetics, Dept. of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy.,Department for the Promotion of Human Science and Quality of Life, University San Raffaele, 00166 Rome, Italy
| | - Florence Malisan
- Laboratory of Signal Transduction, Dept. of Biomedicine and Prevention; University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
31
|
Lees JG, Napierala M, Pébay A, Dottori M, Lim SY. Cellular pathophysiology of Friedreich's ataxia cardiomyopathy. Int J Cardiol 2022; 346:71-78. [PMID: 34798207 DOI: 10.1016/j.ijcard.2021.11.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/01/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022]
Abstract
Friedreich's ataxia (FRDA) is a hereditary neuromuscular disorder. Cardiomyopathy is the leading cause of premature death in FRDA. FRDA cardiomyopathy is a complex and progressive disease with no cure or treatment to slow its progression. At the cellular level, cardiomyocyte hypertrophy, apoptosis and fibrosis contribute to the cardiac pathology. However, the heart is composed of multiple cell types and several clinical studies have reported the involvement of cardiac non-myocytes such as vascular cells, autonomic neurons, and inflammatory cells in the pathogenesis of FRDA cardiomyopathy. In fact, several of the cardiac pathologies associated with FRDA including cardiomyocyte necrosis, fibrosis, and arrhythmia, could be contributed to by a diseased vasculature and autonomic dysfunction. Here, we review available evidence regarding the current understanding of cellular mechanisms for, and the involvement of, cardiac non-myocytes in the pathogenesis of FRDA cardiomyopathy.
Collapse
Affiliation(s)
- Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria 3052, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, School of Medicine, Molecular Horizons, University of Wollongong, New South Wales 2522, Australia; Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
32
|
Seminotti B, Grings M, Tucci P, Leipnitz G, Saso L. Nuclear Factor Erythroid-2-Related Factor 2 Signaling in the Neuropathophysiology of Inherited Metabolic Disorders. Front Cell Neurosci 2021; 15:785057. [PMID: 34955754 PMCID: PMC8693715 DOI: 10.3389/fncel.2021.785057] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/05/2021] [Indexed: 01/14/2023] Open
Abstract
Inherited metabolic disorders (IMDs) are rare genetic conditions that affect multiple organs, predominantly the central nervous system. Since treatment for a large number of IMDs is limited, there is an urgent need to find novel therapeutical targets. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a transcription factor that has a key role in controlling the intracellular redox environment by regulating the expression of antioxidant enzymes and several important genes related to redox homeostasis. Considering that oxidative stress along with antioxidant system alterations is a mechanism involved in the neuropathophysiology of many IMDs, this review focuses on the current knowledge about Nrf2 signaling dysregulation observed in this group of disorders characterized by neurological dysfunction. We review here Nrf2 signaling alterations observed in X-linked adrenoleukodystrophy, glutaric acidemia type I, hyperhomocysteinemia, and Friedreich’s ataxia. Additionally, beneficial effects of different Nrf2 activators are shown, identifying a promising target for treatment of patients with these disorders. We expect that this article stimulates research into the investigation of Nrf2 pathway involvement in IMDs and the use of potential pharmacological modulators of this transcription factor to counteract oxidative stress and exert neuroprotection.
Collapse
Affiliation(s)
- Bianca Seminotti
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mateus Grings
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Guilhian Leipnitz
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Postgraduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Liu Y, Cai J, Shen J, Dong W, Xu L, Fang M, Lin Y, Liu J, Ding Y, Qiao T, Li K. SS-31 efficacy in a mouse model of Friedreich ataxia by upregulation of frataxin expression. Hum Mol Genet 2021; 31:176-188. [PMID: 34387346 DOI: 10.1093/hmg/ddab232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 06/05/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Friedreich ataxia (FRDA) is a serious hereditary neurodegenerative disease, mostly accompanied with hypertrophic cardiomyopathy, caused by the reduced expression of frataxin (FXN). However, there is still no effective treatment. Our previous studies have shown that SS-31, a mitochondrion-targeted peptide, is capable to upregulate the expression of FXN and improve the mitochondrial function in cells derived from FRDA patients. To further explore the potential of SS-31, we used the GAA expansion-based models, including Y47 and YG8R (Fxn KIKO) mice, primary neurons and macrophages from the mice and cells derived from FRDA patients. After once-daily intraperitoneal injection of 1 mg/kg SS-31 for 1 month, we observed the significant improvement of motor function. The vacuolation in dorsal root ganglia, lesions in dentate nuclei and the lost thickness of myelin sheath of spinal cord were all repaired after SS-31 treatment. In addition, the hypertrophic cardiomyocytes and disarrayed abnormal Purkinje cells were dramatically reduced. Interestingly, we found that SS-31 treatment upregulated FXN expression not only at the translational levels as observed in cell culture but also at mRNA levels in vivo. Consequently, mitochondrial morphology and function were greatly improved in all tested tissues. Importantly, our data provided additional evidence that the maintenance of the therapeutic benefits needed continuous drug administration. Taken together, our findings have demonstrated the effectiveness of SS-31 treatment through the upregulation of FXN in vivo and offer guidance of the potential usage in the clinical application for FRDA.
Collapse
Affiliation(s)
- Yutong Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Jing Cai
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jiaqi Shen
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Weichen Dong
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Li Xu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Maoxin Fang
- Department of Biological Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yishan Lin
- Department of Biological Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jiali Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Yibing Ding
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - Tong Qiao
- Department of Vascular Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| |
Collapse
|
34
|
The dynamin-related protein 1 is decreased and the mitochondrial network is altered in Friedreich's ataxia cardiomyopathy. Int J Biochem Cell Biol 2021; 143:106137. [PMID: 34923139 DOI: 10.1016/j.biocel.2021.106137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022]
Abstract
Friedreich ataxia is an autosomal recessive congenital neurodegenerative disease caused by a deficiency in the frataxin protein and is often diagnosed in young adulthood. An expansion of guanine-adenine-adenine repeats in the first intron of the FXN gene leads to decreased frataxin expression. Frataxin plays an essential role in mitochondrial metabolism. Most Friedreich ataxia patients are diagnosed with left ventricular hypertrophic cardiomyopathy, and 60% of patients die with hypertrophic cardiomyopathy. However, the mitochondrial anatomy in Friedreich ataxia hypertrophic cardiomyopathy is still poorly understood. We investigated mitochondrial fission, fusion, and function using biochemical, microscopy, and computational stochastic analysis in human induced pluripotent stem cell derived cardiomyocytes from a patient with Friedreich ataxia hypertrophic cardiomyopathy and a healthy individual. We found a significantly higher mitochondrial footprint, decreased mitochondrial fission protein dynamin-related protein, and mitochondrial fission rate over fusion with more giant mitochondrial clusters in human induced pluripotent stem cell derived cardiomyocytes from a patient with Friedreich ataxia hypertrophic cardiomyopathy, compared to an unaffected individual. We also found significantly depolarized mitochondrial membrane potential and higher reactive oxygen species levels in Friedreich ataxia human induced pluripotent stem cell cardiomyocytes. Our results show that frataxin's depletion may dampen the mitochondrial fission machinery by reducing dynamin-related protein1. The loss of mitochondrial fission might lead to elevated reactive oxygen species and depolarized mitochondrial membrane potential, which may cause oxidative damage in Friedreich ataxia hypertrophic cardiomyopathy. Further investigations are needed to identify the mechanism of downregulating dynamin-related protein1 due to the frataxin deficiency in Friedreich ataxia hypertrophic cardiomyopathy.
Collapse
|
35
|
Russo M, Nuzzo A, Foschi M, Boarin S, Lorenzetti S, Tomasi C, Querzani P, Rubboli A. Left atrial appendage thrombosis in a patient with Friedreich Ataxia-related cardiomyopathy, left ventricular systolic dysfunction, and atrial fibrillation. SAGE Open Med Case Rep 2021; 9:2050313X211056419. [PMID: 34733522 PMCID: PMC8558795 DOI: 10.1177/2050313x211056419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/12/2021] [Indexed: 12/04/2022] Open
Abstract
Friedreich ataxia is the most common form of hereditary ataxia. Heart involvement in Friedreich ataxia is common and can include increased left ventricular wall thickness, atrial fibrillation, and in the later stages, a reduction of left ventricular ejection fraction. We present the case of a 45-year-old man with a history of paroxysmal atrial fibrillation and a congestive heart failure, hypertension, age ⩾ 75 years, diabetes mellitus, stroke, vascular disease, age 65–74 years, and female sex (CHA2DS2-VASc) score of only 1 (because of reduced left ventricular ejection fraction) who presented with pneumonia and was also found to have atrial fibrillation with a rapid ventricular response. Despite already being on long-term therapy with a non-vitamin K-antagonist oral anticoagulant, a transesophageal echocardiogram showed a mobile floating thrombus in the left atrial appendage. In accordance with previous necropsy evidence of thrombosis and thromboembolism in Friedreich ataxia subjects who likely have had only non-sex-related CHA2DS2-VASc score ⩽1, this case suggests that the risk of thromboembolism in Friedreich ataxia subjects with atrial fibrillation may not be adequately predicted by the sole CHA2DS2-VASc score.
Collapse
Affiliation(s)
- Michele Russo
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Annachiara Nuzzo
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Matteo Foschi
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simona Boarin
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Stefano Lorenzetti
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Corrado Tomasi
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Pietro Querzani
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| | - Andrea Rubboli
- Department of Cardiovascular Diseases, Division of Cardiology, S. Maria delle Croci Hospital, AUSL Romagna, Ravenna, Italy
| |
Collapse
|
36
|
Amini O, Lakziyan R, Abavisani M, Sarchahi Z. The cardiomyopathy of Friedreich's ataxia common in a family: A case report. Ann Med Surg (Lond) 2021; 66:102408. [PMID: 34136207 PMCID: PMC8178102 DOI: 10.1016/j.amsu.2021.102408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/09/2021] [Accepted: 05/14/2021] [Indexed: 11/02/2022] Open
Abstract
Introduction and importance Friedreich's Ataxia is an autosomal recessive disease and is usually associated with arterial dysfunction, muscle weakness, spasm in the lower extremities, scoliosis, bladder dysfunction, lack of reflexes in the lower extremities, and imbalance. Approximately 2.3 people have cardiomyopathy. In this article, we have reviewed a case of Friedreich's Ataxia with hypertrophic cardiomyopathy. Case presentation A 19-year-old woman with Friedreich's Ataxia has been protesting since she was 11 years old and complained of chest pains, dyspnea, and heart palpitations without a medical history. In ECG, Asymmetrical invert T wave diffuse, diffuse ST-segment depression, and left ventricular hypertrophy were observed. In echocardiography, the left ventricle was reported as hyperimmobile with increased EF (70-75%). Clinical discussion In the present study, a patient with Friedrich Ataxia was diagnosed with chest pain, dyspnea, and palpitations without any medical history, and was discharged from the hospital after treatment. In the patients introduced and our patient, there was significant fibro-myocardial hypertrophy, in which the ventricular septal hypertrophy was marked by hypertrophic cardiomyopathy. Conclusion Because early diagnosis of the disease is difficult, clinical signs and the patient's current profile at the time of referral will be very helpful.
Collapse
Affiliation(s)
- Omidreza Amini
- Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Rasool Lakziyan
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mahnaz Abavisani
- Department of Nursing, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zohreh Sarchahi
- Department of Nursing, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
37
|
Culley MK, Zhao J, Tai YY, Tang Y, Perk D, Negi V, Yu Q, Woodcock CSC, Handen A, Speyer G, Kim S, Lai YC, Satoh T, Watson AM, Aaraj YA, Sembrat J, Rojas M, Goncharov D, Goncharova EA, Khan OF, Anderson DG, Dahlman JE, Gurkar AU, Lafyatis R, Fayyaz AU, Redfield MM, Gladwin MT, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency promotes endothelial senescence in pulmonary hypertension. J Clin Invest 2021; 131:136459. [PMID: 33905372 PMCID: PMC8159699 DOI: 10.1172/jci136459] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
The dynamic regulation of endothelial pathophenotypes in pulmonary hypertension (PH) remains undefined. Cellular senescence is linked to PH with intracardiac shunts; however, its regulation across PH subtypes is unknown. Since endothelial deficiency of iron-sulfur (Fe-S) clusters is pathogenic in PH, we hypothesized that a Fe-S biogenesis protein, frataxin (FXN), controls endothelial senescence. An endothelial subpopulation in rodent and patient lungs across PH subtypes exhibited reduced FXN and elevated senescence. In vitro, hypoxic and inflammatory FXN deficiency abrogated activity of endothelial Fe-S-containing polymerases, promoting replication stress, DNA damage response, and senescence. This was also observed in stem cell-derived endothelial cells from Friedreich's ataxia (FRDA), a genetic disease of FXN deficiency, ataxia, and cardiomyopathy, often with PH. In vivo, FXN deficiency-dependent senescence drove vessel inflammation, remodeling, and PH, whereas pharmacologic removal of senescent cells in Fxn-deficient rodents ameliorated PH. These data offer a model of endothelial biology in PH, where FXN deficiency generates a senescent endothelial subpopulation, promoting vascular inflammatory and proliferative signals in other cells to drive disease. These findings also establish an endothelial etiology for PH in FRDA and left heart disease and support therapeutic development of senolytic drugs, reversing effects of Fe-S deficiency across PH subtypes.
Collapse
Affiliation(s)
- Miranda K. Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Dror Perk
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Qiujun Yu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Chen-Shan C. Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Adam Handen
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Gil Speyer
- Research Computing, Arizona State University, Tempe, Arizona, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, College of Engineering, Prairie View A&M University, Prairie View, Texas, USA
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Taijyu Satoh
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Annie M.M. Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Dmitry Goncharov
- Lung Center, Pulmonary Vascular Disease Program, Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Elena A. Goncharova
- Lung Center, Pulmonary Vascular Disease Program, Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis School of Medicine, Davis, California, USA
| | - Omar F. Khan
- Institute of Biomedical Engineering, Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Daniel G. Anderson
- Department of Chemical Engineering, Institute of Medical Engineering and Science, Harvard-MIT Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James E. Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Aditi U. Gurkar
- Aging Institute, Division of Geriatric Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, GRECC VA, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ahmed U. Fayyaz
- Department of Cardiovascular Medicine and
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesotta, USA
| | | | - Mark T. Gladwin
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Thomas Bertero
- Université Côte d’Azur, CNRS, UMR7275, IPMC, Valbonne, France
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Divisions of Cardiology, Pulmonary, Allergy, and Critical Care Medicine and Rheumatology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
38
|
Cissé CAK, Cissé L, Ba HO, Samassékou O, Simaga A, Taméga A, Diarra S, Diallo SH, Coulibaly T, Diallo S, Yalcouyé A, Maiga AB, Keita M, Fischbeck KH, Traoré SF, Guinto CO, Landouré G. Friedreich ataxia in a family from Mali, West Africa/Friedreich ataxia in a Malian family. Clin Case Rep 2021; 9:e04065. [PMID: 34084490 PMCID: PMC8142306 DOI: 10.1002/ccr3.4065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/07/2021] [Indexed: 11/10/2022] Open
Abstract
Friedreich ataxia is the most common inherited ataxia in the world, but yet to be reported in black African. We report the first genetically confirmed case in a West African family. Studying genetic diseases in populations with diverse backgrounds may give new insights into their pathophysiology for future therapeutic targets.
Collapse
Affiliation(s)
| | - Lassana Cissé
- Service de NeurologieCentre Hospitalier Universitaire du Point “G”BamakoMali
| | - Hamidou O. Ba
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service de CardiologieCentre Hospitalier Universitaire de Gabriel TouréBamakoMali
| | | | - Assiatou Simaga
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Institut d’Ophtalmologie Tropicale d’AfriqueBamakoMali
| | | | - Salimata Diarra
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Neurogenetics BranchNINDSNIHBethesdaMDUSA
| | - Seybou H. Diallo
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire de Gabriel TouréBamakoMali
| | - Thomas Coulibaly
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire du Point “G”BamakoMali
| | - Salimata Diallo
- Service de NeurologieCentre Hospitalier Universitaire de Gabriel TouréBamakoMali
| | | | | | - Mohamed Keita
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service d’ORLCentre Hospitalier Universitaire de Gabriel TouréBamakoMali
| | | | - Sékou F. Traoré
- Malaria Research and Training CenterDepartment of EntomologyUSTTBBamakoMali
| | - Cheick O. Guinto
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire du Point “G”BamakoMali
| | - Guida Landouré
- Faculté de Médecine et d'OdontostomatologieUSTTBBamakoMali
- Service de NeurologieCentre Hospitalier Universitaire du Point “G”BamakoMali
- Neurogenetics BranchNINDSNIHBethesdaMDUSA
| | | |
Collapse
|
39
|
Mejia E, Lynch A, Hearle P, Okunowo O, Griffis H, Shah M, Lynch D, Lin KY. Ectopic Burden via Holter Monitors in Friedreich Ataxia. Pediatr Neurol 2021; 117:29-33. [PMID: 33652339 PMCID: PMC8085807 DOI: 10.1016/j.pediatrneurol.2021.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/09/2021] [Accepted: 01/17/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Friedreich ataxia is the most commonly inherited ataxia; nearly 60% of deaths are cardiac in nature, with one in eight deaths due to arrhythmia. Additional or irregular heartbeats, measured as ectopy, can be quantified using portable heart rhythm monitoring. We sought to describe the ectopic burden in Friedreich ataxia. METHODS Using a natural history study of patients with Friedreich ataxia at a single center, we analyzed portable heart rhythm monitors (Holters). Ectopic burden was defined as the proportion of atrial or ventricular ectopic beats over total beats. RESULTS Of 456 patients, 131 had Holters. Sixty-eight (52.0%) were male, median age of symptom onset was 8.0 years (5.0 to 13.0, n = 111), median age at time of Holter was 17.3 years (interquartile range [IQR] 12.9 to 22.8, n = 129), and median duration of illness was 8.7 years (IQR 5.3 to 11.6, n = 110). Median GAA length on the shorter FXN allele was 706.0 (IQR 550.0 to 840.0, n = 112). Eight (7.8%, n = 103) had diminished cardiac function, and 74 (74.0%, n = 100) had ventricular hypertrophy. Ninety patients (83.0%) had atrial ectopy (supraventricular ectopy [SVE]): 85 (78.0%) with rare SVE (>0% to 5%) and five (5.0%) with frequent SVE (>10%). Twenty-five (19.0%) had supraventricular runs, and one (0.8%) had atrial fibrillation/flutter. Forty-five (41.0%) had ventricular ectopy (VE): 43 (39.0%) with rare VE (0% to 5%) and two (2.0%) with moderate VE (5% to 10%). Compared with patients with none and rare SVE, patients with frequent SVE had longer disease duration (18.3 versus 4.6 versus 9.0 years, P = 0.0005). CONCLUSION Patients with longer disease duration had higher rates of SVE. Heart rhythm monitoring may be considered for risk stratification; however, longitudinal analysis is needed.
Collapse
Affiliation(s)
- Erika Mejia
- Division of Cardiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Abigail Lynch
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patrick Hearle
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Oluwatimilehin Okunowo
- Data Science & Biostatistics Unit, Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heather Griffis
- Data Science & Biostatistics Unit, Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maully Shah
- Division of Cardiology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David Lynch
- Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Y. Lin
- Division of Cardiology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Viventi S, Frausin S, Howden SE, Lim SY, Finol-Urdaneta RK, McArthur JR, Abu-Bonsrah KD, Ng W, Ivanusic J, Thompson L, Dottori M. In vivo survival and differentiation of Friedreich ataxia iPSC-derived sensory neurons transplanted in the adult dorsal root ganglia. Stem Cells Transl Med 2021; 10:1157-1169. [PMID: 33734599 PMCID: PMC8284774 DOI: 10.1002/sctm.20-0334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/05/2023] Open
Abstract
Friedreich ataxia (FRDA) is an autosomal recessive disease characterized by degeneration of dorsal root ganglia (DRG) sensory neurons, which is due to low levels of the mitochondrial protein Frataxin. To explore cell replacement therapies as a possible approach to treat FRDA, we examined transplantation of sensory neural progenitors derived from human embryonic stem cells (hESC) and FRDA induced pluripotent stem cells (iPSC) into adult rodent DRG regions. Our data showed survival and differentiation of hESC and FRDA iPSC-derived progenitors in the DRG 2 and 8 weeks post-transplantation, respectively. Donor cells expressed neuronal markers, including sensory and glial markers, demonstrating differentiation to these lineages. These results are novel and a highly significant first step in showing the possibility of using stem cells as a cell replacement therapy to treat DRG neurodegeneration in FRDA as well as other peripheral neuropathies.
Collapse
Affiliation(s)
- Serena Viventi
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Stefano Frausin
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Sara E Howden
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Surgery, The University of Melbourne, St Vincent Hospital, Fitzroy, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Keiraville, Australia
| | - Jeffrey R McArthur
- Illawarra Health and Medical Research Institute, University of Wollongong, Keiraville, Australia
| | - Kwaku Dad Abu-Bonsrah
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Wayne Ng
- School of Medicine, Griffith University, Gold Coast, Australia.,Department of Neurosurgery, Gold Coast University Hospital, Southport, Australia
| | - Jason Ivanusic
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Australia
| | - Lachlan Thompson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Mirella Dottori
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Keiraville, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Australia
| |
Collapse
|
41
|
Smith FM, Kosman DJ. Molecular Defects in Friedreich's Ataxia: Convergence of Oxidative Stress and Cytoskeletal Abnormalities. Front Mol Biosci 2020; 7:569293. [PMID: 33263002 PMCID: PMC7686857 DOI: 10.3389/fmolb.2020.569293] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2020] [Indexed: 01/18/2023] Open
Abstract
Friedreich’s ataxia (FRDA) is a multi-faceted disease characterized by progressive sensory–motor loss, neurodegeneration, brain iron accumulation, and eventual death by hypertrophic cardiomyopathy. FRDA follows loss of frataxin (FXN), a mitochondrial chaperone protein required for incorporation of iron into iron–sulfur cluster and heme precursors. After the discovery of the molecular basis of FRDA in 1996, over two decades of research have been dedicated to understanding the temporal manifestations of disease both at the whole body and molecular level. Early research indicated strong cellular iron dysregulation in both human and yeast models followed by onset of oxidative stress. Since then, the pathophysiology due to dysregulation of intracellular iron chaperoning has become central in FRDA relative to antioxidant defense and run-down in energy metabolism. At the same time, limited consideration has been given to changes in cytoskeletal organization, which was one of the first molecular defects noted. These alterations include both post-translational oxidative glutathionylation of actin monomers and differential DNA processing of a cytoskeletal regulator PIP5K1β. Currently unknown in respect to FRDA but well understood in the context of FXN-deficient cell physiology is the resulting impact on the cytoskeleton; this disassembly of actin filaments has a particularly profound effect on cell–cell junctions characteristic of barrier cells. With respect to a neurodegenerative disorder such as FRDA, this cytoskeletal and tight junction breakdown in the brain microvascular endothelial cells of the blood–brain barrier is likely a component of disease etiology. This review serves to outline a brief history of this research and hones in on pathway dysregulation downstream of iron-related pathology in FRDA related to actin dynamics. The review presented here was not written with the intent of being exhaustive, but to instead urge the reader to consider the essentiality of the cytoskeleton and appreciate the limited knowledge on FRDA-related cytoskeletal dysfunction as a result of oxidative stress. The review examines previous hypotheses of neurodegeneration with brain iron accumulation (NBIA) in FRDA with a specific biochemical focus.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| | - Daniel J Kosman
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
42
|
Mastroianno S, Germano M, Maggio A, Massaro R, Potenza DR, Russo A, Carella M, Di Stolfo G. Electrocardiogram in Friedreich's ataxia: A short-term surrogate endpoint for treatment efficacy. Ann Noninvasive Electrocardiol 2020; 26:e12813. [PMID: 33151022 PMCID: PMC8293611 DOI: 10.1111/anec.12813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 01/19/2023] Open
Abstract
Friedreich's ataxia is a rare degenerative neuromuscular disorder, caused by a homozygous GAA triplet repeat expansion in the frataxin (FXN) gene, with a broad clinical phenotype characterized by progressive gait and limb ataxia, dysarthria, and loss of lower limb reflexes; cardiac involvement is represented by hypertrophic cardiomyopathy, ventricular arrhythmias, and sudden cardiac deaths. Currently, no definite therapy is available, while many drugs are under investigation; for this reasons, we need markers of short‐ and long‐term treatment efficacy acting on different tissue for trial evaluation. We describe the case of a 21‐year‐old patient affected by Friedreich's ataxia on wheel‐chair, with initial cardiac involvement and electrocardiographic features characterized by thiamine treatment‐related negative T wave and QTc variations. We discuss plausible physiopathology and potential ECG role implications as an intermediate marker of treatment response in future clinical trials considering patients affected by Friedreich's ataxia.
Collapse
Affiliation(s)
- Sandra Mastroianno
- Cardiology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Michele Germano
- Child Neuropsychiatry Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Angela Maggio
- Paediatric Oncology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Raimondo Massaro
- Cardiology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | - Aldo Russo
- Cardiology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Medical Genetic Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giuseppe Di Stolfo
- Cardiology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
43
|
Manolis AS, Manolis AA, Manolis TA, Apostolaki NE, Apostolopoulos EJ, Melita H, Katsiki N. Mitochondrial dysfunction in cardiovascular disease: Current status of translational research/clinical and therapeutic implications. Med Res Rev 2020; 41:275-313. [PMID: 32959403 DOI: 10.1002/med.21732] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria provide energy to the cell during aerobic respiration by supplying ~95% of the adenosine triphosphate (ATP) molecules via oxidative phosphorylation. These organelles have various other functions, all carried out by numerous proteins, with the majority of them being encoded by nuclear DNA (nDNA). Mitochondria occupy ~1/3 of the volume of myocardial cells in adults, and function at levels of high-efficiency to promptly meet the energy requirements of the myocardial contractile units. Mitochondria have their own DNA (mtDNA), which contains 37 genes and is maternally inherited. Over the last several years, a variety of functions of these organelles have been discovered and this has led to a growing interest in their involvement in various diseases, including cardiovascular (CV) diseases. Mitochondrial dysfunction relates to the status where mitochondria cannot meet the demands of a cell for ATP and there is an enhanced formation of reactive-oxygen species. This dysfunction may occur as a result of mtDNA and/or nDNA mutations, but also as a response to aging and various disease and environmental stresses, leading to the development of cardiomyopathies and other CV diseases. Designing mitochondria-targeted therapeutic strategies aiming to maintain or restore mitochondrial function has been a great challenge as a result of variable responses according to the etiology of the disorder. There have been several preclinical data on such therapies, but clinical studies are scarce. A major challenge relates to the techniques needed to eclectically deliver the therapeutic agents to cardiac tissues and to damaged mitochondria for successful clinical outcomes. All these issues and progress made over the last several years are herein reviewed.
Collapse
Affiliation(s)
- Antonis S Manolis
- First Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | | | | | | | | | | | - Niki Katsiki
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, Diabetes Center, Medical School, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
44
|
Bizzoca A, Caracciolo M, Corsi P, Magrone T, Jirillo E, Gennarini G. Molecular and Cellular Substrates for the Friedreich Ataxia. Significance of Contactin Expression and of Antioxidant Administration. Molecules 2020; 25:E4085. [PMID: 32906751 PMCID: PMC7570916 DOI: 10.3390/molecules25184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 11/16/2022] Open
Abstract
In this study, the neural phenotype is explored in rodent models of the spinocerebellar disorder known as the Friedreich Ataxia (FA), which results from mutations within the gene encoding the Frataxin mitochondrial protein. For this, the M12 line, bearing a targeted mutation, which disrupts the Frataxin gene exon 4 was used, together with the M02 line, which, in addition, is hemizygous for the human Frataxin gene mutation (Pook transgene), implying the occurrence of 82-190 GAA repeats within its first intron. The mutant mice phenotype was compared to the one of wild type littermates in regions undergoing differential profiles of neurogenesis, including the cerebellar cortex and the spinal cord by using neuronal (β-tubulin) and glial (Glial Fibrillary Acidic Protein) markers as well as the Contactin 1 axonal glycoprotein, involved in neurite growth control. Morphological/morphometric analyses revealed that while in Frataxin mutant mice the neuronal phenotype was significantly counteracted, a glial upregulation occurred at the same time. Furthermore, Contactin 1 downregulation suggested that changes in the underlying gene contributed to the disorder pathogenesis. Therefore, the FA phenotype implies an alteration of the developmental profile of neuronal and glial precursors. Finally, epigallocatechin gallate polyphenol administration counteracted the disorder, indicating protective effects of antioxidant administration.
Collapse
Affiliation(s)
| | | | | | | | | | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. I-70124 Bari, Italy; (A.B.); (M.C.); (P.C.); (T.M.); (E.J.)
| |
Collapse
|
45
|
Chiang S, Huang MLH, Richardson DR. Treatment of dilated cardiomyopathy in a mouse model of Friedreich's ataxia using N-acetylcysteine and identification of alterations in microRNA expression that could be involved in its pathogenesis. Pharmacol Res 2020; 159:104994. [PMID: 32534099 DOI: 10.1016/j.phrs.2020.104994] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 01/01/2023]
Abstract
Deficient expression of the mitochondrial protein, frataxin, leads to a deadly cardiomyopathy. Our laboratory reported the master regulator of oxidative stress, nuclear factor erythroid 2-related factor-2 (Nrf2), demonstrates marked down-regulation after frataxin deletion in the heart. This was due, in part, to a pronounced increase in Keap1. To assess if this can be therapeutically targeted, cells were incubated with N-acetylcysteine (NAC), or buthionine sulfoximine (BSO), which increases or decreases glutathione (GSH), respectively, or the NRF2-inducer, sulforaphane (SFN). While SFN significantly (p < 0.05) induced NRF2, KEAP1 and BACH1, NAC attenuated SFN-induced NRF2, KEAP1 and BACH1. The down-regulation of KEAP1 by NAC was of interest, as Keap1 is markedly increased in the MCK conditional frataxin knockout (MCK KO) mouse model and this could lead to the decreased Nrf2 levels. Considering this, MCK KO mice were treated with i.p. NAC (500- or 1500-mg/kg, 5 days/week for 5-weeks) and demonstrated slightly less (p > 0.05) body weight loss versus the vehicle-treated KO. However, NAC did not rescue the cardiomyopathy. To additionally examine the dys-regulation of Nrf2 upon frataxin deletion, studies assessed the role of microRNA (miRNA) in this process. In MCK KO mice, miR-144 was up-regulated, which down-regulates Nrf2. Furthermore, miRNA screening in MCK KO mice demonstrated 23 miRNAs from 756 screened were significantly (p < 0.05) altered in KOs versus WT littermates. Of these, miR-21*, miR-34c*, and miR-200c, demonstrated marked alterations, with functional clustering analysis showing they regulate genes linked to cardiac hypertrophy, cardiomyopathy, and oxidative stress, respectively.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Animals
- Basic-Leucine Zipper Transcription Factors/metabolism
- Cardiomyopathy, Dilated/drug therapy
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cell Line, Tumor
- Disease Models, Animal
- Friedreich Ataxia/complications
- Friedreich Ataxia/genetics
- Gene Expression Regulation
- Humans
- Iron-Binding Proteins/genetics
- Iron-Binding Proteins/metabolism
- Isothiocyanates/pharmacology
- Kelch-Like ECH-Associated Protein 1/metabolism
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/metabolism
- Sulfoxides/pharmacology
- Frataxin
Collapse
Affiliation(s)
- S Chiang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - M L H Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia
| | - D R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), University of Sydney, Sydney, New South Wales, 2006 Australia; Centre for Cancer Cell Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia.
| |
Collapse
|
46
|
Salami CO, Jackson K, Jose C, Alyass L, Cisse GI, De BP, Stiles KM, Chiuchiolo MJ, Sondhi D, Crystal RG, Kaminsky SM. Stress-Induced Mouse Model of the Cardiac Manifestations of Friedreich's Ataxia Corrected by AAV-mediated Gene Therapy. Hum Gene Ther 2020; 31:819-827. [PMID: 32646255 DOI: 10.1089/hum.2019.363] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Friedreich's ataxia (FA), an autosomal recessive disorder caused by a deficiency in the expression of frataxin (FXN), is characterized by progressive ataxia and hypertrophic cardiomyopathy. Although cardiac dysfunction is the most common cause of mortality in FA, the cardiac disease remains subclinical for most of the clinical course because the neurologic disease limits muscle oxygen demands. Previous FXN knockout mouse models exhibit fatal cardiomyopathy similar to human FA, but in contrast to the human condition, untreated mice become moribund by 2 months of age, unlike humans where the cardiac disease often does not manifest until the third decade. The study was designed to create a mouse model for early FA disease relevant to the time for which a gene therapy would likely be most effective. To generate a cardiac-specific mouse model of FA cardiomyopathy similar to the human disease, we used a cardiac promoter (αMyhc) driving CRE recombinase cardiac-specific excision of FXN exon 4 to generate a mild, cardiac-specific FA model that is normal at rest, but exhibits the cardiac phenotype with stress. The hearts of αMyhc mice had decreased levels of FXN and activity of the mitochondrial complex II/complex IV respiratory chain. At rest, αMyhc mice exhibited normal cardiac function as assessed by echocardiographic assessment of ejection fraction and fractional shortening, but when the heart was stressed chemically with dobutamine, αMyhc mice compared with littermate control mice had a 62% reduction in the stress ejection fraction (p < 2 × 10-4) and 71% reduction in stress-related fractional shortening (p < 10-5). When assessing functional cardiac performance using running on an inclined treadmill, αMyhc mice stayed above the midline threefold less than littermate controls (p < 0.02). A one-time intravenous administration of 1011 genome copies of AAVrh.10hFXN, an adeno-associated virus (AAV) serotype rh10 gene transfer vector expressing human FXN, corrected the stress-induced ejection fraction and fractional shortening phenotypes. Treated αMyhc mice exhibited exercise performance on a treadmill indistinguishable from littermate controls (p > 0.07). These αMyhc mice provide an ideal model to study long-term cardiac complications due to FA and AAV-mediated gene therapy correction of stress-induced cardiac phenotypes typical of human FA.
Collapse
Affiliation(s)
- Christiana O Salami
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Katie Jackson
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Clarisse Jose
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laith Alyass
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Georges-Ibrahim Cisse
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Bishnu P De
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Maria J Chiuchiolo
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|