1
|
Piccolo S, Casal M, Rossi V, Ferrigni F, Piccoli A, Bolzan B, Setti M, Butturini C, Benfari G, Ferrero V, Franchi E, Tomasi L, Ribichini FL, Mugnai G. Ventricular arrhythmias and primary prevention of sudden cardiac death in Anderson-Fabry disease. Int J Cardiol 2024; 415:132444. [PMID: 39128566 DOI: 10.1016/j.ijcard.2024.132444] [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: 03/24/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
The Anderson-Fabry disease (AFD) is a X-linked lysosomal storage disorder due to the deficiency in the α-galactosidase A enzyme. Cardiovascular mortality is a major cause of death in patients with AFD and sudden cardiac death (SCD) is one of the main causes of death. The storage of glycosphingolipid along with ionic channel impairment, inflammation and fibrosis are involved in the arrhythmogenesis. Some risk factors have been associated with ventricular tachycardia (VT)/ventricular fibrillation (VF) and SCD. Left ventricular hypertrophy (LVH), cardiac fibrosis, non-sustained VTs seem to be the most important. Older age and male gender might be associated with higher risk of ventricular arrhythmias and SCD. Currently, the implantable cardioverter-defibrillator (ICD) is recommended in patients with AFD who have survived a cardiac arrest secondary to VT/VF or who experienced sustained VT causing syncope or hemodynamic compromise, and have a life expectancy >1 year. ICD implantation is also recommended in patients considered to be at high risk (e.g., patients with severe LVH or fibrosis). The present review sought to summarize the risk of ventricular arrythmias in AFD, the indications for ICD, focusing on pathophysiology and analyzing the role of possible predictors of arrhythmias in preventing SCD, especially as primary prevention.
Collapse
Affiliation(s)
- Solange Piccolo
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Matteo Casal
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Valentina Rossi
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Francesca Ferrigni
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Anna Piccoli
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Bruna Bolzan
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Martina Setti
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Caterina Butturini
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Giovanni Benfari
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Valeria Ferrero
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Elena Franchi
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Luca Tomasi
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Flavio Luciano Ribichini
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy
| | - Giacomo Mugnai
- Division of Cardiology, Cardio-Thoracic Department, University Hospital of Verona, Verona, Italy.
| |
Collapse
|
2
|
Lin CJ, Yang FJ, Wu CJ, Wu MJ, Wu MS. Insights of Fabry disease: Expert consensus approach for screening, diagnosis, and multidisciplinary management in chronic kidney disease. J Formos Med Assoc 2024:S0929-6646(24)00453-4. [PMID: 39358116 DOI: 10.1016/j.jfma.2024.09.033] [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: 03/23/2024] [Revised: 08/15/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
The prevalence of Fabry disease (FD) among males with chronic kidney disease (CKD) of unknown etiology in Taiwan is 0.6%. Despite this, FD is frequently overlooked in clinical settings. To address this issue, two consensus meetings were conducted in Taiwan-one in August 2022 and another in April 2023. The first meeting established screening criteria based on age, gender, family history, cardiac involvement, and symptoms. The second meeting, with a multidisciplinary team, developed treatment recommendations. The consensus emphasizes the importance of proactive data collection in dialysis units and outpatient follow-ups to enhance FD detection and management. The screening algorithm recommends incorporating FD screening into the diagnostic process for CKD patients, regardless of age. Priority is given to patients with a family history of FD, early stroke history, or classical FD symptoms. Comprehensive screening is also advised for CKD patients without obvious classical symptoms. Screening protocols for males include measuring α-galactosidase A enzyme activity, with reduced activity leading to further tests such as lyso-Gb3 level quantification and genetic analysis. For females, the protocol involves evaluating lyso-Gb3 plasma levels and genetic testing. FD, though often underestimated, is more prevalent than recognized and necessitates a multidisciplinary approach for timely diagnosis. Enhancing awareness and adopting a comprehensive approach are essential for improving patient outcomes.
Collapse
Affiliation(s)
- Cheng-Jui Lin
- Division of Nephrology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Feng-Jung Yang
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Yunlin Branch, Douliu, Taiwan
| | - Chih-Jen Wu
- Division of Nephrology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
| | - Mai-Szu Wu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
3
|
Sokolska JM, Károlyi M, Hiestand DR, Gastl M, Weber L, Sokolski M, Kosmala W, Alkadhi H, Gruner C, Manka R. Myocardial Fibrosis Quantification Methods by Cardiovascular Magnetic Resonance Imaging in Patients with Fabry Disease. J Clin Med 2024; 13:5047. [PMID: 39274260 PMCID: PMC11395808 DOI: 10.3390/jcm13175047] [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: 07/14/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: The presence of late gadolinium enhancement (LGE) on cardiac magnetic resonance (CMR) in patients with Fabry disease (FD) is a predictor of adverse cardiac events. The aim of this study was to establish the most reliable and reproducible technique for quantifying LGE in patients with FD. Methods: Twenty FD patients with LGE who underwent CMR on the same scanner and LGE sequence were included. LGE quantifications were done using gray-scale thresholds of 2, 3, 4, 5 and 6 standard deviations (SD) above the mean signal intensity of the remote myocardium, the full width at half maximum method (FWHM), visual assessment with threshold (VAT) and the fully manual method (MM). Results: The mean amount of fibrosis varied between quantification techniques from 36 ± 19 at 2SD to 2 ± 2 g using the FWHM (p < 0.0001). Intraobserver reliability was excellent for most methods, except for the FWHM which was good (ICC 0.84; all p < 0.05). Interobserver reliability was excellent for VAT (ICC 0.94) and good for other techniques (all p < 0.05). Intraobserver reproducibility showed the lowest coefficient of variation (CV, 6%) at 5SD and at 2SD and VAT (35% and 38%) for interobserver reproducibility. The FWHM revealed the highest CV (63% and 94%) for both intra- and interobserver reproducibility. Conclusions: The available methods for LGE quantification demonstrate good to excellent intra- and interobserver reproducibility in patients with FD. The most reliable and reproducible techniques were VAT and 5SD, whereas the FWHM was the least reliable in the setting of our study. The total amount of LGE varies strongly with the quantification technique used.
Collapse
Affiliation(s)
- Justyna M Sokolska
- Institute of Heart Diseases, Faculty of Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mihály Károlyi
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Dana R Hiestand
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mareike Gastl
- Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Lucas Weber
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Mateusz Sokolski
- Institute of Heart Diseases, Faculty of Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Kosmala
- Institute of Heart Diseases, Faculty of Medicine, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Hatem Alkadhi
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Christiane Gruner
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Robert Manka
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
4
|
Weissman D, Dudek J, Sequeira V, Maack C. Fabry Disease: Cardiac Implications and Molecular Mechanisms. Curr Heart Fail Rep 2024; 21:81-100. [PMID: 38289538 PMCID: PMC10923975 DOI: 10.1007/s11897-024-00645-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW This review explores the interplay among metabolic dysfunction, oxidative stress, inflammation, and fibrosis in Fabry disease, focusing on their potential implications for cardiac involvement. We aim to discuss the biochemical processes that operate in parallel to sphingolipid accumulation and contribute to disease pathogenesis, emphasizing the importance of a comprehensive understanding of these processes. RECENT FINDINGS Beyond sphingolipid accumulation, emerging studies have revealed that mitochondrial dysfunction, oxidative stress, and chronic inflammation could be significant contributors to Fabry disease and cardiac involvement. These factors promote cardiac remodeling and fibrosis and may predispose Fabry patients to conduction disturbances, ventricular arrhythmias, and heart failure. While current treatments, such as enzyme replacement therapy and pharmacological chaperones, address disease progression and symptoms, their effectiveness is limited. Our review uncovers the potential relationships among metabolic disturbances, oxidative stress, inflammation, and fibrosis in Fabry disease-related cardiac complications. Current findings suggest that beyond sphingolipid accumulation, other mechanisms may significantly contribute to disease pathogenesis. This prompts the exploration of innovative therapeutic strategies and underscores the importance of a holistic approach to understanding and managing Fabry disease.
Collapse
Affiliation(s)
- David Weissman
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Vasco Sequeira
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
5
|
Bosquetti B, Santana AA, Gregório PC, da Cunha RS, Miniskiskosky G, Budag J, Franco CRC, Ramos EADS, Barreto FC, Stinghen AEM. The Role of α3β1 Integrin Modulation on Fabry Disease Podocyte Injury and Kidney Impairment. Toxins (Basel) 2023; 15:700. [PMID: 38133204 PMCID: PMC10748128 DOI: 10.3390/toxins15120700] [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: 10/06/2023] [Revised: 11/25/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Podocyte dysfunction plays a crucial role in renal injury and is identified as a key contributor to proteinuria in Fabry disease (FD), primarily impacting glomerular filtration function (GFF). The α3β1 integrins are important for podocyte adhesion to the glomerular basement membrane, and disturbances in these integrins can lead to podocyte injury. Therefore, this study aimed to assess the effects of chloroquine (CQ) on podocytes, as this drug can be used to obtain an in vitro condition analogous to the FD. Murine podocytes were employed in our experiments. The results revealed a dose-dependent reduction in cell viability. CQ at a sub-lethal concentration (1.0 µg/mL) induced lysosomal accumulation significantly (p < 0.0001). Morphological changes were evident through scanning electron microscopy and immunofluorescence, highlighting alterations in F-actin and nucleus morphology. No significant changes were observed in the gene expression of α3β1 integrins via RT-qPCR. Protein expression of α3 integrin was evaluated with Western Blotting and immunofluorescence, demonstrating its lower detection in podocytes exposed to CQ. Our findings propose a novel in vitro model for exploring secondary Fabry nephropathy, indicating a modulation of α3β1 integrin and morphological alterations in podocytes under the influence of CQ.
Collapse
Affiliation(s)
- Bruna Bosquetti
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Aline Aparecida Santana
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Paulo Cézar Gregório
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Regiane Stafim da Cunha
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Guilherme Miniskiskosky
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Julia Budag
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Célia Regina Cavichiolo Franco
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Edneia Amancio de Souza Ramos
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| | - Fellype Carvalho Barreto
- Internal Medicine Department, Division of Nephrology, Universidade Federal do Paraná, Curitiba 80060-900, Brazil;
| | - Andréa Emilia Marques Stinghen
- Experimental Nephrology Laboratory, Basic Pathology Department, Universidade Federal do Paraná, Curitiba 81531-980, Brazil; (B.B.); (A.A.S.); (P.C.G.); (R.S.d.C.); (G.M.); (J.B.); (C.R.C.F.); (E.A.d.S.R.)
| |
Collapse
|
6
|
Powell AW, Wittekind SG, Mays WA, Lang SM, Knilans TK, Prada CE, Hopkin RJ, Chin C. Arrhythmia Burden and Heart Rate Response During Exercise in Anderson-Fabry Disease. Tex Heart Inst J 2022; 49:485731. [PMID: 36069908 DOI: 10.14503/thij-20-7363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Patients with Anderson-Fabry disease (AFD) have an elevated incidence of resting arrhythmias and ischemic heart disease, but their exercise arrhythmia burden and ischemic changes are not well understood. In addition, little research has been done on heart rate recovery in these patients. We retrospectively reviewed charts of patients with AFD who underwent maximal effort cardiopulmonary exercise testing (CPET) (n=44; 38.2 ± 13.8 yr; 23 men) from 2012 through 2018. Electrocardiographic, Holter monitoring, echocardiographic, cardiac magnetic resonance imaging, and patient demographic data were collected. No patient had adverse events that necessitated CPET termination, whereas 25 (57%) had ectopy during CPET, including 3 (7%) with frequent premature atrial contractions and 5 (11%) with frequent premature ventricular contractions. The ectopic burden was higher during resting electrocardiographic monitoring before exercise. In addition, 7 patients (16%) had pathologic ST-segment or T-wave changes on CPET, defined as ST-segment changes ≥2 mm. Among the patients who had concurrent cardiac magnetic resonance findings with their CPET (n=27), ST-segment or T-wave changes were associated with left ventricular myocardial mass (r=0.43, P=0.02). Chronotropic incompetence was seen during CPET in 28 patients (64%); however, only 2 patients (4%) had abnormal heart rate recovery at 1 minute. This study shows that patients with AFD can safely undergo exercise testing but have a high incidence of exercise-induced arrhythmias and ischemic changes. Ischemic electrocardiographic changes during exercise testing are associated with myocardial mass. Despite the chronotropic incompetence associated with AFD, heart rate recovery appears to be generally preserved in these patients.
Collapse
Affiliation(s)
- Adam W Powell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Samuel G Wittekind
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Wayne A Mays
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sean M Lang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Timothy K Knilans
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Carlos E Prada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Robert J Hopkin
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Clifford Chin
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
7
|
Hongo K. Cardiac involvement in Fabry disease - A non-invasive assessment and the role of specific therapies. Mol Genet Metab 2022; 137:179-186. [PMID: 36088815 DOI: 10.1016/j.ymgme.2022.08.006] [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: 06/01/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022]
Abstract
Fabry disease is an X-linked inherited metabolic disorder due to the pathogenic mutation of the GLA gene, which codes lysosomal enzyme alpha-galactosidase A. The resultant accumulation of glycosphingolipids causes various systemic symptoms in childhood and adolescence, and major organ damage in adulthood. Cardiac involvement is important as the most frequent cause of death in Fabry disease patients. Progressive left ventricular hypertrophy with varying degrees of contractile dysfunction as well as conduction abnormalities and arrhythmias are typical cardiac features, and these findings can be evaluated in detail via non-invasive modalities, such as an electrocardiogram, echocardiography and cardiac magnetic resonance. In addition, specific therapies of enzyme replacement therapy and pharmacological chaperone therapy are available, and their beneficial effects on cardiac involvement have been reported. This minireview highlights recent evidence concerning non-invasive modalities for assessing cardiac involvement in Fabry disease and the effects of enzyme replacement therapy and pharmacological chaperone therapy on the findings of those modalities.
Collapse
Affiliation(s)
- Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, 105-8461 Tokyo, Japan.
| |
Collapse
|
8
|
Anan I, Sakuma T, Fukuro E, Morimoto S, Nojiri A, Kawai M, Sakurai K, Kobayashi M, Kobayashi H, Ida H, Ohashi T, Yoshimura M, Eto Y, Hongo K. The role of native T1 values on the evaluation of cardiac manifestation in Japanese Fabry disease patients. Mol Genet Metab Rep 2022; 31:100858. [PMID: 35782602 PMCID: PMC9248233 DOI: 10.1016/j.ymgmr.2022.100858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
Aims T1 mapping in cardiac magnetic resonance imaging enables us to distinguish various myocardial diseases showing left ventricular hypertrophy. Fabry disease is a lysosomal storage disorder causing the accumulation of glycosphingolipids into various organs, including the heart, which can be detected by native T1 values in T1 mapping. However, there is no report for the systematic evaluation of native T1 values in Fabry disease in Japan. Methods and results We analyzed native T1 values of 30 Fabry disease patients (14 males and 16 females) obtained by 3-T cardiac magnetic resonance imaging. Averaged T1 values were significantly lower in male patients (septal T1: 1149.5 ± 63.3 ms; total T1: 1145.1 ± 59.5 ms) than in female patients (septal T1: 1210.5 ± 45.5 ms; total T1: 1198.8 ± 51.8 ms) (p < 0.01). We compared the native T1 values of Fabry disease patients with those obtained from 15 hypertrophic cardiomyopathy patients (9 males and 6 females). Native T1 values effectively differentiate Fabry disease from hypertrophic cardiomyopathy (septal T1: sensitivity 93.3% and specificity 80.0%; total T1: sensitivity 86.7% and specificity 73.3%). In addition, native T1 values had a significant negative correlation with the left ventricular mass index in male patients at the pre-hypertrophic stage (p < 0.05). In male and female patients without late-gadolinium enhancement, native T1 values also had a significant negative correlation with the left ventricular mass index (p < 0.05). Conclusion These results suggest that native T1 values can be used to discriminate Fabry disease from hypertrophic cardiomyopathy and can reflect the accumulation of glycosphingolipids in cardiomyocytes.
Collapse
Affiliation(s)
- Ikuko Anan
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toru Sakuma
- Department of Radiology, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Eiko Fukuro
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ayumi Nojiri
- Department of Laboratory Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ken Sakurai
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masahisa Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroshi Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
- Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toya Ohashi
- The Jikei University School of Nursing, 8-3-1, Kokuryo-cho, Chofu-shi, Tokyo 182-8570, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorders, 255, Furusawa-Miyako, Kawasaki Asao-ku, Kanagawa 215-0026, Japan
| | - Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
- Corresponding author.
| |
Collapse
|
9
|
Tolstik E, Ali N, Guo S, Ebersbach P, Möllmann D, Arias-Loza P, Dierks J, Schuler I, Freier E, Debus J, Baba HA, Nordbeck P, Bocklitz T, Lorenz K. CARS Imaging Advances Early Diagnosis of Cardiac Manifestation of Fabry Disease. Int J Mol Sci 2022; 23:5345. [PMID: 35628155 PMCID: PMC9142043 DOI: 10.3390/ijms23105345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 12/12/2022] Open
Abstract
Vibrational spectroscopy can detect characteristic biomolecular signatures and thus has the potential to support diagnostics. Fabry disease (FD) is a lipid disorder disease that leads to accumulations of globotriaosylceramide in different organs, including the heart, which is particularly critical for the patient's prognosis. Effective treatment options are available if initiated at early disease stages, but many patients are late- or under-diagnosed. Since Coherent anti-Stokes Raman (CARS) imaging has a high sensitivity for lipid/protein shifts, we applied CARS as a diagnostic tool to assess cardiac FD manifestation in an FD mouse model. CARS measurements combined with multivariate data analysis, including image preprocessing followed by image clustering and data-driven modeling, allowed for differentiation between FD and control groups. Indeed, CARS identified shifts of lipid/protein content between the two groups in cardiac tissue visually and by subsequent automated bioinformatic discrimination with a mean sensitivity of 90-96%. Of note, this genotype differentiation was successful at a very early time point during disease development when only kidneys are visibly affected by globotriaosylceramide depositions. Altogether, the sensitivity of CARS combined with multivariate analysis allows reliable diagnostic support of early FD organ manifestation and may thus improve diagnosis, prognosis, and possibly therapeutic monitoring of FD.
Collapse
Affiliation(s)
- Elen Tolstik
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
| | - Nairveen Ali
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany; (N.A.); (S.G.); (T.B.)
| | - Shuxia Guo
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany; (N.A.); (S.G.); (T.B.)
| | - Paul Ebersbach
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
| | - Dorothe Möllmann
- Institute of Pathology, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; (D.M.); (H.A.B.)
| | - Paula Arias-Loza
- Department of Nuclear Medicine, Oberdürrbacher Str. 6, 97080 Wuerzburg, Germany;
| | - Johann Dierks
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
| | - Irina Schuler
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
| | - Jörg Debus
- Department of Physics, TU Dortmund University, Otto-Hahn-Str. 4a, 44227 Dortmund, Germany;
| | - Hideo A. Baba
- Institute of Pathology, University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany; (D.M.); (H.A.B.)
| | - Peter Nordbeck
- Department of Internal Medicine I, University of Würzburg, Oberdürrbacher Str. 6, 97080 Wuerzburg, Germany;
| | - Thomas Bocklitz
- Leibniz Institute of Photonic Technology, Member of Leibniz Health Technologies, Albert-Einstein-Str. 9, 07745 Jena, Germany; (N.A.); (S.G.); (T.B.)
| | - Kristina Lorenz
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Bunsen-Kirchhoff-Str. 11, 44139 Dortmund, Germany; (P.E.); (J.D.); (I.S.); (E.F.)
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Str. 9, 97078 Wuerzburg, Germany
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Am Schwarzenberg 15, 97078 Wuerzburg, Germany
| |
Collapse
|
10
|
Ezgu F, Alpsoy E, Bicik Bahcebasi Z, Kasapcopur O, Palamar M, Onay H, Ozdemir BH, Topcuoglu MA, Tufekcioglu O. Expert opinion on the recognition, diagnosis and management of children and adults with Fabry disease: a multidisciplinary Turkey perspective. Orphanet J Rare Dis 2022; 17:90. [PMID: 35236382 PMCID: PMC8889663 DOI: 10.1186/s13023-022-02215-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/06/2022] [Indexed: 11/10/2022] Open
Abstract
This consensus statement by a panel of Fabry experts aimed to identify areas of consensus on conceptual, clinical and therapeutic aspects of Fabry disease (FD) and to provide guidance to healthcare providers on best practice in the management of pediatric and adult patients with FD. This consensus statement indicated the clinical heterogeneity of FD as well as a large number of pathogenic variants in the GLA gene, emphasizing a need for an individualized approach to patient care. The experts reached consensus on the critical role of a high index of suspicion in symptomatic patients and screening of certain at-risk groups to reveal timely and accurate diagnosis of FD along with an increased awareness of the treating physician about the different kinds of pathogenic variants and their clinical implications. The experts emphasized the crucial role of timely recognition of FD with minimal delay from symptom onset to definite diagnosis in better management of FD patients, given the likelihood of changing the disease's natural history, improving the patients' quality of life and the prognosis after enzyme replacement therapy (ERT) administered through a coordinated, multidisciplinary care approach. In this regard, this consensus document is expected to increase awareness among physicians about unique characteristics of FD to assist clinicians in recognizing FD with a well-established clinical suspicion consistent with pathogenic variants and gender-based heterogeneous clinical manifestations of FD and in translating this information into their clinical practice for best practice in the management of patients with FD.
Collapse
Affiliation(s)
- Fatih Ezgu
- Department of Pediatrics, Division of Pediatric Metabolism and Division of Pediatric Genetics, Gazi University Faculty of Medicine, 06560 Ankara, Turkey
| | - Erkan Alpsoy
- Department of Dermatology, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Zerrin Bicik Bahcebasi
- Clinic of Nephrology, Kartal Dr. Lutfu Kirdar Training and Research Hospital, Istanbul, Turkey
| | - Ozgur Kasapcopur
- Department of Pediatrics, Division of Pediatric Rheumatology, Istanbul University Cerrahpasa Faculty of Medicine, Istanbul, Turkey
| | - Melis Palamar
- Department of Ophthalmology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Huseyin Onay
- Department of Medical Genetics, Ege University Faculty of Medicine, Izmir, Turkey
| | | | | | - Omac Tufekcioglu
- University of Health Sciences Department of Cardiology, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
11
|
Verheule S, Schotten U. Electrophysiological Consequences of Cardiac Fibrosis. Cells 2021; 10:cells10113220. [PMID: 34831442 PMCID: PMC8625398 DOI: 10.3390/cells10113220] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/27/2022] Open
Abstract
For both the atria and ventricles, fibrosis is generally recognized as one of the key determinants of conduction disturbances. By definition, fibrosis refers to an increased amount of fibrous tissue. However, fibrosis is not a singular entity. Various forms can be distinguished, that differ in distribution: replacement fibrosis, endomysial and perimysial fibrosis, and perivascular, endocardial, and epicardial fibrosis. These different forms typically result from diverging pathophysiological mechanisms and can have different consequences for conduction. The impact of fibrosis on propagation depends on exactly how the patterns of electrical connections between myocytes are altered. We will therefore first consider the normal patterns of electrical connections and their regional diversity as determinants of propagation. Subsequently, we will summarize current knowledge on how different forms of fibrosis lead to a loss of electrical connectivity in order to explain their effects on propagation and mechanisms of arrhythmogenesis, including ectopy, reentry, and alternans. Finally, we will discuss a histological quantification of fibrosis. Because of the different forms of fibrosis and their diverging effects on electrical propagation, the total amount of fibrosis is a poor indicator for the effect on conduction. Ideally, an assessment of cardiac fibrosis should exclude fibrous tissue that does not affect conduction and differentiate between the various types that do; in this article, we highlight practical solutions for histological analysis that meet these requirements.
Collapse
|
12
|
Lenders M, Brand E. Mechanisms of Neutralizing Anti-drug Antibody Formation and Clinical Relevance on Therapeutic Efficacy of Enzyme Replacement Therapies in Fabry Disease. Drugs 2021; 81:1969-1981. [PMID: 34748189 PMCID: PMC8602155 DOI: 10.1007/s40265-021-01621-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 12/13/2022]
Abstract
Fabry disease (FD) is a rare X-linked lysosomal storage disorder caused by mutations in the α-galactosidase A (AGAL/GLA) gene. The lysosomal accumulation of the substrates globotriaosylceramide (Gb3) and globotriaosylsphingosine (lyso-Gb3) results in progressive renal failure, cardiomyopathy associated with cardiac arrhythmia, and recurrent strokes, significantly limiting life expectancy in affected patients. Current treatment options for FD include recombinant enzyme-replacement therapies (ERTs) with intravenous agalsidase-α (0.2 mg/kg body weight) or agalsidase-β (1 mg/kg body weight) every 2 weeks, facilitating cellular Gb3 clearance and an overall improvement of disease burden. However, ERT can lead to infusion-associated reactions, as well as the formation of neutralizing anti-drug antibodies (ADAs) in ERT-treated males, leading to an attenuation of therapy efficacy and thus disease progression. In this narrative review, we provide a brief overview of the clinical picture of FD and diagnostic confirmation. The focus is on the biochemical and clinical significance of neutralizing ADAs as a humoral response to ERT. In addition, we provide an overview of different methods for ADA measurement and characterization, as well as potential therapeutic approaches to prevent or eliminate ADAs in affected patients, which is representative for other ERT-treated lysosomal storage diseases.
Collapse
Affiliation(s)
- Malte Lenders
- Department of Internal Medicine D, Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany.
| | - Eva Brand
- Department of Internal Medicine D, Nephrology, Hypertension and Rheumatology, Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Muenster, Albert-Schweitzer-Campus 1, 48149, Muenster, Germany
| |
Collapse
|
13
|
Williams S, El-Medany A, Nightingale A, Ismail Y. Rare presentation of Fabry disease as 'burnt-out' hypertrophic cardiomyopathy. BMJ Case Rep 2021; 14:e243604. [PMID: 34479887 PMCID: PMC8420664 DOI: 10.1136/bcr-2021-243604] [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] [Accepted: 07/28/2021] [Indexed: 11/04/2022] Open
Abstract
We herein report the case of a 53-year-old man who was historically diagnosed with hypertrophic cardiomyopathy (HCM) and was lost to follow-up, before presenting with end-stage heart failure. This was initially suspected as dilated cardiomyopathy and then 'burnt-out phase' of HCM but subsequently the underlying diagnosis was Fabry disease. Fabry disease is an uncommon lysosomal-storage disease due to reduced or absent activity of the alpha-galactosidase A enzyme. Cardiac involvement most frequently comprises left ventricular hypertrophy. Early treatment of the underlying condition with enzyme replacement therapy may prevent the progression to end-stage heart failure. Fabry disease should be considered in all patients presenting with a clinical phenotype of HCM and a historical diagnosis should be re-evaluated in light of new diagnostic tools. Untreated Fabry can progress to a 'burnt out' phase, whereby initial hypertrophy undergoes eccentric remodelling to a dilated, severely impaired left ventricle.
Collapse
Affiliation(s)
- Sam Williams
- Division of Cardiology, Bristol Heart Institute, Bristol, UK
| | - Ahmed El-Medany
- Division of Cardiology, Bristol Heart Institute, Bristol, UK
| | | | - Yasmin Ismail
- Division of Cardiology, Bristol Heart Institute, Bristol, UK
| |
Collapse
|
14
|
Viggiano E, Politano L. X Chromosome Inactivation in Carriers of Fabry Disease: Review and Meta-Analysis. Int J Mol Sci 2021; 22:ijms22147663. [PMID: 34299283 PMCID: PMC8304911 DOI: 10.3390/ijms22147663] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022] Open
Abstract
Anderson-Fabry disease is an X-linked inborn error of glycosphingolipid catabolism caused by a deficiency of α-galactosidase A. The incidence ranges between 1: 40,000 and 1:117,000 of live male births. In Italy, an estimate of incidence is available only for the north-western Italy, where it is of approximately 1:4000. Clinical symptoms include angiokeratomas, corneal dystrophy, and neurological, cardiac and kidney involvement. The prevalence of symptomatic female carriers is about 70%, and in some cases, they can exhibit a severe phenotype. Previous studies suggest a correlation between skewed X chromosome inactivation and symptoms in carriers of X-linked disease, including Fabry disease. In this review, we briefly summarize the disease, focusing on the clinical symptoms of carriers and analysis of the studies so far published in regards to X chromosome inactivation pattern, and manifesting Fabry carriers. Out of 151 records identified, only five reported the correlation between the analysis of XCI in leukocytes and the related phenotype in Fabry carriers, in particular evaluating the Mainz Severity Score Index or cardiac involvement. The meta-analysis did not show any correlation between MSSI or cardiac involvement and skewed XCI, likely because the analysis of XCI in leukocytes is not useful for predicting the phenotype in Fabry carriers.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Prevention, UOC Hygiene Service and Public Health, ASL Roma 2, 00142 Rome, Italy
- Correspondence: (E.V.); (L.P.)
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, Luigi Vanvitelli University, 80138 Naples, Italy
- Correspondence: (E.V.); (L.P.)
| |
Collapse
|
15
|
Schumann A, Schaller K, Belche V, Cybulla M, Grünert SC, Moers N, Sass JO, Kaech A, Hannibal L, Spiekerkoetter U. Defective lysosomal storage in Fabry disease modifies mitochondrial structure, metabolism and turnover in renal epithelial cells. J Inherit Metab Dis 2021; 44:1039-1050. [PMID: 33661535 DOI: 10.1002/jimd.12373] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 12/16/2022]
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder. Deficiency of the lysosomal enzyme alpha-galactosidase (GLA) leads to accumulation of potentially toxic globotriaosylceramide (Gb3) on a multisystem level. Cardiac and cerebrovascular abnormalities as well as progressive renal failure are severe, life-threatening long-term complications. The complete pathophysiology of chronic kidney disease (CKD) in FD and the role of tubular involvement for its progression are unclear. We established human renal tubular epithelial cell lines from the urine of male FD patients and male controls. The renal tubular system is rich in mitochondria and involved in transport processes at high-energy costs. Our studies revealed fragmented mitochondria with disrupted cristae structure in FD patient cells. Oxidative stress levels were elevated and oxidative phosphorylation was upregulated in FD pointing at enhanced energetic needs. Mitochondrial homeostasis and energy metabolism revealed major changes as evidenced by differences in mitochondrial number, energy production and fuel consumption. The changes were accompanied by activation of the autophagy machinery in FD. Sirtuin1, an important sensor of (renal) metabolic stress and modifier of different defense pathways, was highly expressed in FD. Our data show that lysosomal FD impairs mitochondrial function and results in severe disturbance of mitochondrial energy metabolism in renal cells. This insight on a tissue-specific level points to new therapeutic targets which might enhance treatment efficacy.
Collapse
Affiliation(s)
- Anke Schumann
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Kristin Schaller
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Véronique Belche
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Markus Cybulla
- Center of Internal Medicine, Department of Nephrology and Rheumatology, Fachinternistische Gemeinschaftspraxis Markgraeflerland, Muellheim, Germany
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Nicolai Moers
- Department of Natural Sciences, Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Jörn O Sass
- Department of Natural Sciences, Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Luciana Hannibal
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Laboratory of Clinical Biochemistry and Metabolism, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Center-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
16
|
Yim J, Yau O, Yeung DF, Tsang TSM. Fabry Cardiomyopathy: Current Practice and Future Directions. Cells 2021; 10:cells10061532. [PMID: 34204530 PMCID: PMC8233708 DOI: 10.3390/cells10061532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder caused by mutations in the galactosidase A (GLA) gene that result in deficient galactosidase A enzyme and subsequent accumulation of glycosphingolipids throughout the body. The result is a multi-system disorder characterized by cutaneous, corneal, cardiac, renal, and neurological manifestations. Increased left ventricular wall thickness represents the predominant cardiac manifestation of FD. As the disease progresses, patients may develop arrhythmias, advanced conduction abnormalities, and heart failure. Cardiac biomarkers, point-of-care dried blood spot testing, and advanced imaging modalities including echocardiography with strain imaging and magnetic resonance imaging (MRI) with T1 mapping now allow us to detect Fabry cardiomyopathy much more effectively than in the past. While enzyme replacement therapy (ERT) has been the mainstay of treatment, several promising therapies are now in development, making early diagnosis of FD even more crucial. Ongoing initiatives involving artificial intelligence (AI)-empowered interpretation of echocardiographic images, point-of-care dried blood spot testing in the echocardiography laboratory, and widespread dissemination of point-of-care ultrasound devices to community practices to promote screening may lead to more timely diagnosis of FD. Fabry disease should no longer be considered a rare, untreatable disease, but one that can be effectively identified and treated at an early stage before the development of irreversible end-organ damage.
Collapse
Affiliation(s)
- Jeffrey Yim
- Department of Medicine, University of British Columbia, Vancouver, BC V6H 0A5, Canada;
| | - Olivia Yau
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 0A5, Canada;
| | - Darwin F. Yeung
- Vancouver General Hospital and University of British Columbia Echocardiography Laboratory, Division of Cardiology, University of British Columbia, Vancouver, BC V6H 0A5, Canada
- Correspondence: (D.F.Y.); (T.S.M.T.)
| | - Teresa S. M. Tsang
- Vancouver General Hospital and University of British Columbia Echocardiography Laboratory, Division of Cardiology, University of British Columbia, Vancouver, BC V6H 0A5, Canada
- Correspondence: (D.F.Y.); (T.S.M.T.)
| |
Collapse
|
17
|
Massalha S, Slart RHJA. Anderson-Fabry disease: Worthy to in-SPECT the nerves? J Nucl Cardiol 2021; 28:650-652. [PMID: 31218586 DOI: 10.1007/s12350-019-01787-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Samia Massalha
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB, Groningen, The Netherlands.
- Department of Biomedical Photonic Imaging, TechMed Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
18
|
Mattig I, Canaan-Kühl S, Tillmanns C, Knebel F. Progression of electrocardiogram changes in an untreated fabry disease: a case report. EUROPEAN HEART JOURNAL-CASE REPORTS 2021; 5:ytab045. [PMID: 33738419 PMCID: PMC7954241 DOI: 10.1093/ehjcr/ytab045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/21/2020] [Accepted: 01/19/2021] [Indexed: 12/01/2022]
Abstract
Background Fabry disease (FD) is a rare lysosomal storage disorder with multiorgan manifestation and associated with an increased morbidity and mortality. Fabry cardiomyopathy includes left ventricular ‘hypertrophy’ (LVH), cardiac arrhythmias, and heart failure. We report a case of an untreated FD with characteristic findings in electrocardiogram (ECG) over a follow-up period of 10 years. Case summary A 53-year-old man with FD presented to our outpatient department. He suffered from symptomatic ventricular extrasystoles. Echocardiography detected LVH and reduced global longitudinal strain. Twelve years ago, first examination was conducted due to ventricular arrhythmias. Electrocardiogram showed a short PQ minus P-wave (PendQ) interval and negative T-waves. Over time, the number of leads with negative T-waves increased. Moreover, the echocardiography revealed a thickened left ventricular wall. Without any further examinations at that time, the patient was treated for arterial hypertension with proteinuria. Ten years after first symptoms appeared, FD was diagnosed utilizing cardiac magnetic resonance imaging and genetic tests. Hence, enzyme replacement therapy was initiated. Discussion The ECG is a fast diagnostic method and it may — even without additional organ manifestations — provide preliminary suspicion of FD. In particular, as shown in our case, a short PendQ and QT interval indicate FD. Over time, disease progression can be detected through ECG changes. T-waves correlate with an increasing LVH and a reduction in longitudinal function in echocardiographic examinations. Unexplained LVH must be followed by differential diagnosis. In case of confirmed FD, patients should be treated by multidisciplinary teams in experienced centres.
Collapse
Affiliation(s)
- Isabel Mattig
- Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Corresponding author. Tel: +49 30 450 613305, Fax: +49 30 450 7 513932,
| | - Sima Canaan-Kühl
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Fabry Zentrum, Zentrum für seltene Nierenerkrankungen (CeRKiD), Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | - Fabian Knebel
- Medizinische Klinik mit Schwerpunkt Kardiologie und Angiologie, Campus Charité Mitte, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Oudenarder Straße 16, 13347 Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| |
Collapse
|
19
|
Morimoto S, Nojiri A, Fukuro E, Anan I, Kawai M, Sakurai K, Kobayashi M, Kobayashi H, Ida H, Ohashi T, Shibata T, Yoshimura M, Eto Y, Hongo K. Characteristics of the Electrocardiogram in Japanese Fabry Patients Under Long-Term Enzyme Replacement Therapy. Front Cardiovasc Med 2021; 7:614129. [PMID: 33521063 PMCID: PMC7840582 DOI: 10.3389/fcvm.2020.614129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022] Open
Abstract
Objective: An electrocardiogram (ECG) is an important tool for demonstrating cardiac manifestations in various heart diseases. The present study clarified the characteristics of ECG parameters in Japanese Fabry patients under long-term enzyme replacement therapy (ERT). Methods: We analyzed the ECGs of 40 Fabry patients (male, n = 17; female, n = 23) before and after treatment with ERT. To evaluate the atrio-ventricular conduction, the PQ interval, corrected PQ and PQ minus P-wave in lead II (Pend-Q) were calculated. The QRS duration, QTc, Sokolow-Lyon index, and strain pattern were also examined. Results: At the baseline, the shortening of the PQ interval, corrected PQ and Pend-Q was identified in 7.5, 25.0, and 47.5% of cases, respectively. The prolongation of QRS duration and QTc was found in 7.5 and 40.0% of cases, respectively. The strain pattern was mainly identified in female patients, irrespective of left ventricular hypertrophy (LVH). During long-term ERT, the PQ interval, corrected PQ and Pend-Q did not change significantly. The QRS duration was significantly prolonged in both genders, whereas the QTc was significantly prolonged only in male patients. A subgroup analysis revealed that the prolongation of the QRS duration and QTc only occurred in male patients with LVH and only occurred in female patients with the classical type mutation. The prevalence of the strain was significantly increased only in male patients with LVH. Conclusions: These results suggest that the shortening of the Pend-Q is a specific finding in Japanese Fabry patients, and the strain pattern without LVH in female patients can be considered Fabry disease. During long-term ERT, prolongation of the QRS duration and QTc can indicate the progression of myocardial damage in male patients with LVH and in female patients with the classical type mutation.
Collapse
Affiliation(s)
- Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayumi Nojiri
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Eiko Fukuro
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ikuko Anan
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ken Sakurai
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Masahisa Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan.,Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Toya Ohashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan.,Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Shibata
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorders, Kanagawa, Japan
| | - Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
[Fabry disease: A review]. Rev Med Interne 2020; 42:110-119. [PMID: 33172708 DOI: 10.1016/j.revmed.2020.08.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 02/03/2023]
Abstract
Fabry disease is the second most frequent lysosomal storage disorder. It is a X-linked genetic disease secondary to alpha-galactosidase A enzyme deficiency. This is a progressive and systemic disease that affects both males and females. Classical symptoms and organ involvements are acral pain crisis, cornea verticillata, hypertrophic cardiomyopathy, stroke and chronic kidney disease with proteinuria. Nevertheless, organ damages can be missing or pauci-symptomatic and other common symptoms are poorly recognised, such as gastrointestinal or ear involvement. In classical Fabry disease, symptoms first appear during childhood or teenage in males, but later in females. Patients may have non-classical or late-onset Fabry disease with delayed manifestations or with single-organ involvement. Recognition of Fabry disease is important because treatments are available, but it may be challenging. Diagnosis is easy in males, with dosage of alpha-galactosidase A enzyme activity into leukocytes, but more difficult in females who can express normal residual activity. Other plasmatic biomarkers, such as lyso-globotriaosylceramide (lyso-Gb3), are interesting in females, but need to be associated with GLA gene analysis. In this review, we aimed at summarize the main clinical manifestations of Fabry disease and propose a practical algorithm to know how to diagnose this complex disease.
Collapse
|
21
|
Abstract
RATIONALE The most common cardiac involvement of Fabry disease (FD) is left ventricular hypertrophy (LVH), which usually occurs in male patients over the age of 30. In rare cases, it can progress to ventricular dilation in the late stage of the disease. PATIENT CONCERNS A 16-year-old boy presenting with recurrent extremity pain and chest distress was admitted to our hospital. Imaging examinations revealed ventricular dilation. DIAGNOSIS α-Galactosidase A enzyme assay and GLA gene sequencing confirmed the diagnosis of FD and revealed a novel mutation c.76_77insT. INTERVENTIONS The patient was treated using metoprolol (23.75 mg qd) and angiotensin-converting enzyme inhibitor (fosinopril sodium 5 mg qd). He refused enzyme replacement therapy for financial reasons. OUTCOMES The echocardiography, electrocardiography, renal function, and routine blood and urine tests performed 20 months after the patients discharge from hospital showed no significant changes. The patient reported a slow and gradual decrease in the frequency and degree of pain and chest distress, starting approximately 24 months after discharge. LESSONS Cardiac involvement of FD can progress rapidly in some cases. Screening for FD should be considered in patients with unexplained ventricular dilation, especially in those with a history of typical FD manifestations.
Collapse
Affiliation(s)
- Jia Tang
- Peking Union Medical College Hospital (PUMCH)
| | - Chao Wu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases
| | | | - Liang Wang
- Department of Cardiology, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Science (CAMS) and Peking Union Medical College (PUMC), Beijing, China
| |
Collapse
|
22
|
Naresh NK, Misener S, Zhang Z, Yang C, Ruh A, Bertolino N, Epstein FH, Collins JD, Markl M, Procissi D, Carr JC, Allen BA. Cardiac MRI Myocardial Functional and Tissue Characterization Detects Early Cardiac Dysfunction in a Mouse Model of Chemotherapy-Induced Cardiotoxicity. NMR IN BIOMEDICINE 2020; 33:e4327. [PMID: 32567177 DOI: 10.1002/nbm.4327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Doxorubicin and doxorubicin-trastuzumab combination chemotherapy have been associated with cardiotoxicity that eventually leads to heart failure and may limit dose-effective cancer treatment. Current diagnostic strategies rely on decreased ejection fraction (EF) to diagnose cardiotoxicity. PURPOSE The aim of this study is to explore the potential of cardiac MR (CMR) imaging to identify imaging biomarkers in a mouse model of chemotherapy-induced cardiotoxicity. METHODS A cumulative dose of 25 mg/kg doxorubicin was administered over three weeks using subcutaneous pellets (n = 9, Dox). Another group (n = 9) received same dose of Dox and a total of 10 mg/kg trastuzumab (DT). Mice were imaged at baseline, 5/6 weeks and 10 weeks post-treatment on a 7T MRI system. The protocol included short-axis cine MRI covering the left ventricle (LV) and mid-ventricular short-axis tissue phase mapping (TPM), pre- and post-contrast T1 mapping, T2 mapping and Displacement Encoding with Stimulated Echoes (DENSE) strain encoded MRI. EF, peak myocardial velocities, native T1, T2, extracellular volume (ECV), and myocardial strain were quantified. N = 7 mice were sacrificed for histopathologic assessment of apoptosis at 5/6 weeks. RESULTS Global peak systolic longitudinal velocity was reduced at 5/6 weeks in Dox (0.6 ± 0.3 vs 0.9 ± 0.3, p = 0.02). In the Dox group, native T1 was reduced at 5/6 weeks (1.3 ± 0.2 ms vs 1.6 ± 0.2 ms, p = 0.02), and relatively normalized at week 10 (1.4 ± 0.1 ms vs 1.6 ± 0.2 ms, p > 0.99). There was no change in EF and other MRI parameters and histopathologic results demonstrated minimal apoptosis in all mice (~1-2 apoptotic cell/high power field), suggesting early-stage cardiotoxicity. CONCLUSIONS In a mouse model of chemotherapy-induced cardiotoxicity using doxorubicin and trastuzumab, advanced CMR shows promise in identifying treatment-related decrease in myocardial velocity and native T1 prior to the onset of cardiomyocyte apoptosis and reduction of EF.
Collapse
Affiliation(s)
- Nivedita K Naresh
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Sol Misener
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Zhouli Zhang
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Cynthia Yang
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Alexander Ruh
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Nicola Bertolino
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Frederick H Epstein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Jeremy D Collins
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Michael Markl
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
- McCormick School of Engineering, Northwestern University, Chicago, IL, USA
| | - Daniele Procissi
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - James C Carr
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| | - Bradley A Allen
- Department of Radiology, Northwestern University, 737 N. Michigan Ave, Chicago, IL, USA
| |
Collapse
|
23
|
Michaud M, Mauhin W, Belmatoug N, Garnotel R, Bedreddine N, Catros F, Ancellin S, Lidove O, Gaches F. When and How to Diagnose Fabry Disease in Clinical Pratice. Am J Med Sci 2020; 360:641-649. [PMID: 32723516 DOI: 10.1016/j.amjms.2020.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/30/2020] [Accepted: 07/09/2020] [Indexed: 01/27/2023]
Abstract
Fabry disease is a frequent lysosomal storage disorder secondary to the deficiency of alpha-galactosidase A enzyme. This X-linked genetic disease realizes progressive and systemic manifestations that affect both male and female. Fabry disease may present as "classical", as "late-onset" or "non-classical" forms. Symptoms and organ involvements of classical Fabry disease are acral pain crisis, cornea verticillata, hypertrophic cardiomyopathy, stroke and chronic kidney disease with proteinuria. Other common symptoms are often poorly recognized, such as gastrointestinal or ear involvements. In classical Fabry disease, symptoms first appear during childhood or during teenage years in males, but later in females. Patients with non-classical or late-onset Fabry disease have delayed manifestations or a single-organ involvement. Diagnosis is therefore difficult when classical organ involvements are missing, in paucisymptomatic patients or in late-onset forms. Recognition of Fabry disease is important because effective treatments are available. They have to be prescribed early. In male, diagnosis is made with alpha-galactosidase A enzyme activity dosage in leukocyte, that is very low or null in classical forms and under 30 percent in late-onset forms. Diagnosis is more challenging in females who may express normal residual enzyme activity. Other plasmatic biomarkers, such as lyso-globotriaosylceramide are interesting, especially in females. In this review, we aimed to summarize main clinical manifestations of Fabry disease to know when to evoke Fabry disease and propose a practical diagnosis algorithm to know how to diagnose.
Collapse
Affiliation(s)
- Martin Michaud
- Department of Internal Medicine, Hopital Joseph Ducuing, Toulouse, France; Competence Center for Lysosomal Storage Diseases, Hopital Joseph Ducuing, Toulouse, France.
| | - Wladimir Mauhin
- Internal Medicine and Rheumatology Department, Diaconesses-Croix Saint Simon Hospital Group, Paris, France; Referral Center for Lysosomal Diseases, site Avron, Paris, France
| | - Nadia Belmatoug
- Department of Internal Medicine, University Hospital Paris Nord Val de Seine, Assistance Publique-Hôpitaux de Paris, Clichy, France; Referral Center for Lysosomal Diseases, University Hospital Paris Nord Val de Seine, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Roselyne Garnotel
- Biochemistry Laboratory, American Memorial Hospital Reims, Reims, France
| | - Naiya Bedreddine
- Association des patients de la maladie de Fabry, Marsannay La Cote, France
| | - Florian Catros
- Department of Internal Medicine, Hopital Joseph Ducuing, Toulouse, France; Competence Center for Lysosomal Storage Diseases, Hopital Joseph Ducuing, Toulouse, France
| | - Sophie Ancellin
- Department of Internal Medicine, Hopital Joseph Ducuing, Toulouse, France; Competence Center for Lysosomal Storage Diseases, Hopital Joseph Ducuing, Toulouse, France
| | - Olivier Lidove
- Internal Medicine and Rheumatology Department, Diaconesses-Croix Saint Simon Hospital Group, Paris, France; Referral Center for Lysosomal Diseases, site Avron, Paris, France
| | - Francis Gaches
- Department of Internal Medicine, Hopital Joseph Ducuing, Toulouse, France; Competence Center for Lysosomal Storage Diseases, Hopital Joseph Ducuing, Toulouse, France
| |
Collapse
|
24
|
Joye E, Zheng WQ, Torosoff M. Pulse‐cancellation echocardiography in Fabry disease diagnosis. Echocardiography 2020; 37:908-912. [DOI: 10.1111/echo.14707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Evan Joye
- Department of Cardiology Albany Medical College Albany NY USA
| | - Wen Qian Zheng
- Department of Internal Medicine Albany Medical College Albany NY USA
| | | |
Collapse
|
25
|
Nojiri A, Anan I, Morimoto S, Kawai M, Sakuma T, Kobayashi M, Kobayashi H, Ida H, Ohashi T, Eto Y, Shibata T, Yoshimura M, Hongo K. Clinical findings of gadolinium-enhanced cardiac magnetic resonance in Fabry patients. J Cardiol 2019; 75:27-33. [PMID: 31623930 DOI: 10.1016/j.jjcc.2019.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/14/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Fabry disease is one of the causes of left ventricular hypertrophy (LVH) and can be treated with enzyme replacement therapy or pharmacological chaperone therapy. Late gadolinium enhancement (LGE) in cardiac magnetic resonance (CMR) can identify myocardial fibrosis and be used for the stratification in LVH. However, the details of the prevalence and characteristics of LGE in Japanese Fabry patients have not been reported. METHODS We evaluated myocardial involvement in 26 Fabry patients (10 males, 16 females) using gadolinium-enhanced CMR. LGE areas were analyzed using the previously reported scoring method. Echocardiography was also performed to evaluate the left ventricular function and left ventricular mass. RESULTS LGE on CMR images was positive in 5 out of 26 patients, and all patients with LGE-positive findings suffered from LVH (2 out of 5 male patients and 3 out of 4 female patients with LVH on echocardiography). LGE was specifically localized at the mid-wall in the infero-lateral area of the left ventricle. LGE-positive patients seemed to be older, and tended to have a larger left ventricular mass index and higher B-type natriuretic peptide level than LGE-negative patients. CONCLUSIONS These results revealed that specific localization of LGE was present in Fabry patients.
Collapse
Affiliation(s)
- Ayumi Nojiri
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ikuko Anan
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Toru Sakuma
- Department of Radiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masahisa Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan; Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan; Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Toya Ohashi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan; Division of Gene Therapy, Research Center for Molecular Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorders, Kanagawa, Japan
| | - Takahiro Shibata
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| |
Collapse
|
26
|
Arbeláez-Cortés Á, Quintero-González DC, Cuesta-Astroz Y, Villadiego JS, González-Buriticá H, Rueda JM. Restrictive cardiomyopathy in a patient with systemic sclerosis and Fabry disease: a case-based review. Rheumatol Int 2019; 40:489-497. [PMID: 31599343 DOI: 10.1007/s00296-019-04453-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/26/2019] [Indexed: 12/29/2022]
Abstract
Systemic sclerosis (SSc) is a rare immune-mediated vasculopathy characterized by fibrosis of the skin and internal organs. Fabry disease (FD) is an X-linked lysosomal storage disorder caused by mutations in the GLA gene producing α-galactosidase-A enzyme (α-Gal A) deficiency. Being a systemic disease, cardiac involvement in FD has a high mortality rate due to heart failure and arrhythmia. The coexistence of these two entities has not been reported previously. We describe the case of a female patient with limited SSc (lcSSc), a diagnosis based on the presence of sclerodactyly, Raynaud phenomenon, microvascular involvement, and positive anti-centromere antibodies. On follow-up, she developed chest pain, a second-degree A-V block, and restrictive cardiomyopathy (without cardiovascular risk factors). Although heart involvement is common in these two entities, the abnormal thickening of lateral and inferior wall, the infiltration pattern and the conduction system disorders presented herein are more characteristic in a heterozygous female with a cardiac variant of FD. The diagnosis of FD was confirmed with high globotriaosylsphingosine (Lyso-Gb3) levels and identification of GLA gene mutation. The patient was treated with enzymatic replacement (agalsidase alpha) following mild improvement in ventricular mass at 6th month, without clinical deterioration. The related literature on SSc associated with FD is also reviewed.
Collapse
Affiliation(s)
- Álvaro Arbeláez-Cortés
- Internal Medicine, Universidad Libre, Cali, Colombia. .,Arthritis and Rheumatology Clinic, Centro Médico Imbanaco, Cali, Colombia.
| | | | - Yesid Cuesta-Astroz
- School of Microbiology, Universidad de Antioquia, Medellín, Colombia.,Instituto Colombiano de Medicina Tropical, Universidad CES, Sabaneta, Colombia
| | | | - Herman González-Buriticá
- Internal Medicine, Universidad Libre, Cali, Colombia.,Arthritis and Rheumatology Clinic, Centro Médico Imbanaco, Cali, Colombia
| | - Jorge M Rueda
- Internal Medicine, Universidad Libre, Cali, Colombia.,Arthritis and Rheumatology Clinic, Centro Médico Imbanaco, Cali, Colombia
| |
Collapse
|
27
|
Sepehri Shamloo A, Schoene K, Stauber A, Darma A, Dagres N, Dinov B, Bertagnolli L, Hilbert S, Müssigbrodt A, Husser D, Bollmann A, Hindricks G, Arya A. Epicardial adipose tissue thickness as an independent predictor of ventricular tachycardia recurrence following ablation. Heart Rhythm 2019; 16:1492-1498. [DOI: 10.1016/j.hrthm.2019.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Indexed: 01/20/2023]
|
28
|
Chaves-Markman ÂV, Markman M, Calado EB, Pires RF, Santos-Veloso MAO, Pereira CMF, Lordsleem ABDMDS, Lima SGD, Markman Filho B, Oliveira DCD. GLA Gene Mutation in Hypertrophic Cardiomyopathy with a New Variant Description: Is it Fabry's Disease? Arq Bras Cardiol 2019; 113:77-84. [PMID: 31291414 PMCID: PMC6684188 DOI: 10.5935/abc.20190112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is an X-linked lysosomal storage disorder caused by mutations in the alpha galactosidase A gene (GLA) that lead to the enzymatic deficiency of alpha galactosidase (α-Gal A), resulting in the accumulation of globotriaosylceramide (Gb3) and globotriaosylsphingosine (lyso-Gb3), causing multiple organ dysfunctions. OBJECTIVE To perform GLA gene screening in a group of patients with echocardiographic diagnosis of hypertrophic cardiomyopathy (HCM). METHODS a cross-sectional study was conducted with HCM patients from a university hospital. Patients with coronary artery disease and valvulopathies were excluded. Mutation analysis of the GLA gene was performed. In male subjects, the analysis was performed after evidence of low α-Gal A activity. RESULTS 60 patients with echocardiographic diagnosis of HCM were included. Age ranged from 12 to 85 years and 60% were women. Mean myocardial fibrosis percentage on MRI was 10.7 ± 13.1% and mean ventricular thickness was18.7 ± 6.7 mm. Four patients had the following GLA gene mutations: c.967C>A (p.Pro323Thr), not yet described in the literature; c.937G>T (p.Asp313Tyr); and c.352C>T (p.Arg118Cys). All patients had normal levels of lyso-Gb3 and non-ischemic myocardial fibrosis on magnetic resonance imaging; one patient had proteinuria and one patient had ventricular tachycardia. CONCLUSION in this study, the frequency of mutation in the GLA gene in patients with HCM was 6.7%. A novel mutation in exon 6 of the GLA gene, c.967C>A (p.Pro323Thr), was identified. Patients with HCM may have GLA mutations and FD should be ruled out. Plasma (lyso-Gb3) levels do not seem to be sufficient to attain a diagnosis and organ biopsy should be considered.
Collapse
Affiliation(s)
- Ândrea Virgínia Chaves-Markman
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil.,Hospital Agamenon Magalhães, Recife, PE - Brazil.,Universidade Federal de Pernambuco - Grupo de pesquisa em Epidemiologia e Cardiologia (EPICARDIO), Recife, PE - Brazil.,Pós Graduação em Inovação Terapêutica (PPGIT) - Universidade Federal de Pernambuco, Recife, PE - Brasil.,Centro Universitário Mauricio de Nassau - Curso de Medicina, Recife, PE - Brasil
| | | | - Eveline Barros Calado
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil
| | | | - Marcelo Antônio Oliveira Santos-Veloso
- Universidade Federal de Pernambuco - Grupo de pesquisa em Epidemiologia e Cardiologia (EPICARDIO), Recife, PE - Brazil.,Pós Graduação em Inovação Terapêutica (PPGIT) - Universidade Federal de Pernambuco, Recife, PE - Brasil.,Centro Universitário Mauricio de Nassau - Curso de Medicina, Recife, PE - Brasil
| | | | - Andréa Bezerra de Melo da Silveira Lordsleem
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil.,Universidade Federal de Pernambuco - Grupo de pesquisa em Epidemiologia e Cardiologia (EPICARDIO), Recife, PE - Brazil.,Centro Universitário Mauricio de Nassau - Curso de Medicina, Recife, PE - Brasil
| | - Sandro Gonçalves de Lima
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil.,Universidade Federal de Pernambuco - Grupo de pesquisa em Epidemiologia e Cardiologia (EPICARDIO), Recife, PE - Brazil.,Centro Universitário Mauricio de Nassau - Curso de Medicina, Recife, PE - Brasil
| | - Brivaldo Markman Filho
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil
| | - Dinaldo Cavalcanti de Oliveira
- Universidade Federal de Pernambuco - Hospital das Clínicas - Área Acadêmica de Medicina Clínica - Centro de Medicina Clínica - CCM, Recife, PE - Brazil.,Pós Graduação em Inovação Terapêutica (PPGIT) - Universidade Federal de Pernambuco, Recife, PE - Brasil.,Centro Universitário Mauricio de Nassau - Curso de Medicina, Recife, PE - Brasil
| |
Collapse
|
29
|
Baig S, Edward NC, Kotecha D, Liu B, Nordin S, Kozor R, Moon JC, Geberhiwot T, Steeds RP. Ventricular arrhythmia and sudden cardiac death in Fabry disease: a systematic review of risk factors in clinical practice. Europace 2019; 20:f153-f161. [PMID: 29045633 DOI: 10.1093/europace/eux261] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/10/2017] [Indexed: 01/08/2023] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder caused by deficiency of α-galactosidase A enzyme. Cardiovascular (CV) disease is a common cause of mortality in FD, in particular as a result of heart failure and arrhythmia, with a significant proportion of events categorized as sudden. There are no clear models for risk prediction in FD. This systematic review aims to identify the risk factors for ventricular arrhythmia (VA) and sudden cardiac deaths (SCD) in FD. A systematic search was performed following PRISMA guidelines of EMBASE, Medline, PubMed, Web of Science, and Cochrane from inception to August 2016, focusing on identification of risk factors for the development of VA or SCD. Thirteen studies were included in the review (n = 4185 patients) from 1189 articles, with follow-up of 1.2-10 years. Weighted average age was 37.6 years, and 50% were male. Death from any cause was reported in 8.3%. Of these, 75% was due to CV problems, with the majority being SCD events (62% of reported deaths). Ventricular tachycardia was reported in 7 studies, with an average prevalence of 15.3%. Risk factors associated with SCD events were age, male gender, left ventricular hypertrophy, late gadolinium enhancement on CV magnetic resonance imaging, and non-sustained ventricular tachycardia. Although a multi-system disease, FD is a predominantly cardiac disease from a mortality perspective, with death mainly from SCD events. Limited evidence highlights the importance of clinical and imaging risk factors that could contribute to improved decision-making in the management of FD.
Collapse
Affiliation(s)
- Shanat Baig
- Department of Cardiology, First Floor, Nuffield House, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Nicky C Edward
- Department of Cardiology, First Floor, Nuffield House, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Dipak Kotecha
- Department of Cardiology, First Floor, Nuffield House, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Boyang Liu
- Department of Cardiology, First Floor, Nuffield House, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Sabrina Nordin
- Barts Heart Centre, London, UK.,Institute of Cardiovascular Science, University College London, London, UK
| | - Rebecca Kozor
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - James C Moon
- Barts Heart Centre, London, UK.,Institute of Cardiovascular Science, University College London, London, UK
| | - Tarekegn Geberhiwot
- Centre for Rare Diseases, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Richard P Steeds
- Department of Cardiology, First Floor, Nuffield House, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
30
|
Vijapurapu R, Kozor R, Hughes DA, Woolfson P, Jovanovic A, Deegan P, Rusk R, Figtree GA, Tchan M, Whalley D, Kotecha D, Leyva F, Moon J, Geberhiwot T, Steeds RP. A randomised controlled trial evaluating arrhythmia burden, risk of sudden cardiac death and stroke in patients with Fabry disease: the role of implantable loop recorders (RaILRoAD) compared with current standard practice. Trials 2019; 20:314. [PMID: 31151481 PMCID: PMC6544923 DOI: 10.1186/s13063-019-3425-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/11/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Fabry disease (FD) is a genetic disorder caused by a deficiency in the enzyme alpha-galactosidase A, leading to an accumulation of glycosphingolipids in tissues across the body. Cardiac disease is the leading cause of morbidity and mortality. Advanced disease, characterised by extensive left ventricular hypertrophy, ventricular dysfunction and fibrosis, is known to be associated with an increase in arrhythmia. Data identifying risk factors for arrhythmia are limited, and no Fabry-specific risk stratification tool is available to select those who may benefit from initiation of medical or device therapy (implantable cardiac defibrillators). Current monitoring strategies have a limited diagnostic yield, and implantable loop recorders (ILRs) have the potential to change treatment and clinical outcomes. AIM The aim of this study is to determine whether ILRs can (1) improve arrhythmia detection in FD and (2) identify risk predictors of arrhythmia. METHODS A prospective, 5-year, open-label, international, multi-centre randomised controlled trial of a minimum of 164 participants with genetically or enzymatically confirmed FD (or both) who have evidence of cardiac disease will be recruited from five centres: Queen Elizabeth Hospital, Birmingham, UK; Salford Royal Hospital, Salford, UK; Royal Free Hospital, London, UK; Addenbrookes Hospital, Cambridge, UK; and Westmead Hospital, Sydney, Australia. Participants will be block-randomised (1:1) to two study arms for cardiac monitoring (i) control arm: standard of care with annual 24 h or 5-day Holter monitor or (ii) treatment arm: continuous cardiac monitoring with ILR implantation plus standard of care. Participants will undergo multiple investigations-blood/urine biomarkers, 12-lead and advanced electrocardiogram (ECG) recording, echocardiography and cardiovascular magnetic resonance (CMR) imaging-at baseline and 6-12 monthly follow-up visits. The primary endpoint is identification of arrhythmia requiring initiation or alteration in therapy. Secondary outcome measures include characterising the risk factors associated with arrhythmia and outcome data in the form of imaging, ECG and blood biomarkers. DISCUSSION This is the first study evaluating arrhythmia burden and the use of ILR across the spectrum of risk profiles in Fabry cardiomyopathy. This will enable detailed characterisation of arrhythmic risk predictors in FD and ultimately support formulation of Fabry-specific guidance in this high-risk population. TRIAL REGISTRATION ClinicalTrials.gov ( NCT03305250 ). Registered on 9 October 2017.
Collapse
Affiliation(s)
- Ravi Vijapurapu
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Mindlesohn Way, Birmingham, B15 2TH UK
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, Mindlesohn Way, Birmingham, B15 2TH UK
| | - Rebecca Kozor
- Sydney Medical School, University of Sydney, Sydney, 2006 NSW Australia
- Cardiology Department, Royal North Shore Hospital, Reserve Road, St. Leonards, NSW 2065 Australia
| | - Derralynn A. Hughes
- Lysosomal Storage Disorder Unit, Royal London NHS Foundation Trust, University College London, Pond Street, London, NW3 2QG UK
| | - Peter Woolfson
- Department of Cardiology, Salford Royal Hospital, Stott Lane, Salford, M6 8HD UK
| | - Ana Jovanovic
- Mark Holland Metabolic Unit, Salford Royal Hospital, Stott Lane, Salford, M6 8HD UK
| | - Patrick Deegan
- Department of Medicine, Addenbrooke’s Hospital, Hill Road, Cambridge, CB2 0QQ UK
| | - Rosemary Rusk
- Department of Cardiology, Addenbrookes Hospital, Hill Road, Cambridge, CB2 0QQ UK
| | - Gemma A. Figtree
- Sydney Medical School, University of Sydney, Sydney, 2006 NSW Australia
- Cardiology Department, Royal North Shore Hospital, Reserve Road, St. Leonards, NSW 2065 Australia
| | - Michel Tchan
- Sydney Medical School, University of Sydney, Sydney, 2006 NSW Australia
- Department of Genetics, Westmead Hospital, Hawkesbury Road, Westmead, NSW 2145 Australia
| | - David Whalley
- Sydney Medical School, University of Sydney, Sydney, 2006 NSW Australia
- Cardiology Department, Royal North Shore Hospital, Reserve Road, St. Leonards, NSW 2065 Australia
| | - Dipak Kotecha
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Mindlesohn Way, Birmingham, B15 2TH UK
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Francisco Leyva
- Aston Medical Research Institute, Aston Medical School, Birmingham, B4 7ET UK
| | - James Moon
- Barts Heart Centre, Barts Health NHS Trust, 16-18 Westmoreland Street, London, W1G 8PH UK
| | - Tarekegn Geberhiwot
- Department of Endocrinology, Queen Elizabeth Hospital Birmingham, Mindlesohn Way, Birmingham, B15 2TH UK
- Institute of Metabolism and System Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Richard P. Steeds
- Department of Cardiology, Queen Elizabeth Hospital Birmingham, Mindlesohn Way, Birmingham, B15 2TH UK
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| |
Collapse
|
31
|
Hernández-Romero D, Sánchez-Quiñones J, Vílchez JA, Rivera-Caravaca JM, de la Morena G, Lip GYH, Climent V, Marín F. Galectin-3 and β-trace protein concentrations are higher in clinically unaffected patients with Fabry disease. Sci Rep 2019; 9:6235. [PMID: 30996283 PMCID: PMC6470309 DOI: 10.1038/s41598-019-42727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/05/2019] [Indexed: 11/13/2022] Open
Abstract
Current therapies have not shown benefit in organ damage reversal in Fabry disease (FD), but biomarkers could help risk stratification and prognosis. We investigated if several biomarkers of cardiac fibrosis, cardiac wall stress, myocardial injury, renal function and inflammation, are associated with early cardiac affectation in FD patients. We included FD patients from four cardiology outpatient clinics of southeastern Spain. At inclusion, Galectin-3 (Gal-3), N-terminal proB-type natriuretic peptide, high sensitivity troponin T (hsTnT), β-trace protein (BTP) and interleukin-6 concentrations were measured. The relation of biomarkers concentrations with clinical features, cardiac involvement and organ affectation according to the Mainz Severity Score Index (MSSI) was investigated. 44 FD patients (n = 21 affected and n = 23 unaffected) were compared to age and sex-respectively matched healthy controls. Significant differences in biomarkers’ concentration between FD groups were observed. Importantly, Gal-3 and BTP levels were higher in unaffected patients when compared with age and sex-matched healthy controls (both p < 0.05). All the biomarkers correlated with clinical features. When cut-off values for clinical affectation (measured as MSSI ≥ 20) were established, only hsTnT (OR 30.69, 95% CI 2.70–348.42) and male sex (OR 8.17, 95% CI 1.16–57.75) were independently associated with cardiac damage by multivariate regression analysis. Gal-3 and BTP levels are increased in unaffected FD patients compared to healthy controls. This suggests that these biomarkers could be useful for the early detection of cardiac affectation in FD patients. On the other hand, hsTnT and male sex are independent risk factors for established clinical cardiac damage in FD.
Collapse
Affiliation(s)
- Diana Hernández-Romero
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, CIBERCV, Murcia, Spain.
| | | | - Juan Antonio Vílchez
- Department of Clinical Analysis, Hospital General Universitario Santa Lucía, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Cartagena, Spain
| | - José Miguel Rivera-Caravaca
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, CIBERCV, Murcia, Spain
| | - Gonzalo de la Morena
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, CIBERCV, Murcia, Spain
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, United Kingdom.,Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Vicente Climent
- Department of Cardiology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Francisco Marín
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), University of Murcia, CIBERCV, Murcia, Spain
| |
Collapse
|
32
|
Zhu X, Yin L, Theisen M, Zhuo J, Siddiqui S, Levy B, Presnyak V, Frassetto A, Milton J, Salerno T, Benenato KE, Milano J, Lynn A, Sabnis S, Burke K, Besin G, Lukacs CM, Guey LT, Finn PF, Martini PG. Systemic mRNA Therapy for the Treatment of Fabry Disease: Preclinical Studies in Wild-Type Mice, Fabry Mouse Model, and Wild-Type Non-human Primates. Am J Hum Genet 2019; 104:625-637. [PMID: 30879639 DOI: 10.1016/j.ajhg.2019.02.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 11/27/2022] Open
Abstract
Fabry disease is an X-linked lysosomal storage disease caused by loss of alpha galactosidase A (α-Gal A) activity and is characterized by progressive accumulation of globotriaosylceramide and its analogs in all cells and tissues. Although enzyme replacement therapy (ERT) is considered standard of care, the long-term effects of ERT on renal and cardiac manifestations remain uncertain and thus novel therapies are desirable. We herein report preclinical studies evaluating systemic messenger RNA (mRNA) encoding human α-Gal A in wild-type (WT) mice, α-Gal A-deficient mice, and WT non-human primates (NHPs). The pharmacokinetics and distribution of h-α-Gal A mRNA encoded protein in WT mice demonstrated prolonged half-lives of α-Gal A in tissues and plasma. Single intravenous administration of h-α-Gal A mRNA to Gla-deficient mice showed dose-dependent protein activity and substrate reduction. Moreover, long duration (up to 6 weeks) of substrate reductions in tissues and plasma were observed after a single injection. Furthermore, repeat i.v. administration of h-α-Gal A mRNA showed a sustained pharmacodynamic response and efficacy in Fabry mice model. Lastly, multiple administrations to non-human primates confirmed safety and translatability. Taken together, these studies across species demonstrate preclinical proof-of-concept of systemic mRNA therapy for the treatment of Fabry disease and this approach may be useful for other lysosomal storage disorders.
Collapse
|
33
|
Jaurretche S, Antongiovanni N, Perretta F. Fabry nephropathy. Role of nephrologist and clinical variables associated with the diagnosis. Nefrologia 2019; 39:294-300. [PMID: 30890379 DOI: 10.1016/j.nefro.2018.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 09/20/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The early detection of Fabry nephropathy is of interest to us. Its treatment is more effective in early stages. It has been studied by analysing molecular and tissue biomarkers. These have certain disadvantages that hinder its routine use. The aim of this study is to describe the role of the nephrologist in the diagnosis of the disease, and to describe the clinical variables associated with nephropathy in affected patients. MATERIAL AND METHODS Cross-sectional study. Patients were included from three reference centres in Argentina. RESULTS Seventy two patients were studied (26.26±16.48years): 30 of which (41.6%) were men and 42 of which (58.4%) were women; 27 paediatric patients and 45 adults. Fourteen "index cases" were detected, 50% of which were diagnosed by nephrologists. Nephropathy was found in 44 patients (61%): 6 paediatric patients and 38 adults. Two types of clinical variables were associated with nephropathy: (i)peripheral nervous system compromise (P≤.001), angiokeratomas (P≤.001) and auditory compromise (P=.01-.001), with these being early clinical manifestations of the most severe disease phenotype, and (ii)structural heart disease (P=.01-.001) and central nervous system compromise (P=.05-.01), which are major and late complications, responsible for increased morbidity and mortality and lower life expectancy. CONCLUSION The nephrologist plays an important role in the diagnosis of Fabry nephropathy, although the detection thereof owing to its renal involvement would represent a late diagnosis, because nephropathy is associated with late complications of the most severe disease phenotype.
Collapse
Affiliation(s)
- Sebastián Jaurretche
- Centro de Neurociencias Los Manantiales, Grupo Gamma Rosario, Rosario, Santa Fe, Argentina; Cátedra de Biofísica y Fisiología, Instituto Universitario Italiano de Rosario, Rosario, Santa Fe, Argentina.
| | - Norberto Antongiovanni
- Centro de Infusión y Estudio de Enfermedades Lisosomales, Instituto de Nefrología Clínica Pergamino, Pergamino, Buenos Aires, Argentina
| | - Fernando Perretta
- Servicio de Terapia Intensiva, Hospital Dr. Enrique Erill de Escobar, Belén de Escobar, Buenos Aires, Argentina
| |
Collapse
|
34
|
Czepluch FS, Hasenfuß G. [Inherited heart diseases and storage diseases with cardiac involvement]. Internist (Berl) 2018; 59:1063-1073. [PMID: 30178098 DOI: 10.1007/s00108-018-0485-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Rare diseases mostly have a genetic cause. Many rare cardiovascular diseases also have a genetic cause. For target-oriented cardiogenetic diagnostics, expert knowledge in human genetics as well as in clinical cardiology is needed. In recent years, the genetic cause of a number of heart diseases have been, at least in part, elucidated. Especially, certain arrhythmias and cardiomyopathy forms have a monogenetic cause. An early genetic diagnosis means that patients can be treated more effectively. Rare storage diseases also usually have a genetic cause and can manifest themselves in the heart; prominent examples are Fabry disease and amyloidosis. As patients with Fabry disease or amyloidosis suffer from a diverse and variable symptomatology, the correct diagnosis is often difficult.
Collapse
Affiliation(s)
- Frauke S Czepluch
- Klinik für Kardiologie und Pneumologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland.
| | - Gerd Hasenfuß
- Klinik für Kardiologie und Pneumologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Deutschland
| |
Collapse
|
35
|
Powell AW, Jefferies JL, Hopkin RJ, Mays WA, Goa Z, Chin C. Cardiopulmonary fitness assessment on maximal and submaximal exercise testing in patients with Fabry disease. Am J Med Genet A 2018; 176:1852-1857. [PMID: 30178908 DOI: 10.1002/ajmg.a.40369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/26/2022]
Abstract
The cardiopulmonary exercise test (CPET) is a valuable tool to assess a patient's aerobic fitness and cardiac function, including the response to stress. There have been few studies using CPET to evaluate cardiopulmonary exercise capacity in patients with Fabry disease. We performed a retrospective chart review of patients with Fabry disease from 2001 to 2016, compared to age, gender, and size-matched normal controls. A total of 18 patients were evaluated using the Bruce protocol (treadmill) and 11 patients were evaluated with the ramp protocol (cycle ergometer). The Fabry group demonstrated significantly lower heart rate at peak exercise (151.2 ± 22.5 vs. 178.6 ± 16.2, p < .05), max indexed VO2 (23.7 ± 7 vs. 33.9 ± 8.4, p < .05), and peak index oxygen pulse (12.1 ± 3 vs. 15.2 ± 4.2, p < .05). When the groups were further separated into treadmill or cycle ergometry testing only, there remained statistically significant differences in peak indexed oxygen pulse, heart rate at peak exercise, and max indexed VO2 . There was a statistically significant difference between the Fabry patients evaluated by treadmill testing for systolic blood pressure at peak exercise that was not seen in the cycle ergometry group. Additionally, when looking at the patients who had concurrent cardiac MRI (cMRI) with their CPET, there was a positive correlation with max indexed VO2 and right ventricular end-diastolic volume (r = .55, p = .007) and end-systolic volume (r = .59, p = .007). Patients with Fabry disease have impaired cardiopulmonary exercise capacity as measured by CPET. Additionally, in patients with Fabry disease there is a positive correlation with functional capacity and right ventricular volumes on cMRI.
Collapse
Affiliation(s)
- Adam W Powell
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John L Jefferies
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Robert J Hopkin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Wayne A Mays
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Zhiqian Goa
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Clifford Chin
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
36
|
Lenders M, Brand E. Effects of Enzyme Replacement Therapy and Antidrug Antibodies in Patients with Fabry Disease. J Am Soc Nephrol 2018; 29:2265-2278. [PMID: 30093456 DOI: 10.1681/asn.2018030329] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Malte Lenders
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany
| | - Eva Brand
- Internal Medicine D, Department of Nephrology, Hypertension and Rheumatology, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
37
|
Favalli V, Serio A, Giuliani LP, Arbustini E. 'Precision and personalized medicine,' a dream that comes true? J Cardiovasc Med (Hagerstown) 2018; 18 Suppl 1:e1-e6. [PMID: 27661611 DOI: 10.2459/jcm.0000000000000423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Valentina Favalli
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation, Policlinico San Matteo, University Hospital, Pavia, Italy
| | | | | | | |
Collapse
|
38
|
Trujillo-Quintero JP, Palomino-Doza J, Cárdenas-Reyes I, Ochoa JP, Monserrat L. Abordaje de las cardiopatías familiares desde la Medicina genómica. REVISTA COLOMBIANA DE CARDIOLOGÍA 2018. [DOI: 10.1016/j.rccar.2018.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
39
|
Wanner C, Arad M, Baron R, Burlina A, Elliott PM, Feldt-Rasmussen U, Fomin VV, Germain DP, Hughes DA, Jovanovic A, Kantola I, Linhart A, Mignani R, Monserrat L, Namdar M, Nowak A, Oliveira JP, Ortiz A, Pieroni M, Spada M, Tylki-Szymańska A, Tøndel C, Viana-Baptista M, Weidemann F, Hilz MJ. European expert consensus statement on therapeutic goals in Fabry disease. Mol Genet Metab 2018; 124:189-203. [PMID: 30017653 DOI: 10.1016/j.ymgme.2018.06.004] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/31/2018] [Accepted: 06/10/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Fabry disease, an inherited lysosomal storage disorder, causes multi-organ pathology resulting in substantial morbidity and a reduced life expectancy. Although Fabry disease is an X-linked disorder, both genders may be affected, but generally to a lesser extent in females. The disease spectrum ranges from classic early-onset disease to non-classic later-onset phenotypes, with complications occurring in multiple organs or being confined to a single organ system depending on the stage of the disease. The impact of therapy depends upon patient- and disease-specific factors and timing of initiation. METHODS A European panel of experts collaborated to develop a set of organ-specific therapeutic goals for Fabry disease, based on evidence identified in a recent systematic literature review and consensus opinion. RESULTS A series of organ-specific treatment goals were developed. For each organ system, optimal treatment strategies accounted for inter-patient differences in disease severity, natural history, and treatment responses as well as the negative burden of therapy and the importance of multidisciplinary care. The consensus therapeutic goals and proposed patient management algorithm take into account the need for early disease-specific therapy to delay or slow the progression of disease as well as non-specific adjunctive therapies that prevent or treat the effects of organ damage on quality of life and long-term prognosis. CONCLUSIONS These consensus recommendations help advance Fabry disease management by considering the balance between anticipated clinical benefits and potential therapy-related challenges in order to facilitate individualized treatment, optimize patient care and improve quality of life.
Collapse
Affiliation(s)
- Christoph Wanner
- Division of Nephrology, University Clinic, University of Würzburg, Würzburg, Germany.
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Tel Aviv University, Israel
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | | | - Perry M Elliott
- Barts Heart Centre, University College London, London, United Kingdom
| | - Ulla Feldt-Rasmussen
- Department of Medical Endocrinology, Section 2132, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Victor V Fomin
- I.M. Sechenov First Moscow State Medical University, Department of Internal Diseases No. 1, Moscow, Russian Federation
| | - Dominique P Germain
- French Referral Center for Fabry disease, Division of Medical Genetics and INSERM U1179, University of Versailles, Paris-Saclay University, Montigny, France
| | - Derralynn A Hughes
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free London NHS Foundation Trust, University College London, United Kingdom
| | - Ana Jovanovic
- Mark Holland Metabolic Unit, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Ilkka Kantola
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Aleš Linhart
- Second Department of Medicine - Department of Cardiovascular Medicine, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Renzo Mignani
- Department of Nephrology, Infermi Hospital, Rimini, Italy
| | | | - Mehdi Namdar
- Service de Cardiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Albina Nowak
- University Heart Center, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| | - João-Paulo Oliveira
- Department of Genetics, São João Hospital Centre & Faculty of Medicine and "Instituto de Investigação e Inovação em Saúde (i3S)", University of Porto, Porto, Portugal
| | - Alberto Ortiz
- Unidad de Diálisis, IIS-Fundación Jiménez Díaz/UAM, IRSIN and REDINREN, Madrid, Spain
| | | | - Marco Spada
- Department of Paediatrics, University of Torino, Torino, Italy
| | - Anna Tylki-Szymańska
- Department of Paediatrics, Nutrition and Metabolic Diseases, Children's Memorial Health Institute, Warsaw, Poland
| | - Camilla Tøndel
- Department of Paediatrics, Haukeland University Hospital and Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Miguel Viana-Baptista
- Serviço de Neurologia, Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, CEDOC Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Frank Weidemann
- Department of Cardiology, Innere Klinik II, Katharinen-Hospital, Unna, Germany
| | - Max J Hilz
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
40
|
Hongo K, Ito K, Date T, Anan I, Inoue Y, Morimoto S, Ogawa K, Kawai M, Kobayashi H, Kobayashi M, Ida H, Ohashi T, Taniguchi I, Yoshimura M, Eto Y. The beneficial effects of long-term enzyme replacement therapy on cardiac involvement in Japanese Fabry patients. Mol Genet Metab 2018; 124:143-151. [PMID: 29747997 DOI: 10.1016/j.ymgme.2018.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 04/21/2018] [Indexed: 01/02/2023]
Abstract
Fabry disease is a hereditary disorder that occurs due to the reduction or absence of alpha-galactosidase A activity, which leads to cardiac involvement including left ventricular hypertrophy (LVH). Enzyme replacement therapy (ERT) provides better patient outcomes by preventing serious complications. However, there have been very few studies on the long-term effects of ERT on the cardiac manifestations in Japanese Fabry patients. We retrospectively analyzed the data from the medical records of 42 Fabry patients (male, n = 17; female, n = 25) who were followed at Jikei University Hospital, and in whom the long-term effects of ERT could be evaluated (median follow-up period: male, 11 years; female, 8 years). The slope of the left ventricular mass (LVM) increase was 3.02 ± 3.41 g/m2/year in males and 1.69 ± 2.73 g/m2/year in females. In a subgroup analysis, the slopes of males with and without LVH did not differ to a statistically significant extent; however, the slope in female patients without LVH was significantly smaller than that of female patients with LVH. We then compared our data to the natural historical data that have previously been reported. In comparison to the previously reported data, we found a significant reduction in the LVM changes (g/height2.7/year) of patients who received long-term ERT (male, 4.07 ± 1.03 to 1.25 ± 1.39; female, 2.31 ± 0.81 to 0.78 ± 1.23). Long-term ERT effectively prevents LVH in Fabry patients. This effect was also observed in the patients with LVH prior to the initiation of ERT.
Collapse
Affiliation(s)
- Kenichi Hongo
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan.
| | - Keiichi Ito
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Taro Date
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ikuko Anan
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yasunori Inoue
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Satoshi Morimoto
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroshi Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Masahisa Kobayashi
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Hiroyuki Ida
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Toya Ohashi
- Department of Pediatrics, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Ikuo Taniguchi
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yoshikatsu Eto
- Advanced Clinical Research Center, Institute of Neurological Disorders, Furusawa-Miyako 255, Asou-ku, Kawasaki city, Kanagawa Prefecture 215-0026, Japan
| |
Collapse
|
41
|
Lavalle L, Thomas AS, Beaton B, Ebrahim H, Reed M, Ramaswami U, Elliott P, Mehta AB, Hughes DA. Phenotype and biochemical heterogeneity in late onset Fabry disease defined by N215S mutation. PLoS One 2018; 13:e0193550. [PMID: 29621274 PMCID: PMC5886405 DOI: 10.1371/journal.pone.0193550] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/13/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Fabry disease (FD) results from X-linked inheritance of a mutation in the GLA gene, encoding for alpha galactosidase A, and is characterized by heterogeneous clinical manifestations. Two phenotypes have been described "Classic" and "late onset" which cannot be predicted exclusively by genotype. The latter has been considered an attenuated form of the disease often affecting a single organ system commonly the heart. Recent studies have demonstrated that cardiac outcomes are similar in patients with classic and late onset mutations. In this study we investigate the relationship between clinical heterogeneity and plasma lyso-Gb3 in a large single centre cohort of N215S patients and compare this to patients with other mutations. METHODS In this single-centre, retrospective, cross-sectional study we analysed a cohort of 251 FD patients: 84 N215S mutation (37 males) and 167 non-N215S mutations (58 males). The Mainz severity score index (MSSI) was used as an index of overall disease severity. Cardiac function and morphology were assessed by electrocardiogram and echocardiogram. Left ventricular mass was calculated using the Devereux formula and the left ventricular mass index (LVMI) calculated to adjust for height (g/m2.7). The presence of white matter lesions was assessed by cerebral MRI or computed tomography (CT). GFR was measured by radio-isotope (chromium-EDTA) method and adjusted for patient height (ml/min/m2.7), and urinary protein quantification was undertaken by 24 hour urine collection. Plasma globotriaosylsphingosine (lyso-Gb3) was analysed prior to ERT in 84 patients. RESULTS N215S patients showed later symptom onset (males: p< 0.0001, females: p<0.03), later development of left ventricular hypertrophy (LVH) (median survival without LVH: 41 (non-N215S) vs. 64 (N215S) years, p< 0.0001), later development of proteinuria (median survival without proteinuria 43 (non-N215S) vs 71 years (N215S), p< 0.0001), later occurrence of cerebrovascular events (stroke/ Transient Ischaemic Attacks (TIA); median survival without stroke: 74 years (non-N215S) vs. not reached (N215S), p< 0.02), later decline in renal function to GFR <60 ml/min/1.73m2 (median survival: 56 (non-N215S) vs. 72 (N215S) years, p< 0.01), and greater overall survival (median survival 81 (N215S) vs. 66 (non-N215S) years, p< 0.0006). Lyso-Gb3 was found to be less elevated in N215S compared to non-N215S male and female patients. However, the N215S population eventually reached an overall severity measured by MSSI comparable to the non-N215S without equivalent elevation of lyso-Gb3 (means: 6.7 vs. 74.3 nmol/L, p < 0.001). In addition, N215S patients showed strong correlations between lyso-Gb3 levels and LVMI, GFR, and MSSI. These associations became stronger when we investigated individuals' life time exposure to lyso-Gb3 (calculated as [lyso-Gb3]*age): MSSI (r2 = 0.88, p< 0.0001), LVMI (r2 = 0.59, p< 0.005), and GFR (r2 = 0.75, p = 0.0001). CONCLUSION These results demonstrate that the N215S mutation results in a late onset phenotype involving the heart and other organs. Correlations between clinical manifestations and plasma lyso-Gb3 variations in this group suggest a Fabry-relevant disease mechanism for the heterogeneity observed in this group.
Collapse
Affiliation(s)
- L. Lavalle
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - A. S. Thomas
- UCL Institute of Cardiovascular Science, Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
| | - B. Beaton
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - H. Ebrahim
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - M. Reed
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - U. Ramaswami
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - P. Elliott
- Haematology Department, St George’s Hospital NHS Foundation Trust, London, United Kingdom
| | - A. B. Mehta
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| | - D. A. Hughes
- Lysosomal Storage Disorders Unit, Department of Haematology, Royal Free Hospital and University College Medical School, London, United Kingdom
| |
Collapse
|
42
|
Adalsteinsdottir B, Palsson R, Desnick RJ, Gardarsdottir M, Teekakirikul P, Maron M, Appelbaum E, Neisius U, Maron BJ, Burke MA, Chen B, Pagant S, Madsen CV, Danielsen R, Arngrimsson R, Feldt-Rasmussen U, Seidman JG, Seidman CE, Gunnarsson GT. Fabry Disease in Families With Hypertrophic Cardiomyopathy: Clinical Manifestations in the Classic and Later-Onset Phenotypes. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.116.001639. [PMID: 28798024 DOI: 10.1161/circgenetics.116.001639] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 06/15/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND The screening of Icelandic patients clinically diagnosed with hypertrophic cardiomyopathy resulted in identification of 8 individuals from 2 families with X-linked Fabry disease (FD) caused by GLA(α-galactosidase A gene) mutations encoding p.D322E (family A) or p.I232T (family B). METHODS AND RESULTS Familial screening of at-risk relatives identified mutations in 16 family A members (8 men and 8 heterozygotes) and 25 family B members (10 men and 15 heterozygotes). Clinical assessments, α-galactosidase A (α-GalA) activities, glycosphingolipid substrate levels, and in vitro mutation expression were used to categorize p.D322E as a classic FD mutation and p.I232T as a later-onset FD mutation. In vitro expression revealed that p.D322E and p.I232T had α-GalA activities of 1.4% and 14.9% of the mean wild-type activity, respectively. Family A men had markedly decreased α-GalA activity and childhood-onset classic manifestations, except for angiokeratoma and cornea verticillata. Family B men had residual α-GalA activity and developed FD manifestations in adulthood. Despite these differences, all family A and family B men >30 years of age had left ventricular hypertrophy, which was mainly asymmetrical, and had similar late gadolinium enhancement patterns. Ischemic stroke and severe white matter lesions were more frequent among family A men, but neither family A nor family B men had overt renal disease. Family A and family B heterozygotes had less severe or no clinical manifestations. CONCLUSIONS Men with classic or later-onset FD caused by GLA missense mutations developed prominent and similar cardiovascular disease at similar ages, despite markedly different α-GalA activities.
Collapse
Affiliation(s)
- Berglind Adalsteinsdottir
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.).
| | - Runolfur Palsson
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Robert J Desnick
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Marianna Gardarsdottir
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Polakit Teekakirikul
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Martin Maron
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Evan Appelbaum
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Ulf Neisius
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Barry J Maron
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Michael A Burke
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Brenden Chen
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Silvere Pagant
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Christoffer V Madsen
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Ragnar Danielsen
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Reynir Arngrimsson
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Ulla Feldt-Rasmussen
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Jonathan G Seidman
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Christine E Seidman
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| | - Gunnar Th Gunnarsson
- From the Faculty of Medicine, University of Iceland, Reykjavik, Iceland (B.A., R.P., R.A., G.T.G.); Division of Cardiology (B.A., R.D.), Department of Genetics (R.A.), Division of Nephrology (R.P.), and Department of Radiology (M.G.), Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland; Department of Cardiology, Haukeland University Hospital, Bergen, Norway (B.A.); Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY (R.J.D., B.C., S.P.); Department of Genetics, Harvard Medical School, Boston, MA (P.T., M.A.B., J.G.S., C.E.S.); Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (E.A., U.N.); Division of Cardiology, Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA (M.M., B.J.M.); Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (M.A.B., C.E.S.); Division of Cardiology, Emory University School of Medicine, Atlanta, GA (M.A.B.); Department of Medical Endocrinology, Rigshospitalet and University of Copenhagen, Denmark (C.V.M., U.F.-R.); Howard Hughes Medical Institute, Boston, MA (C.E.S.); and Department of Medicine, Akureyri Hospital, Iceland (G.T.G.)
| |
Collapse
|
43
|
|
44
|
Limongelli G, Masarone D, Verrengia M, Gravino R, Salerno G, Castelletti S, Rubino M, Marrazzo T, Pisani A, Cecchi F, Elliott PM, Pacileo G. Diagnostic Clues for the Diagnosis of Nonsarcomeric Hypertrophic Cardiomyopathy (Phenocopies): Amyloidosis, Fabry Disease, and Mitochondrial Disease. J Cardiovasc Echogr 2018; 28:120-123. [PMID: 29911009 PMCID: PMC5989543 DOI: 10.4103/jcecho.jcecho_2_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common known inherited heart disorder, with a prevalence of 1:500 of the adult population. Etiology of HCM can be heterogeneous, with sarcomeric gene disease as the leading cause in up to 60% of the patients, and with a number of possible different diseases (phenocopies) in about 10%–15% of the patients. Early diagnosis of storage and infiltrative disorders, particularly those with specific treatments (i.e., Fabry disease and/or amyloidosis), means early management and treatment, with a significant impact on patients prognosis. Here, we report on four different cases of HCM, highlighting difficulties to make differential diagnosis of different forms of cardiomyopathies, and their potential impact on the management.
Collapse
Affiliation(s)
- Giuseppe Limongelli
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy.,Institute of Cardiovascular Sciences University College of London, London, UK
| | - Daniele Masarone
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Marina Verrengia
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Rita Gravino
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Gemma Salerno
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Silvia Castelletti
- IRCCS Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Marta Rubino
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Tommaso Marrazzo
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| | - Antonio Pisani
- Department of Public Health, Nefrology Unit, Federico II University, Naples, Italy
| | - Franco Cecchi
- IRCCS Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy
| | - Perry Mark Elliott
- Institute of Cardiovascular Sciences University College of London, London, UK
| | - Giuseppe Pacileo
- Department of Cardiothoracic Sciences, "Luigi Vanvitelli"-Campania University, Naples, Italy
| |
Collapse
|
45
|
Manna R, Cauda R, Feriozzi S, Gambaro G, Gasbarrini A, Lacombe D, Livneh A, Martini A, Ozdogan H, Pisani A, Riccio E, Verrecchia E, Dagna L. Recommendations for the inclusion of Fabry disease as a rare febrile condition in existing algorithms for fever of unknown origin. Intern Emerg Med 2017; 12:1059-1067. [PMID: 28726033 DOI: 10.1007/s11739-017-1704-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 06/27/2017] [Indexed: 12/26/2022]
Abstract
Fever of unknown origin (FUO) is a rather rare clinical syndrome representing a major diagnostic challenge. The occurrence of more than three febrile attacks with fever-free intervals of variable duration during 6 months of observation has recently been proposed as a subcategory of FUO, Recurrent FUO (RFUO). A substantial number of patients with RFUO have auto-inflammatory genetic fevers, but many patients remain undiagnosed. We hypothesize that this undiagnosed subgroup may be comprised of, at least in part, a number of rare genetic febrile diseases such as Fabry disease. We aimed to identify key features or potential diagnostic clues for Fabry disease as a model of rare genetic febrile diseases causing RFUO, and to develop diagnostic guidelines for RFUO, using Fabry disease as an example of inserting other rare diseases in the existing FUO algorithms. An international panel of specialists in recurrent fevers and rare diseases, including internists, infectious disease specialists, rheumatologists, gastroenterologists, nephrologists, and medical geneticists convened to review the existing diagnostic algorithms, and to suggest recommendations for arriving at accurate diagnoses on the basis of available literature and clinical experience. By combining specific features of rare diseases with other diagnostic considerations, guidelines have been designed to raise awareness and identify rare diseases among other causes of FUO. The proposed guidelines may be useful for the inclusion of rare diseases in the diagnostic algorithms for FUO. A wide spectrum of patients will be needed to validate the algorithm in different clinical settings.
Collapse
Affiliation(s)
- Raffaele Manna
- Centro di Ricerca delle Febbri Periodiche e Malattie Rare, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy.
| | - Roberto Cauda
- Istituto di Malattie Infettive, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Giovanni Gambaro
- Divisione di Nefrologia, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- Divisione di Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Didier Lacombe
- CHU Bordeaux, MRGM Laboratory, Department Medical Genetics, University of Bordeaux, Bordeaux, France
| | - Avi Livneh
- Department of Medicine, Sheba Medical Center, Tel Hashomer, Israel, and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | - Huri Ozdogan
- Division of Rheumatology, Department of Internal Medicine, Cerrahpasa Medical Faculty, Istanbul University Fatih, Istanbul, Turkey
| | | | | | - Elena Verrecchia
- Centro di Ricerca delle Febbri Periodiche e Malattie Rare, Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168, Rome, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
46
|
Olivera-González S, Josa-Laorden C, Torralba-Cabeza MA. The pathophysiology of Fabry disease. Rev Clin Esp 2017; 218:22-28. [PMID: 28843599 DOI: 10.1016/j.rce.2017.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/12/2017] [Accepted: 06/12/2017] [Indexed: 10/19/2022]
Abstract
Fabry disease is a lysosomal condition with systemic clinical expression, caused by the tissue deposit of globotriaosylceramide, due to a deficit in its degradation. As with most lysosomal diseases, the presence of a mutation in a gene does not explain the pathophysiological disorders shown by patients. We conducted a comprehensive review of the pathogenic mechanisms that occur in Fabry disease.
Collapse
Affiliation(s)
- S Olivera-González
- Unidad de Enfermedades Minoritarias, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España.
| | - C Josa-Laorden
- Servicio de Medicina Interna, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| | - M A Torralba-Cabeza
- Unidad de Enfermedades Minoritarias, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España
| |
Collapse
|
47
|
Affiliation(s)
- Ryuichiro Anan
- Clinical Research Unit, National Miyakonojo Medical Center
| |
Collapse
|
48
|
Chang WH, Niu DM, Lu CY, Lin SY, Liu TC, Chang JG. Modulation the alternative splicing of GLA (IVS4+919G>A) in Fabry disease. PLoS One 2017; 12:e0175929. [PMID: 28430823 PMCID: PMC5400244 DOI: 10.1371/journal.pone.0175929] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/03/2017] [Indexed: 12/31/2022] Open
Abstract
While a base substitution in intron 4 of GLA (IVS4+919G>A) that causes aberrant alternative splicing resulting in Fabry disease has been reported, its molecular mechanism remains unclear. Here we reported that upon IVS4+919G>A transversion, H3K36me3 was enriched across the alternatively spliced region. PSIP1, an adapter of H3K36me3, together with Hsp70 and NONO were recruited and formed a complex with SF2/ASF and SRp20, which further promoted GLA splicing. Amiloride, a splicing regulator in cancer cells, could reverse aberrant histone modification patterns and disrupt the association of splicing complex with GLA. It could also reverse aberrant GLA splicing in a PP1-dependant manner. Our findings revealed the alternative splicing mechanism of GLA (IVS4+919G>A), and a potential treatment for this specific genetic type of Fabry disease by amiloride in the future.
Collapse
Affiliation(s)
- Wen-Hsin Chang
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Dau-Ming Niu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- * E-mail: (SYL); (TCL); (JGC)
| | - Ta-Chih Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (SYL); (TCL); (JGC)
| | - Jan-Gowth Chang
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- * E-mail: (SYL); (TCL); (JGC)
| |
Collapse
|
49
|
Chou SJ, Yu WC, Chang YL, Chen WY, Chang WC, Chien Y, Yen JC, Liu YY, Chen SJ, Wang CY, Chen YH, Niu DM, Lin SJ, Chen JW, Chiou SH, Leu HB. Energy utilization of induced pluripotent stem cell-derived cardiomyocyte in Fabry disease. Int J Cardiol 2017; 232:255-263. [DOI: 10.1016/j.ijcard.2017.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 12/29/2022]
|
50
|
Fulton N, Rajiah P. Utility of magnetic resonance imaging in the evaluation of left ventricular thickening. Insights Imaging 2017; 8:279-293. [PMID: 28281159 PMCID: PMC5359150 DOI: 10.1007/s13244-017-0549-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 12/14/2022] Open
Abstract
Left ventricular (LV) thickening can be due to hypertrophy (concentric, asymmetric, eccentric) or remodelling (concentric or asymmetric). Pathological thickening may be caused by pressure overload, volume overload, infiltrative disorders, hypertrophic cardiomyopathy, athlete's heart or neoplastic infiltration. Magnetic resonance imaging (MRI) plays an important role in the comprehensive evaluation of LV thickening, including: establishing diagnosis, determining LV geometry, establishing aetiology, quantification, identifying prognostic factors, serial follow-up and treatment response. In this article, we review the aetiologies and pathophysiology of LV thickening, and demonstrate the comprehensive role of MRI in the evaluation of LV thickening. TEACHING POINTS • MRI plays an important role in the evaluation of LV thickening. • LV thickening can be due to either hypertrophy or remodelling. • Pathological thickening can be due to pressure/volume overload or infiltrative disorders.
Collapse
Affiliation(s)
- Nicholas Fulton
- Department of Radiology, University Hospital Case Medical Center, Cleveland, OH, USA
| | - Prabhakar Rajiah
- Department of Radiology, University Hospital Case Medical Center, Cleveland, OH, USA.
- Department of Radiology Cardiothoracic Imaging, UT Southwestern Medical Center, E6.120 B, Mail code 9316, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8896, USA.
| |
Collapse
|