1
|
Huang S, Abutaleb K, Mishra S. Glycosphingolipids in Cardiovascular Disease: Insights from Molecular Mechanisms and Heart Failure Models. Biomolecules 2024; 14:1265. [PMID: 39456198 PMCID: PMC11506000 DOI: 10.3390/biom14101265] [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: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
This review explores the crucial role of glycosphingolipids (GSLs) in the context of cardiovascular diseases (CVDs), focusing on their biosynthesis, metabolic pathways, and implications for clinical outcomes. GSLs are pivotal in regulating a myriad of cellular functions that are essential for heart health and disease progression. Highlighting findings from both human cohorts and animal models, this review emphasizes the potential of GSLs as biomarkers and therapeutic targets. We advocate for more detailed mechanistic studies to deepen our understanding of GSL functions in cardiovascular health, which could lead to innovative strategies for diagnosis, treatment, and personalized medicine in cardiovascular care.
Collapse
Affiliation(s)
- Sarah Huang
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Karima Abutaleb
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24061, USA
| | - Sumita Mishra
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24061, USA
- Center for Exercise Medicine Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24061, USA
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24061, USA
- Department of Human Nutrition, Foods, and Exercise, College of Life Sciences, Virginia Tech, Roanoke, VA 24061, USA
| |
Collapse
|
2
|
Gatterer C, Beitzke D, Sunder-Plassmann G, Friedl M, Hohensinner P, Mann C, Ponleitner M, Graf S, Lenz M. NT-proBNP Reflects Left Ventricular Hypertrophy Rather than Left Ventricular Dilatation or Systolic Dysfunction in Patients with Fabry Disease. J Clin Med 2024; 13:5953. [PMID: 39408013 PMCID: PMC11478308 DOI: 10.3390/jcm13195953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The diagnosis and follow-up of cardiac involvement in Fabry disease constitutes an important challenge for clinicians caring for affected patients. Combining cardiac imaging with laboratory biomarkers appears most appropriate for longitudinal monitoring. Therefore, we examined the use of NT-proBNP and its association with imaging findings in patients with Fabry disease. Methods: We analysed cardiac MRI and echocardiography data, as well as laboratory results, from a single-centre prospective registry. Results: Repetitive follow-ups of 38 patients with Fabry disease, of whom 18 presented with left ventricular hypertrophy (LVH), revealed a correlation of NT-proBNP with left ventricular (LV) interventricular septal thickness, LV maximum wall thickness, LV and right ventricular (RV) mass index and trabecular mass in patients with LVH. Patients without LVH did not exhibit any tangible association between NT-proBNP and the mentioned parameters. Conversely, we could not detect an association of NT-proBNP with impairment of LV or RV ejection fraction or diastolic volume. Conclusions: NT-proBNP plays a pivotal role as a biomarker for cardiac involvement in patients with Fabry disease. Interestingly, in this specific population with mostly preserved ejection fraction, it seems to reflect ventricular hypertrophy rather than ventricular dysfunction or dilatation. While strong associations were found in hypertrophic patients, NT-proBNP's prognostic value appears limited in non- or pre-hypertrophic stages.
Collapse
Affiliation(s)
- Constantin Gatterer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Gere Sunder-Plassmann
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Maximilian Friedl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| | - Christopher Mann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| | - Markus Ponleitner
- Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Senta Graf
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| | - Max Lenz
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (S.G.)
| |
Collapse
|
3
|
Hung CL, Wu YW, Kuo L, Sung KT, Lin HH, Chang WT, Chang CH, Lai CH, Huang CY, Wang CL, Lin CC, Juang JMJ, Chen PS, Wang CY, Chang HC, Chu CY, Wang WH, Tseng H, Kao YT, Wang TD, Yu WC, Chen WJ. 2024 Update of the TSOC Expert Consensus of Fabry Disease. ACTA CARDIOLOGICA SINICA 2024; 40:544-568. [PMID: 39308653 PMCID: PMC11413953 DOI: 10.6515/acs.202409_40(5).20240731a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024]
Abstract
As an X-linked inherited lysosomal storage disease that is caused by α-galactosidase A gene variants resulting in progressive accumulation of pathogenic glycosphingolipid (Gb3) accumulation in multiple tissues and organs, Fabry disease (FD) can be classified into classic or late-onset phenotypes. In classic phenotype patients, α-galactosidase A activity is absent or severely reduced, resulting in a more progressive disease course with multi-systemic involvement. Conversely, late-onset phenotype, often with missense variants (e.g., IVS4+919G>A) in Taiwan, may present with a more chronic clinical course with predominant cardiac involvement (cardiac subtype), as they tend to have residual enzyme activity, remaining asymptomatic or clinically silent during childhood and adolescence. In either form, cardiac hypertrophy remains the most common feature of cardiac involvement, potentially leading to myocardial fibrosis, arrhythmias, and heart failure. Diagnosis is established through α-galactosidase enzyme activity assessment or biomarker analyisis (globotriaosylsphingosine, Lyso-Gb3), advanced imaging modalities (echocardiography and cardiac magnetic resonance imaging), and genotyping to differentiate FD from other cardiomyopathy. Successful therapeutic response relies on early recognition and by disease awareness from typical features in classic phenotype and cardiac red flags in cardiac variants for timely therapeutic interventions. Recent advances in pharmacological approach including enzyme replacement therapy (agalsidase alfa or beta), oral chaperone therapy (migalastat), and substrate reduction therapy (venglustat) aim to prevent from irreversible organ damage. Genotype- and gender-based monitoring of treatment effects through biomarker (Lyso-Gb3), renal assessment, and cardiac responses using advanced imaging modalities are key steps to optimizing patient care in FD.
Collapse
Affiliation(s)
- Chung-Lieh Hung
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei
| | - Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, and Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan
| | - Ling Kuo
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei
| | - Kuo-Tzu Sung
- Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei
- Department of Medicine, MacKay Medical College
| | - Heng-Hsu Lin
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City
| | - Wei-Ting Chang
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan
| | - Chia-Hsiu Chang
- Division of Cardiology, Cardiovascular Center, Cathay General Hospital, Taipei
| | - Chih-Hung Lai
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University
| | - Chun-Yao Huang
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital
- Taipei Heart institute, Taipei Medical University, Taipei
| | - Chun-Li Wang
- Division of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan
| | - Chih-Chan Lin
- Division of Cardiology, Department of Internal Medicine, An Nan Hospital, China Medical University, Tainan
| | - Jyh-Ming Jimmy Juang
- Heart Failure Center and Division of Cardiology, Department of Internal Medicine, Center of Genetic Heart Diseases, National Taiwan University Hospital
- Department of Medicine, National Taiwan University College of Medicine, Taipei
| | - Po-Sheng Chen
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan
| | - Chao-Yung Wang
- Division of Cardiology, Chang Gung Memorial Hospital, Linkou Medical Center
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli
- Department of Medical Science, National Tsing Hua University, Hsinchu
| | - Hao-Chih Chang
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
- Department of Medicine, Taipei Veterans General Hospital Taoyuan Branch, Taoyuan
| | - Chun-Yuan Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
| | - Wen-Hwa Wang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung
| | - Hsinyu Tseng
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu
| | - Yung-Ta Kao
- Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital
- Taipei Heart institute, Taipei Medical University, Taipei
| | - Tzung-Dau Wang
- Department of Medicine, National Taiwan University College of Medicine, Taipei
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan
| | - Wen-Chung Yu
- Department of Internal Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Jone Chen
- Division of Cardiology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan
| |
Collapse
|
4
|
Yuan Y, Zhao Y, Li F, Ling C, Wu Y, Ma W, Wang Z, Yuan Y, Hao H, Zhang W. Inflammatory cytokine expression in Fabry disease: impact of disease phenotype and alterations under enzyme replacement therapy. Front Immunol 2024; 15:1367252. [PMID: 39234251 PMCID: PMC11371600 DOI: 10.3389/fimmu.2024.1367252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Objectives The aim of this study is to explore the expression of inflammatory cytokines (ICs) in Fabry disease (FD), the correlation between ICs and FD phenotypes, and the impact of enzyme replacement therapy (ERT) on IC expression. Methods We recruited 67 FD patients and 44 healthy controls (HCs) and detected concentrations of the following ICs: interferon-γ, interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12P70, IL-17A, IL-17F, IL-22, tumor necrosis factor (TNF)-α, and TNF-β. We also analyzed the impact of ERT on IC expression in FD patients and the relationship between IC expression and sex, genotype, phenotype, disease burden, and biomarkers. Results Most ICs were significantly higher in FD patients than in HCs. A number of ICs were positively correlated with clinical aspects, including disease burden (Mainz Severity Score Index [MSSI]) and cardiac and renal markers. IL-8 was higher in the high MSSI (P-adj=0.026*) than in the low MSSI. Conclusions ICs were upregulated in FD patients, indicating the role of the innate immune process in FD etiology. ERT ameliorated FD-related inflammatory activation, at least to some extent. IC expression was positively correlated with disease burden and clinical markers in FD. Our findings indicated that the inflammatory pathway may be a promising therapeutic target for FD.
Collapse
Affiliation(s)
- Yujing Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yawen Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Fan Li
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chen Ling
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yuan Wu
- Department of Ophthalmology, Peking University First Hospital, Beijing, China
| | - Wei Ma
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Diseases, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Diseases, Beijing, China
| | - Hongjun Hao
- Department of Neurology, Peking University First Hospital, Beijing, China
- Department of Neuroimmunity, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Neurovascular Diseases, Beijing, China
| |
Collapse
|
5
|
Faro DC, Di Pino FL, Monte IP. Inflammation, Oxidative Stress, and Endothelial Dysfunction in the Pathogenesis of Vascular Damage: Unraveling Novel Cardiovascular Risk Factors in Fabry Disease. Int J Mol Sci 2024; 25:8273. [PMID: 39125842 PMCID: PMC11312754 DOI: 10.3390/ijms25158273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Anderson-Fabry disease (AFD), a genetic disorder caused by mutations in the α-galactosidase-A (GLA) gene, disrupts lysosomal function, leading to vascular complications. The accumulation of globotriaosylceramide (Gb3) in arterial walls triggers upregulation of adhesion molecules, decreases endothelial nitric oxide synthesis, and induces reactive oxygen species production. This cascade results in fibrotic thickening, endothelial dysfunction, hypercontractility, vasospasm, and a pro-thrombotic phenotype. AFD patients display increased intima-media thickness (IMT) and reduced flow-mediated dilation (FMD), indicating heightened cardiovascular risk. Nailfold capillaroscopy (NFC) shows promise in diagnosing and monitoring microcirculatory disorders in AFD, though it remains underexplored. Morphological evidence of AFD as a storage disorder can be demonstrated through electron microscopy and immunodetection of Gb3. Secondary pathophysiological disturbances at cellular, tissue, and organ levels contribute to the clinical manifestations, with prominent lysosomal inclusions observed in vascular, cardiac, renal, and neuronal cells. Chronic accumulation of Gb3 represents a state of ongoing toxicity, leading to increased cell turnover, particularly in vascular endothelial cells. AFD-related vascular pathology includes increased renin-angiotensin system activation, endothelial dysfunction, and smooth muscle cell proliferation, resulting in IMT increase. Furthermore, microvascular alterations, such as atypical capillaries observed through NFC, suggest early microvascular involvement. This review aims to unravel the complex interplay between inflammation, oxidative stress, and endothelial dysfunction in AFD, highlighting the potential connections between metabolic disturbances, oxidative stress, inflammation, and fibrosis in vascular and cardiac complications. By exploring novel cardiovascular risk factors and potential diagnostic tools, we can advance our understanding of these mechanisms, which extend beyond sphingolipid accumulation to include other significant contributors to disease pathogenesis. This comprehensive approach can pave the way for innovative therapeutic strategies and improved patient outcomes.
Collapse
Affiliation(s)
| | | | - Ines Paola Monte
- Department of General Surgery and Medical-Surgical Specialties (CHIRMED), University of Catania, Via S. Sofia 78, 95100 Catania, Italy; (D.C.F.); (F.L.D.P.)
| |
Collapse
|
6
|
López-Valverde L, Vázquez-Mosquera ME, Colón-Mejeras C, Bravo SB, Barbosa-Gouveia S, Álvarez JV, Sánchez-Martínez R, López-Mendoza M, López-Rodríguez M, Villacorta-Argüelles E, Goicoechea-Diezhandino MA, Guerrero-Márquez FJ, Ortolano S, Leao-Teles E, Hermida-Ameijeiras Á, Couce ML. Characterization of the plasma proteomic profile of Fabry disease: Potential sex- and clinical phenotype-specific biomarkers. Transl Res 2024; 269:47-63. [PMID: 38395389 DOI: 10.1016/j.trsl.2024.02.006] [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: 11/23/2023] [Revised: 01/25/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Fabry disease (FD) is a X-linked rare lysosomal storage disorder caused by deficient α-galactosidase A (α-GalA) activity. Early diagnosis and the prediction of disease course are complicated by the clinical heterogeneity of FD, as well as by the frequently inconclusive biochemical and genetic test results that do not correlate with clinical course. We sought to identify potential biomarkers of FD to better understand the underlying pathophysiology and clinical phenotypes. We compared the plasma proteomes of 50 FD patients and 50 matched healthy controls using DDA and SWATH-MS. The >30 proteins that were differentially expressed between the 2 groups included proteins implicated in processes such as inflammation, heme and haemoglobin metabolism, oxidative stress, coagulation, complement cascade, glucose and lipid metabolism, and glycocalyx formation. Stratification by sex revealed that certain proteins were differentially expressed in a sex-dependent manner. Apolipoprotein A-IV was upregulated in FD patients with complications, especially those with chronic kidney disease, and apolipoprotein C-III and fetuin-A were identified as possible markers of FD with left ventricular hypertrophy. All these proteins had a greater capacity to identify the presence of complications in FD patients than lyso-GB3, with apolipoprotein A-IV standing out as being more sensitive and effective in differentiating the presence and absence of chronic kidney disease in FD patients than renal markers such as creatinine, glomerular filtration rate and microalbuminuria. Identification of these potential biomarkers can help further our understanding of the pathophysiological processes that underlie the heterogeneous clinical manifestations associated with FD.
Collapse
Affiliation(s)
- Laura López-Valverde
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - María E Vázquez-Mosquera
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Cristóbal Colón-Mejeras
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Susana B Bravo
- Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Proteomic Platform, University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Sofía Barbosa-Gouveia
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - J Víctor Álvarez
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Rosario Sánchez-Martínez
- Internal Medicine Department, Alicante General University Hospital-Alicante Institute of Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante 03010, Spain
| | - Manuel López-Mendoza
- Department of Nephrology, Hospital Universitario Virgen del Rocío, Manuel Siurot s/n, Sevilla 41013, Spain
| | - Mónica López-Rodríguez
- Internal Medicine Department, Hospital Universitario Ramón y Cajal, IRYCIS, Colmenar Viejo, Madrid 28034, Spain; Faculty of Medicine and Health Sciences, Universidad de Alcalá (UAH), Av. de Madrid, Alcalá de Henares 28871, Spain
| | - Eduardo Villacorta-Argüelles
- Department of Cardiology, Complejo Asistencial Universitario de Salamanca, P°. de San Vicente 58, Salamanca 37007, Spain
| | | | - Francisco J Guerrero-Márquez
- Department of Cardiology, Internal Medicine Service, Hospital de la Serranía, San Pedro, Ronda, Málaga 29400, Spain
| | - Saida Ortolano
- Rare Diseases and Pediatric Medicine Research Group, Galicia Sur Health Research Institute-SERGAS-UVIGO, Clara Campoamor 341, Vigo 36213, Spain
| | - Elisa Leao-Teles
- Centro de Referência de Doenças Hereditárias do Metabolismo, Centro Hospitalar Universitário de São João, Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Álvaro Hermida-Ameijeiras
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain.
| | - María L Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain.
| |
Collapse
|
7
|
Mauhin W, Dzangue-Tchoupou G, Amelin D, Corneau A, Lamari F, Allenbach Y, Dussol B, Leguy-Seguin V, D'Halluin P, Matignon M, Maillot F, Ly KH, Besson G, Willems M, Labombarda F, Masseau A, Lavigne C, Lacombe D, Maillard H, Lidove O, Benveniste O. Mass cytometry reveals atypical immune profile notably impaired maturation of memory CD4 T with Gb3-related CD27 expression in CD4 T cells in Fabry disease. J Inherit Metab Dis 2024; 47:818-833. [PMID: 38623626 DOI: 10.1002/jimd.12727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 04/17/2024]
Abstract
Fabry disease (FD) is an X-linked disease characterized by an accumulation of glycosphingolipids, notably of globotriaosylceramide (Gb3) and globotriaosylsphingosine (lysoGb3) leading to renal failure, cardiomyopathy, and cerebral strokes. Inflammatory processes are involved in the pathophysiology. We investigated the immunological phenotype of peripheral blood mononuclear cells in Fabry patients depending on the clinical phenotype, treatment, Gb3, and lysoGb3 levels and the presence of anti-drug antibodies (ADA). Leucocytes from 41 male patients and 20 controls were analyzed with mass cytometry using both unsupervised and supervised algorithms. FD patients had an increased expression of CD27 and CD28 in memory CD45- and CD45 + CCR7-CD4 T cells (respectively p < 0.014 and p < 0.02). Percentage of CD45RA-CCR7-CD27 + CD28+ cells in CD4 T cells was correlated with plasma lysoGb3 (r = 0.60; p = 0.0036) and phenotype (p < 0.003). The correlation between Gb3 and CD27 in CD4 T cells almost reached significance (r = 0.33; p = 0.058). There was no immune profile associated with the presence of ADA. Treatment with agalsidase beta was associated with an increased proportion of Natural Killer cells. These findings provide valuable insights for understanding FD, linking Gb3 accumulation to inflammation, and proposing new prognostic biomarkers.
Collapse
Affiliation(s)
- Wladimir Mauhin
- Internal Medicine Department, Reference Center for Lysosomal Diseases, Groupe Hospitalier Diaconesses-Croix Saint Simon, Paris, France
- Centre de Recherche en Myologie, Unité Mixte de Recherche Scientifique 974, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Gaelle Dzangue-Tchoupou
- Centre de Recherche en Myologie, Unité Mixte de Recherche Scientifique 974, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Damien Amelin
- Centre de Recherche en Myologie, Unité Mixte de Recherche Scientifique 974, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Aurélien Corneau
- Plateforme de Cytométrie de la Pitié-Salpétrière (CyPS), UMS037-PASS, Faculté de Médecine, Sorbonne Université, Paris, France
| | - Foudil Lamari
- UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Yves Allenbach
- Centre de Recherche en Myologie, Unité Mixte de Recherche Scientifique 974, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Bertrand Dussol
- Nephrology Department, Aix Marseille Université et Centre d'Investigation Clinique 1409, INSERM/AMU/AP-HM, Marseille, France
| | - Vanessa Leguy-Seguin
- Internal Medicine and Clinical Immunology Department, Francois Mitterrand Hospital, Dijon, France
| | - Pauline D'Halluin
- Nephrology and Hemodialysis Department, Centre Hospitalier Côte Basque, Bayonne, France
| | - Marie Matignon
- Nephrology and Renal Transplantation Department, Institut Francilien de Recherche en Néphrologie et Transplantation (IFRNT), Henri-Mondor/Albert-Chenevier University Hospital, Assistance Publique Hôpitaux de Paris, Créteil, France
| | - François Maillot
- Internal Medicine Department, Tours University Hospital, Tours, France
| | - Kim-Heang Ly
- Internal Medicine Department, Dupuytren University Hospital, Limoges, France
| | - Gérard Besson
- Neurology Department, Grenoble University Hospital, Grenoble, France
| | - Marjolaine Willems
- Medical Genetics and Rare Diseases Department, Montpellier University Hospital, Montpellier, France
| | | | - Agathe Masseau
- Internal Medicine Department, Hôtel-Dieu University Hospital, Nantes, France
| | - Christian Lavigne
- Internal Medicine and Clinical Immunology Department, Angers University Hospital, Angers, France
| | - Didier Lacombe
- Medical Genetics Department, CHU de Bordeaux, INSERM U1211, Université de Bordeaux, Bordeaux, France
| | - Hélène Maillard
- Department of Internal Medicine and Clinical Immunology, Referral Centre for rare systemic autoimmune diseases North and North-West of France (CeRAINO), CHU Lille, Lille, France
| | - Olivier Lidove
- Internal Medicine Department, Reference Center for Lysosomal Diseases, Groupe Hospitalier Diaconesses-Croix Saint Simon, Paris, France
| | - Olivier Benveniste
- Centre de Recherche en Myologie, Unité Mixte de Recherche Scientifique 974, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
| |
Collapse
|
8
|
Kurdi H, Lavalle L, Moon JCC, Hughes D. Inflammation in Fabry disease: stages, molecular pathways, and therapeutic implications. Front Cardiovasc Med 2024; 11:1420067. [PMID: 38932991 PMCID: PMC11199868 DOI: 10.3389/fcvm.2024.1420067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Fabry disease, a multisystem X-linked disorder caused by mutations in the alpha-galactosidase gene. This leads to the accumulation of globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), culminating in various clinical signs and symptoms that significantly impact quality of life. Although treatments such as enzyme replacement, oral chaperone, and emerging therapies like gene therapy exist; delayed diagnosis often curtails their effectiveness. Our review highlights the importance of delineating the stages of inflammation in Fabry disease to enhance the timing and efficacy of diagnosis and interventions, particularly before the progression to fibrosis, where treatment options are less effective. Inflammation is emerging as an important aspect of the pathogenesis of Fabry disease. This is thought to be predominantly mediated by the innate immune response, with growing evidence pointing towards the potential involvement of adaptive immune mechanisms that remain poorly understood. Highlighted by the fact that Fabry disease shares immune profiles with systemic autoinflammatory diseases, blurring the distinctions between these disorders and highlighting the need for a nuanced understanding of immune dynamics. This insight is crucial for developing targeted therapies and improving the administration of current treatments like enzyme replacement. Moreover, our review discusses the complex interplay between these inflammatory processes and current treatments, such as the challenges posed by anti-drug antibodies. These antibodies can attenuate the effectiveness of therapies, necessitating more refined approaches to mitigate their impact. By advancing our understanding of the molecular changes, inflammatory mediators and causative factors that drive inflammation in Fabry disease, we aim to clarify their role in the disease's progression. This improved understanding will help us see how these processes fit into the current landscape of Fabry disease. Additionally, it will guide the development of more effective diagnostic and therapeutic approaches, ultimately improving patient care.
Collapse
Affiliation(s)
- Hibba Kurdi
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Lucia Lavalle
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| | - James C. C. Moon
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Derralynn Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| |
Collapse
|
9
|
Roy A, Cumberland MJ, O'Shea C, Holmes A, Kalla M, Gehmlich K, Geberhiwot T, Steeds RP. Arrhythmogenesis in Fabry Disease. Curr Cardiol Rep 2024; 26:545-560. [PMID: 38607539 PMCID: PMC11199244 DOI: 10.1007/s11886-024-02053-2] [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: 03/25/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW Fabry Disease (FD) is a rare lysosomal storage disorder characterised by multiorgan accumulation of glycosphingolipid due to deficiency in the enzyme α-galactosidase A. Cardiac sphingolipid accumulation triggers various types of arrhythmias, predominantly ventricular arrhythmia, bradyarrhythmia, and atrial fibrillation. Arrhythmia is likely the primary contributor to FD mortality with sudden cardiac death, the most frequent cardiac mode of death. Traditionally FD was seen as a storage cardiomyopathy triggering left ventricular hypertrophy, diastolic dysfunction, and ultimately, systolic dysfunction in advanced disease. The purpose of this review is to outline the current evidence exploring novel mechanisms underlying the arrhythmia substrate. RECENT FINDINGS There is growing evidence that FD cardiomyopathy is a primary arrhythmic disease with each stage of cardiomyopathy (accumulation, hypertrophy, inflammation, and fibrosis) contributing to the arrhythmia substrate via various intracellular, extracellular, and environmental mechanisms. It is therefore important to understand how these mechanisms contribute to an individual's risk of arrhythmia in FD. In this review, we outline the epidemiology of arrhythmia, pathophysiology of arrhythmogenesis, risk stratification, and cardiac therapy in FD. We explore how advances in conventional cardiac investigations performed in FD patients including 12-lead electrocardiography, transthoracic echocardiography, and cardiac magnetic resonance imaging have enabled early detection of pro-arrhythmic substrate. This has allowed for appropriate risk stratification of FD patients. This paves the way for future work exploring the development of therapeutic initiatives and risk prediction models to reduce the burden of arrhythmia.
Collapse
Affiliation(s)
- Ashwin Roy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK.
| | - Max J Cumberland
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Holmes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Manish Kalla
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Division of Cardiovascular Medicine, Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Tarekegn Geberhiwot
- Department of Inherited Metabolic Diseases, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, UK
| | - Richard P Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
- Department of Cardiology, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| |
Collapse
|
10
|
Pieroni M, Namdar M, Olivotto I, Desnick RJ. Anderson-Fabry disease management: role of the cardiologist. Eur Heart J 2024; 45:1395-1409. [PMID: 38486361 DOI: 10.1093/eurheartj/ehae148] [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: 09/03/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 04/22/2024] Open
Abstract
Anderson-Fabry disease (AFD) is a lysosomal storage disorder characterized by glycolipid accumulation in cardiac cells, associated with a peculiar form of hypertrophic cardiomyopathy (HCM). Up to 1% of patients with a diagnosis of HCM indeed have AFD. With the availability of targeted therapies for sarcomeric HCM and its genocopies, a timely differential diagnosis is essential. Specifically, the therapeutic landscape for AFD is rapidly evolving and offers increasingly effective, disease-modifying treatment options. However, diagnosing AFD may be difficult, particularly in the non-classic phenotype with prominent or isolated cardiac involvement and no systemic red flags. For many AFD patients, the clinical journey from initial clinical manifestations to diagnosis and appropriate treatment remains challenging, due to late recognition or utter neglect. Consequently, late initiation of treatment results in an exacerbation of cardiac involvement, representing the main cause of morbidity and mortality, irrespective of gender. Optimal management of AFD patients requires a dedicated multidisciplinary team, in which the cardiologist plays a decisive role, ranging from the differential diagnosis to the prevention of complications and the evaluation of timing for disease-specific therapies. The present review aims to redefine the role of cardiologists across the main decision nodes in contemporary AFD clinical care and drug discovery.
Collapse
Affiliation(s)
- Maurizio Pieroni
- Cardiovascular Department, San Donato Hospital, Via Pietro Nenni 22, 52100 Arezzo, Italy
| | - Mehdi Namdar
- Cardiology Division, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi Hospital and Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Robert J Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Fekete N, Li LK, Kozma GT, Fekete G, Pállinger É, Kovács ÁF. Flow Cytometry-Based Assay to Detect Alpha Galactosidase Enzymatic Activity at the Cellular Level. Cells 2024; 13:706. [PMID: 38667321 PMCID: PMC11049294 DOI: 10.3390/cells13080706] [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: 02/15/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Fabry disease is a progressive, X chromosome-linked lysosomal storage disorder with multiple organ dysfunction. Due to the absence or reduced activity of alpha-galactosidase A (AGAL), glycosphingolipids, primarily globotriaosyl-ceramide (Gb3), concentrate in cells. In heterozygous women, symptomatology is heterogenous and currently routinely used fluorometry-based assays measuring mean activity mostly fail to uncover AGAL dysfunction. The aim was the development of a flow cytometry assay to measure AGAL activity in individual cells. METHODS Conventional and multispectral imaging flow cytometry was used to detect AGAL activity. Specificity was validated using the GLA knockout (KO) Jurkat cell line and AGAL inhibitor 1-deoxygalactonojirimycin. The GLA KO cell line was generated via CRISPR-Cas9-based transfection, validated with exome sequencing, gene expression and substrate accumulation. RESULTS Flow cytometric detection of specific AGAL activity is feasible with fluorescently labelled Gb3. In the case of Jurkat cells, a substrate concentration of 2.83 nmol/mL and 6 h of incubation are required. Quenching of the aspecific exofacial binding of Gb3 with 20% trypan blue solution is necessary for the specific detection of lysosomal substrate accumulation. CONCLUSION A flow cytometry-based assay was developed for the quantitative detection of AGAL activity at the single-cell level, which may contribute to the diagnosis of Fabry patients.
Collapse
Affiliation(s)
- Nóra Fekete
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary; (N.F.); (É.P.)
- For Human Genome Foundation, 1094 Budapest, Hungary
| | - Luca Kamilla Li
- Pediatrics Centre, Tűzoltó Street Department, Semmelweis University, 1085 Budapest, Hungary; (L.K.L.); (G.F.)
| | - Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary;
- SeroScience LCC, 1089 Budapest, Hungary
| | - György Fekete
- Pediatrics Centre, Tűzoltó Street Department, Semmelweis University, 1085 Budapest, Hungary; (L.K.L.); (G.F.)
| | - Éva Pállinger
- Department of Genetics, Cell and Immunobiology, Semmelweis University, 1085 Budapest, Hungary; (N.F.); (É.P.)
| | - Árpád Ferenc Kovács
- For Human Genome Foundation, 1094 Budapest, Hungary
- Pediatrics Centre, Tűzoltó Street Department, Semmelweis University, 1085 Budapest, Hungary; (L.K.L.); (G.F.)
| |
Collapse
|
12
|
Coelho-Ribeiro B, Silva HG, Sampaio-Marques B, Fraga AG, Azevedo O, Pedrosa J, Ludovico P. Inflammation and Exosomes in Fabry Disease Pathogenesis. Cells 2024; 13:654. [PMID: 38667269 PMCID: PMC11049543 DOI: 10.3390/cells13080654] [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: 02/22/2024] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Fabry Disease (FD) is one of the most prevalent lysosomal storage disorders, resulting from mutations in the GLA gene located on the X chromosome. This genetic mutation triggers glo-botriaosylceramide (Gb-3) buildup within lysosomes, ultimately impairing cellular functions. Given the role of lysosomes in immune cell physiology, FD has been suggested to have a profound impact on immunological responses. During the past years, research has been focusing on this topic, and pooled evidence strengthens the hypothesis that Gb-3 accumulation potentiates the production of pro-inflammatory mediators, revealing the existence of an acute inflammatory process in FD that possibly develops to a chronic state due to stimulus persistency. In parallel, extracellular vesicles (EVs) have gained attention due to their function as intercellular communicators. Considering EVs' capacity to convey cargo from parent to distant cells, they emerge as potential inflammatory intermediaries capable of transporting cytokines and other immunomodulatory molecules. In this review, we revisit the evidence underlying the association between FD and altered immune responses and explore the potential of EVs to function as inflammatory vehicles.
Collapse
Affiliation(s)
- Bruna Coelho-Ribeiro
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Helena G. Silva
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Alexandra G. Fraga
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Olga Azevedo
- Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, 4835-044 Guimarães, Portugal;
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| |
Collapse
|
13
|
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
|
14
|
Lenzini L, Iori E, Vettore M, Gugelmo G, Radu C, Padoan A, Carraro G, Simioni P, Calò L, Avogaro A, Rossi GP, Vitturi N. Increased Soluble Interleukin 6 Receptors in Fabry Disease. J Clin Med 2023; 13:218. [PMID: 38202225 PMCID: PMC10780051 DOI: 10.3390/jcm13010218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Fabry disease (FD) is an X-linked lysosome storage disease that results in the accumulation of globotriaosylceramide (Gb3) throughout the body leading to irreversible target organ damage. As the role of secondary mediators (inflammatory molecules) and their mechanisms has not been fully elucidated, we focused on the interleukin (IL)-6 system in adult FD patients and in matched healthy subjects. To obtain insights into the complex regulation of IL-6 actions, we used a novel approach that integrates information from plasma and exosomes of FD patients (n = 20) and of healthy controls (n = 15). Soluble IL-6 receptor (sIL-6R) levels were measured in plasma with the ELISA method, and membrane-bound IL-6R was quantified in plasma and urinary exosomes using flow cytometry. In FD patients, the levels of soluble IL-6R in plasma were higher than in control subjects (28.0 ± 5.4 ng/mL vs. 18.9 ± 5.4 ng/mL, p < 0.0001); they were also higher in FD subjects with the classical form as compared to those with the late-onset form of the disease (36.0 ± 11.4 ng/mL vs. 26.1 ± 4.5 ng/mL, p < 0.0001). The percentage of urinary exosomes positive for IL-6R was slightly lower in FD (97 ± 1 vs. 100 ± 0% of events positive for IL-6R, p < 0.05); plasma IL-6 levels were not increased. These results suggest a potential role of IL-6 in triggering the inflammatory response in FD. As in FD patients only the levels of sIL-6Rs are consistently higher than in healthy controls, the IL-6 pathogenic signal seems to prevail over the homeostatic one, suggesting a potential mechanism causing multi-systemic damage in FD.
Collapse
Affiliation(s)
- Livia Lenzini
- Internal & Emergency Medicine Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (L.L.); (G.P.R.)
| | - Elisabetta Iori
- Division of Metabolic Diseases, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (E.I.); (M.V.); (A.A.)
| | - Monica Vettore
- Division of Metabolic Diseases, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (E.I.); (M.V.); (A.A.)
| | - Giorgia Gugelmo
- Division of Clinical Nutrition, Department of Medicine, Padova University Hospital, 35128 Padova, Italy;
| | - Claudia Radu
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (C.R.); (P.S.)
| | - Andrea Padoan
- Laboratory Medicine Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy;
| | - Gianni Carraro
- Nephrology, Dialysis and Transplant Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (G.C.); (L.C.)
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (C.R.); (P.S.)
| | - Lorenzo Calò
- Nephrology, Dialysis and Transplant Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (G.C.); (L.C.)
| | - Angelo Avogaro
- Division of Metabolic Diseases, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (E.I.); (M.V.); (A.A.)
| | - Gian Paolo Rossi
- Internal & Emergency Medicine Unit, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (L.L.); (G.P.R.)
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine, Padova University Hospital, 35128 Padova, Italy; (E.I.); (M.V.); (A.A.)
| |
Collapse
|
15
|
Izhar R, Borriello M, La Russa A, Di Paola R, De A, Capasso G, Ingrosso D, Perna AF, Simeoni M. Fabry Disease in Women: Genetic Basis, Available Biomarkers, and Clinical Manifestations. Genes (Basel) 2023; 15:37. [PMID: 38254927 PMCID: PMC10815601 DOI: 10.3390/genes15010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
Fabry Disease (FD) is a rare lysosomal storage disorder caused by mutations in the GLA gene on the X chromosome, leading to a deficiency in α-galactosidase A (AGAL) enzyme activity. This leads to the accumulation of glycosphingolipids, primarily globotriaosylceramide (Gb3), in vital organs such as the kidneys, heart, and nervous system. While FD was initially considered predominantly affecting males, recent studies have uncovered that heterozygous Fabry women, carrying a single mutated GLA gene, can manifest a wide array of clinical symptoms, challenging the notion of asymptomatic carriers. The mechanisms underlying the diverse clinical manifestations in females remain not fully understood due to X-chromosome inactivation (XCI). XCI also known as "lyonization", involves the random inactivation of one of the two X chromosomes. This process is considered a potential factor influencing phenotypic variation. This review delves into the complex landscape of FD in women, discussing its genetic basis, the available biomarkers, clinical manifestations, and the potential impact of XCI on disease severity. Additionally, it highlights the challenges faced by heterozygous Fabry women, both in terms of their disease burden and interactions with healthcare professionals. Current treatment options, including enzyme replacement therapy, are discussed, along with the need for healthcare providers to be well-informed about FD in women, ultimately contributing to improved patient care and quality of life.
Collapse
Affiliation(s)
- Raafiah Izhar
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.D.P.); (A.D.)
| | - Margherita Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (D.I.)
| | - Antonella La Russa
- Department of Sperimental Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Rossella Di Paola
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.D.P.); (A.D.)
| | - Ananya De
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.D.P.); (A.D.)
| | | | - Diego Ingrosso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.B.); (D.I.)
| | - Alessandra F. Perna
- Nephrology and Dialysis Unit, Department of Translation Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Mariadelina Simeoni
- Nephrology and Dialysis Unit, Department of Translation Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| |
Collapse
|
16
|
Stankowski K, Figliozzi S, Battaglia V, Catapano F, Francone M, Monti L. Fabry Disease: More than a Phenocopy of Hypertrophic Cardiomyopathy. J Clin Med 2023; 12:7061. [PMID: 38002674 PMCID: PMC10671939 DOI: 10.3390/jcm12227061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Fabry disease (FD) is a genetic lysosomal storage disease with frequent cardiovascular involvement, whose presence is a major determinant of adverse clinical outcomes. As a potentially treatable cause of left ventricular hypertrophy (LVH) and heart failure with preserved ejection fraction, the early recognition of FD is crucial to initiate enzyme replacement therapy and improve long-term prognosis. Multimodality imaging plays a central role in the evaluation of patients with FD and helps in the differential diagnosis of other conditions presenting with LVH. In the present review, we explore the current applications of multimodality cardiac imaging, in particular echocardiography and cardiovascular magnetic resonance, in the diagnosis, prognostic assessment, and follow-up of patients with FD.
Collapse
Affiliation(s)
- Kamil Stankowski
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| | - Stefano Figliozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| | - Vincenzo Battaglia
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| | - Federica Catapano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| | - Marco Francone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| | - Lorenzo Monti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, Pieve Emanuele, 20090 Milano, Italy; (K.S.); (S.F.); (V.B.); (F.C.); (M.F.)
- Humanitas Research Hospital IRCCS, Via Alessandro Manzoni, 56, Rozzano, 20089 Milano, Italy
| |
Collapse
|
17
|
Spinelli L. Impairment of sympathetic activity in Fabry disease cardiomyopathy: A further challenge for cardiac imaging. J Nucl Cardiol 2023; 30:1822-1824. [PMID: 37142879 DOI: 10.1007/s12350-023-03261-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 05/06/2023]
Affiliation(s)
- Letizia Spinelli
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini, 5, 80131, Naples, Italy.
| |
Collapse
|
18
|
Lillo R, Graziani F, Franceschi F, Iannaccone G, Massetti M, Olivotto I, Crea F, Liuzzo G. Inflammation across the spectrum of hypertrophic cardiac phenotypes. Heart Fail Rev 2023; 28:1065-1075. [PMID: 37115472 PMCID: PMC10403403 DOI: 10.1007/s10741-023-10307-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
The hypertrophic cardiomyopathy phenotype encompasses a heterogeneous spectrum of genetic and acquired diseases characterized by the presence of left ventricular hypertrophy in the absence of abnormal cardiac loading conditions. This "umbrella diagnosis" includes the "classic" hypertrophic cardiomyopathy (HCM), due to sarcomere protein gene mutations, and its phenocopies caused by intra- or extracellular deposits, such as Fabry disease (FD) and cardiac amyloidosis (CA). All these conditions share a wide phenotypic variability which results from the combination of genetic and environmental factors and whose pathogenic mediators are poorly understood so far. Accumulating evidence suggests that inflammation plays a critical role in a broad spectrum of cardiovascular conditions, including cardiomyopathies. Indeed, inflammation can trigger molecular pathways which contribute to cardiomyocyte hypertrophy and dysfunction, extracellular matrix accumulation, and microvascular dysfunction. Growing evidence suggests that systemic inflammation is a possible key pathophysiologic process potentially involved in the pathogenesis of cardiac disease progression, influencing the severity of the phenotype and clinical outcome, including heart failure. In this review, we summarize current knowledge regarding the prevalence, clinical significance, and potential therapeutic implications of inflammation in HCM and two of its most important phenocopies, FD and CA.
Collapse
Affiliation(s)
- Rosa Lillo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesca Graziani
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy.
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Giulia Iannaccone
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Iacopo Olivotto
- Cardiology Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, Rome, 00168, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
19
|
Frustaci A, Verardo R, Magnocavallo M, Scialla R, Sansone L, Russo MA. Circulating Anti-GB3 Antibody as a Biomarker of Myocardial Inflammation in Patients with Fabry Disease Cardiomyopathy. J Clin Med 2023; 12:4068. [PMID: 37373761 DOI: 10.3390/jcm12124068] [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: 04/06/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Fabry disease cardiomyopathy (FDCM) has manifested some resistance to enzyme replacement therapy (ERT), particularly in its advanced phase. Recently, myocardial inflammation of autoimmune origin has been demonstrated in FDCM. AIMS The objective of this study was the assessment of circulating anti-globotriaosylceramide (GB3) antibodies as potential biomarkers of myocardial inflammation in FDCM, defined by the additional presence of ≥CD3+ 7 T lymphocytes/low-power field associated with focal necrosis of adjacent myocytes. Its sensitivity was based on the evidence of overlapping myocarditis at left ventricular endomyocardial biopsy. METHODS AND RESULTS From January 1996 to December 2021, 85 patients received a histological diagnosis of FDCM in our department and 48 (56.5%) of them had an overlapping myocardial inflammation with negative PCR for common cardiotropic viruses, positive antiheart, and antimyosin abs. The presence of anti-GB3 antibodies was evaluated with an in-house ELISA assay (BioGeM scarl Medical Investigational Research, MIR-Ariano Irpino, Italy), along with antiheart and antimyosin abs, in the FDCM patients and compared with control healthy individuals. The correlation between levels of circulating anti-GB3 autoantibody myocardial inflammation and FDCM severity was assessed. Anti-Gb3 antibodies were above the positivity cut-off in 87.5% of FDCM subjects with myocarditis (42 out of 48), while 81.1% of FDCM patients without myocarditis were identified as negative for Gb3 antibodies. Positive anti-Gb3 abs correlated with positive antiheart and antimyosin abs. CONCLUSIONS The present study suggests a potential positive role of anti-GB3 antibodies as a marker of overlapping cardiac inflammation in patients with FDCM.
Collapse
Affiliation(s)
| | - Romina Verardo
- Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Roma, Italy
| | - Michele Magnocavallo
- Arrhythmology Unit, Ospedale Fatebenefratelli Isola Tiberina-Gemelli Isola, 00186 Rome, Italy
| | - Rossella Scialla
- Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Roma, Italy
| | - Luigi Sansone
- IRCCS San Raffaele Rome, 00166 Roma, Italy
- MEBIC Consortium, San Raffaele Open University, 00166 Rome, Italy
| | - Matteo Antonio Russo
- IRCCS San Raffaele Rome, 00166 Roma, Italy
- MEBIC Consortium, San Raffaele Open University, 00166 Rome, Italy
| |
Collapse
|
20
|
Burlina A, Brand E, Hughes D, Kantola I, Krӓmer J, Nowak A, Tøndel C, Wanner C, Spada M. An expert consensus on the recommendations for the use of biomarkers in Fabry disease. Mol Genet Metab 2023; 139:107585. [PMID: 37207471 DOI: 10.1016/j.ymgme.2023.107585] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by the accumulation of glycosphingolipids in various tissues and body fluids, leading to progressive organ damage and life-threatening complications. Phenotypic classification is based on disease progression and severity and can be used to predict outcomes. Patients with a classic Fabry phenotype have little to no residual α-Gal A activity and have widespread organ involvement, whereas patients with a later-onset phenotype have residual α-Gal A activity and disease progression can be limited to a single organ, often the heart. Diagnosis and monitoring of patients with Fabry disease should therefore be individualized, and biomarkers are available to support with this. Disease-specific biomarkers are useful in the diagnosis of Fabry disease; non-disease-specific biomarkers may be useful to assess organ damage. For most biomarkers it can be challenging to prove they translate to differences in the risk of clinical events associated with Fabry disease. Therefore, careful monitoring of treatment outcomes and collection of prospective data in patients are needed. As we deepen our understanding of Fabry disease, it is important to regularly re-evaluate and appraise published evidence relating to biomarkers. In this article, we present the results of a literature review of evidence published between February 2017 and July 2020 on the impact of disease-specific treatment on biomarkers and provide an expert consensus on clinical recommendations for the use of those biomarkers.
Collapse
Affiliation(s)
- Alessandro Burlina
- Neurological Unit, St. Bassiano Hospital, Via dei Lotti 40, I-36061 Bassano del Grappa, Italy.
| | - Eva Brand
- Internal Medicine, Department of Nephrology, Hypertension and Rheumatology; Interdisciplinary Fabry Center Münster (IFAZ), University Hospital Münster, Münster, Germany
| | - Derralynn Hughes
- Lysosomal Storage Disorders Unit, Royal Free London NHS Foundation Trust, University College London, United Kingdom
| | - Ilkka Kantola
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Johannes Krӓmer
- Pediatric Neurology and Metabolism, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Albina Nowak
- Department of Endocrinology and Clinical Nutrition, University Hospital of Zurich, Zurich, Switzerland
| | - Camilla Tøndel
- Department of Clinical Science, University of Bergen and Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Christoph Wanner
- Department of Internal Medicine, Division of Nephrology, Fabry Center for Interdisciplinary Therapy (FAZIT), University Hospital of Würzburg, Würzburg, Germany
| | - Marco Spada
- Department of Pediatrics, University of Torino, Torino, Italy
| |
Collapse
|
21
|
Hiestand R, Nowak A, Sokolska JM, Chan R, Ruschitzka F, Manka R, Gruner C. Clinical and CMR characteristics associated with cardiac events in patients with Fabry disease. Int J Cardiol 2023; 382:46-51. [PMID: 37044180 DOI: 10.1016/j.ijcard.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND The assessment of late gadolinium enhancement (LGE) and left ventricular hypertrophy (LVH) by cardiac magnetic resonance (CMR) as diagnostic and prognostic maker in Fabry disease is advancing. We aimed to investigate the impact of clinical characteristics and CMR findings on cardiac outcome in patients with FD. METHODS In this study 55 patients with genetically confirmed FD and available CMR imaging were included. The primary endpoint was defined as a composite of cardiac events including cardiac death, new occurrence of atrial fibrillation, heart failure, ventricular tachycardia and bradycardia requiring device insertion. RESULTS During a median follow-up of 4.9 years (IQR 3.7-5.9), 9 patients (16.3%) reached the primary cardiac end point. The global amount of LGE was associated with an increased risk for primary endpoint in the univariate analysis (HR 1.4 per 10% increase in LGE, p = 0.002). However maximal wall thickness (MWT) was the sole independent predictor of the primary endpoint in a stepwise logistic regression model (HR 9.8 per mm increase in MWT, p < 0.0001). Kaplan-Meier analysis revealed significant difference in event free survival rate between patients with and without LVH (Long-rank p = 0.006) and in patients with and without LGE (Long-rank p < 0.001). Patients without LVH and LGE were free of adverse cardiac events. CONCLUSION LVH and LGE detected by CMR were associated with adverse cardiac events in FD. In particular maximal wall thickness can be useful in cardiac risk stratification of FD patients.
Collapse
Affiliation(s)
- Roxana Hiestand
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland.
| | - Albina Nowak
- Department of Endocrinology and Clinical Nutrition, University Hospital Zurich and University of Zurich, Switzerland; Department of Internal Medicine, Psychiatry University Hospital Zurich, Switzerland
| | - Justyna M Sokolska
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland; Department of Cardiovascular Imaging, Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Raymond Chan
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland
| | - Robert Manka
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland
| | - Christiane Gruner
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Tebani A, Barbey F, Dormond O, Ducatez F, Marret S, Nowak A, Bekri S. Deep next-generation proteomics and network analysis reveal systemic and tissue-specific patterns in Fabry disease. Transl Res 2023:S1931-5244(23)00038-5. [PMID: 36863609 DOI: 10.1016/j.trsl.2023.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
Fabry disease (FD) is an X-linked lysosomal rare disease due to a deficiency of α-galactosidase A activity. The accumulation of glycosphingolipids mainly affects the kidney, heart, and central nervous system, considerably reducing life expectancy. Although the accumulation of undegraded substrate is considered the primary cause of FD, it is established that secondary dysfunctions at the cellular, tissue, and organ levels ultimately give rise to the clinical phenotype. To parse this biological complexity, a large-scale deep plasma targeted proteomic profiling has been performed. We analyzed the plasma protein profiles of FD deeply phenotyped patients (n = 55) compared to controls (n = 30) using next-generation plasma proteomics including 1463 proteins. Systems biology and machine learning approaches have been used. The analysis enabled the identification of proteomic profiles that unambiguously separated FD patients from controls (615 differentially expressed proteins, 476 upregulated, and 139 downregulated) and 365 proteins are newly reported. We observed functional remodeling of several processes, such as cytokine-mediated pathways, extracellular matrix, and vacuolar/lysosomal proteome. Using network strategies, we probed patient-specific tissue metabolic remodeling and described a robust predictive consensus protein signature including 17 proteins CD200, SPINT1, CD34, FGFR2, GRN, ERBB4, AXL, ADAM15, PTPRM, IL13RA1, NBL1, NOTCH1, VASN, ROR1, AMBP, CCN3, and HAVCR2. Our findings highlight the pro-inflammatory cytokines' involvement in FD pathogenesis along with extracellular matrix remodeling. The study shows a tissue-wide metabolic remodeling connection to plasma proteomics in FD. These results will facilitate further studies to understand the molecular mechanisms in FD to pave the way for better diagnostics and therapeutics.
Collapse
Affiliation(s)
- Abdellah Tebani
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, Rouen, France
| | - Frédéric Barbey
- University of Lausanne and University Hospital of Lausanne, Department of Immunology, Switzerland
| | - Olivier Dormond
- Lausanne University Hospital and University of Lausanne, Department of Visceral Surgery, Lausanne, Switzerland
| | - Franklin Ducatez
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, Rouen, France; Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Neonatal Pediatrics, Intensive Care, and Neuropediatrics, Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Neonatal Pediatrics, Intensive Care, and Neuropediatrics, Rouen, France
| | - Albina Nowak
- University Hospital and University of Zurich, Department of Endocrinology and Clinical Nutrition, Zurich, Switzerland
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, Rouen, France.
| |
Collapse
|
23
|
Enzyme Replacement Therapy for FABRY Disease: Possible Strategies to Improve Its Efficacy. Int J Mol Sci 2023; 24:ijms24054548. [PMID: 36901983 PMCID: PMC10003632 DOI: 10.3390/ijms24054548] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Enzyme replacement therapy is the only therapeutic option for Fabry patients with completely absent AGAL activity. However, the treatment has side effects, is costly, and requires conspicuous amounts of recombinant human protein (rh-AGAL). Thus, its optimization would benefit patients and welfare/health services (i.e., society at large). In this brief report, we describe preliminary results paving the way for two possible approaches: i. the combination of enzyme replacement therapy with pharmacological chaperones; and ii. the identification of AGAL interactors as possible therapeutic targets on which to act. We first showed that galactose, a low-affinity pharmacological chaperone, can prolong AGAL half-life in patient-derived cells treated with rh-AGAL. Then, we analyzed the interactomes of intracellular AGAL on patient-derived AGAL-defective fibroblasts treated with the two rh-AGALs approved for therapeutic purposes and compared the obtained interactomes to the one associated with endogenously produced AGAL (data available as PXD039168 on ProteomeXchange). Common interactors were aggregated and screened for sensitivity to known drugs. Such an interactor-drug list represents a starting point to deeply screen approved drugs and identify those that can affect (positively or negatively) enzyme replacement therapy.
Collapse
|
24
|
Umer M, Kalra DK. Treatment of Fabry Disease: Established and Emerging Therapies. Pharmaceuticals (Basel) 2023; 16:320. [PMID: 37259462 PMCID: PMC9967779 DOI: 10.3390/ph16020320] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 01/14/2024] Open
Abstract
Fabry disease (FD) is a rare, X-linked inherited disorder of glycosphingolipid metabolism. It leads to the progressive accumulation of globotriaosylceramide within lysosomes due to a deficiency of α-galactosidase A enzyme. It involves multiple organs, predominantly the renal, cardiac, and cerebrovascular systems. Early diagnosis and treatment are critical to prevent progression to irreversible tissue damage and organ failure, and to halt life-threatening complications that can significantly reduce life expectancy. This review will focus on the established and emerging treatment options for FD.
Collapse
Affiliation(s)
| | - Dinesh K. Kalra
- Division of Cardiology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
25
|
Perera K, Kashyap N, Wang K, Omar F, Prosia E, Thompson RB, Paterson DI, Fine NM, White JA, Khan A, Oudit GY. Integrating Cardiac MRI Imaging and Multidisciplinary Clinical Care is Associated With Improved Outcomes in Patients With Fabry Disease. Curr Probl Cardiol 2023; 48:101476. [PMID: 36328338 DOI: 10.1016/j.cpcardiol.2022.101476] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/26/2022] [Indexed: 01/04/2023]
Abstract
Given the inherent complexities of Fabry disease (FD) and evolving landscape of cardiovascular clinical management, there is no established ideal clinical care model for these patients. We identified clinical factors predictive of increased risk of major adverse cardiac events (MACE) in patients with FD targeted to improve clinical outcomes. Ninety-five patients studied over a median follow-up time of 6.3 years, and 26 patients reached the composite endpoint with a high prevalence of heart failure and cerebrovascular events and no cardiac-related mortality. Patients with MACE had worse health-related quality of life scores. Hypertrophy and presence of myocardial fibrosis increase risk of MACE by 4-5 times, and dyslipidemia increases risk of MACE by 3 times. Early Fabry-specific treatment and close monitoring of comorbidities reduce cardiac complications and mortality. These findings highlight the importance of comprehensive multidisciplinary management to help improve outcomes in FD patients.
Collapse
Affiliation(s)
- Kevin Perera
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Niharika Kashyap
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kaiming Wang
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Fadya Omar
- School of Health and Public Safety, Southern Alberta Institute of Technology, Calgary, Alberta, Canada; Metabolics and Genetics in Canada (M.A.G.I.C.) Clinic Ltd., Calgary, Alberta, Canada
| | - Easter Prosia
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary Alberta, Canada
| | - Richard B Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - D Ian Paterson
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Nowell M Fine
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary Alberta, Canada
| | - James A White
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary Alberta, Canada
| | - Aneal Khan
- Metabolics and Genetics in Canada (M.A.G.I.C.) Clinic Ltd., Calgary, Alberta, Canada; Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
26
|
Rudenko KV, Nevmerzhytska LO, Unitska OM, Danchenko PA, Leiko NS. Fabry Disease, a Rare Disorder with Cardiac Manifestations. The Problem of Diagnosis and Treatment: a Literature Review. UKRAINIAN JOURNAL OF CARDIOVASCULAR SURGERY 2022. [DOI: 10.30702/ujcvs/22.30(04)/rn047-7380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disease caused by a mutation in the gene encoding α-galactosidase A and leads to reduced activity or complete absence of this enzyme, which causes the accumulation of globotriaosylceramide (Gb3) and its deacylated form (lyso-Gb3) in cells of the whole body. FD can occur both with multisystem manifestations, including damage to the nervous system, kidneys, and skin, and can affect only the heart. Cardiac involvement is a major cause of poor quality of life and death in patients with FD and an underrecognized cause of heart failure with preserved ejection fraction and ventricular arrhythmias in men over 30 years of age and women over 40 years of age. Cardiac damage begins at an early age, progresses subclinically until the appearance of significant symptoms, and usually manifests as leftventricular hypertrophy, mimicking hypertrophic cardiomyopathy.
After the introduction of enzyme replacement therapy, early recognition of FD and differential diagnosis with other causes of leftventricular hypertrophy have become crucial to limit the progression of the disease. Recent advances in the understanding of cardiac pathophysiology and imaging have improved diagnostic and therapeutic approaches to the cardiac manifestations of this pathology.
Modern achievements in the study of cardiac manifestations of FD have made it possible to significantly improve diagnostic and therapeutic approaches, in particular, in relation to the identification of pathogenetic mechanisms of organ damage and early disruption of their function. A better understanding of secondary pathogenic pathways, such as myocardial inflammation, may influence future therapeutic strategies and timely diagnosis of FD.
Delay in diagnosis and untimely initiation of treatment remain critical problems for many patients with FD, especially for patients with late-onset cardiovascular manifestations, in whom treatment effects may be more limited and ineffective.
Cooperation between specialists in genetic diseases and cardiologists remains important to identify patients before the appearance of cardiac symptoms in order to obtain maximum therapeutic effects.
Collapse
|
27
|
Tran BTT, Gelin A, Durand S, Texier M, Daste A, Toullec C, Benihoud K, Breuskin I, Gorphe P, Garic F, Brenner C, Le Tourneau C, Fayette J, Niki T, David M, Busson P, Even C. Plasma galectins and metabolites in advanced head and neck carcinomas: evidence of distinct immune characteristics linked to hypopharyngeal tumors. Oncoimmunology 2022; 12:2150472. [PMID: 36545254 PMCID: PMC9762837 DOI: 10.1080/2162402x.2022.2150472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Extra-cellular galectins 1, 3 and 9 (gal-1, -3 and -9) are known to act as soluble immunosuppressive agents in various malignancies. Previous publications have suggested that their expression is dependent on the metabolic status of producing cells and reciprocally that they can influence metabolic pathways in their target cells. Very little is known about the status of gal-1, -3 and -9 in patients bearing head and neck squamous cell carcinomas (HNSCC) and about their relationships with the systemic metabolic condition. This study was conducted in plasma samples from a prospective cohort of 83 HNSCC patients with advanced disease. These samples were used to explore the distribution of gal-1, -3 and -9 and simultaneously to profile a series of 87 metabolites assessed by mass spectrometry. We identified galectin and metabolic patterns within five disease categories defined according to the primary site and human papillomavirus (HPV) status (HPV-positive and -negative oropharyngeal carcinomas, carcinomas of the oral cavity, hypopharynx and larynx carcinomas). Remarkably, samples related to hypopharyngeal carcinomas displayed the highest average concentration of gal-9 (p = .017) and a trend toward higher concentrations of kynurenine, a potential factor of tumor growth and immune suppression. In contrast, there was a tendency toward higher concentrations of fatty acids in samples related to oral cavity. These observations emphasize the diversity of HPV-negative HNSCCs. Depending on their primary site, they evolve into distinct types of immune and metabolic landscapes that seem to be congruent with specific oncogenic mechanisms.
Collapse
Affiliation(s)
- Bao-Tram Thi Tran
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Aurore Gelin
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Sylvère Durand
- Plateforme de Métabolomique/UMR 1138, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Matthieu Texier
- Service d’Epidémiologie et de Biostatistiques, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Amaury Daste
- Department of Medical Oncology, Hôpital Saint André, Bordeaux, France
| | - Clémence Toullec
- GI and Liver/Head and Neck unit, Institut du Cancer-Avignon Provence, Avignon, France
| | - Karim Benihoud
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Ingrid Breuskin
- Service de Cancérologie Cervico-Faciale, Gustave Roussy, Villejuif, France
| | - Philippe Gorphe
- Service de Cancérologie Cervico-Faciale, Gustave Roussy, Villejuif, France
| | | | - Catherine Brenner
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie and Paris-Saclay University, Paris, France
| | - Jérôme Fayette
- Claude Bernard Lyon 1 University, INSERM 1052, CNRS 5286 & Department of Medical Oncology, Centre Léon Bérard, Cancer Research Center of Lyon, Lyon, France
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Kita-gun, Japan
| | - Muriel David
- HiFiBiO Therapeutics, Pépinière Paris Santé Cochin, Paris, France
| | - Pierre Busson
- CNRS UMR 9018-METSY, Gustave Roussy and Université Paris-Saclay, Villejuif, France,CONTACT Pierre Busson CNRS UMR 9018-METSY, Gustave Roussy, 39, Rue Camile Desmoulins, F-94805Villejuif, France
| | - Caroline Even
- Service de Cancérologie Cervico-Faciale, Gustave Roussy, Villejuif, France
| |
Collapse
|
28
|
Li X, Ren X, Zhang Y, Ding L, Huo M, Li Q. Fabry disease: Mechanism and therapeutics strategies. Front Pharmacol 2022; 13:1025740. [PMID: 36386210 PMCID: PMC9643830 DOI: 10.3389/fphar.2022.1025740] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Fabry disease is a monogenic disease characterized by a deficiency or loss of the α-galactosidase A (GLA). The resulting impairment in lysosomal GLA enzymatic activity leads to the pathogenic accumulation of enzymatic substrate and, consequently, the progressive appearance of clinical symptoms in target organs, including the heart, kidney, and brain. However, the mechanisms involved in Fabry disease-mediated organ damage are largely ambiguous and poorly understood, which hinders the development of therapeutic strategies for the treatment of this disorder. Although currently available clinical approaches have shown some efficiency in the treatment of Fabry disease, they all exhibit limitations that need to be overcome. In this review, we first introduce current mechanistic knowledge of Fabry disease and discuss potential therapeutic strategies for its treatment. We then systemically summarize and discuss advances in research on therapeutic approaches, including enzyme replacement therapy (ERT), gene therapy, and chaperone therapy, as well as strategies targeting subcellular compartments, such as lysosomes, the endoplasmic reticulum, and the nucleus. Finally, the future development of potential therapeutic strategies is discussed based on the results of mechanistic studies and the limitations associated with these therapeutic approaches.
Collapse
Affiliation(s)
- Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiangyi Ren
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yabing Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Ding
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Minfeng Huo
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| |
Collapse
|
29
|
An Overview of Molecular Mechanisms in Fabry Disease. Biomolecules 2022; 12:biom12101460. [PMID: 36291669 PMCID: PMC9599883 DOI: 10.3390/biom12101460] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 12/24/2022] Open
Abstract
Fabry disease (FD) (OMIM #301500) is a rare genetic lysosomal storage disorder (LSD). LSDs are characterized by inappropriate lipid accumulation in lysosomes due to specific enzyme deficiencies. In FD, the defective enzyme is α-galactosidase A (α-Gal A), which is due to a mutation in the GLA gene on the X chromosome. The enzyme deficiency leads to a continuous deposition of neutral glycosphingolipids (globotriaosylceramide) in the lysosomes of numerous tissues and organs, including endothelial cells, smooth muscle cells, corneal epithelial cells, renal glomeruli and tubules, cardiac muscle and ganglion cells of the nervous system. This condition leads to progressive organ failure and premature death. The increasing understanding of FD, and LSD in general, has led in recent years to the introduction of enzyme replacement therapy (ERT), which aims to slow, if not halt, the progression of the metabolic disorder. In this review, we provide an overview of the main features of FD, focusing on its molecular mechanism and the role of biomarkers.
Collapse
|
30
|
Umer M, Motwani M, Jefferies JL, Kalra DK. Cardiac involvement in Fabry Disease and the Role of Multimodality Imaging in Diagnosis and Disease Monitoring. Curr Probl Cardiol 2022; 48:101439. [DOI: 10.1016/j.cpcardiol.2022.101439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022]
|
31
|
Fu L, Wasiak S, Tsujikawa LM, Rakai BD, Stotz SC, Wong NCW, Johansson JO, Sweeney M, Mohan CM, Khan A, Kulikowski E. Inhibition of epigenetic reader proteins by apabetalone counters inflammation in activated innate immune cells from Fabry disease patients receiving enzyme replacement therapy. Pharmacol Res Perspect 2022; 10:e00949. [PMID: 35417091 PMCID: PMC9007222 DOI: 10.1002/prp2.949] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 12/14/2022] Open
Abstract
Fabry disease (FD) is a rare X‐linked disorder of lipid metabolism, characterized by the accumulation of globotriaosylceramide (Gb3) due to defective the lysosomal enzyme, α‐galactosidase. Gb3 deposits activate immune‐mediated systemic inflammation, ultimately leading to life‐threatening consequences in multiple organs such as the heart and kidneys. Enzyme replacement therapy (ERT), the standard of care, is less effective with advanced tissue injury and inflammation in patients with FD. Here, we showed that MCP‐1 and TNF‐α cytokine levels were almost doubled in plasma from ERT‐treated FD patients. Chemokine receptor CCR2 surface expression was increased by twofold on monocytes from patients with low eGFR. We also observed an increase in IL12B transcripts in unstimulated peripheral blood mononuclear cells (PBMCs) over a 2‐year period of continuous ERT. Apabetalone is a clinical‐stage oral bromodomain and extra terminal protein inhibitor (BETi), which has beneficial effects on cardiovascular and kidney disease related pathways including inflammation. Here, we demonstrate that apabetalone, a BD2‐selective BETi, dose dependently reduced the production of MCP‐1 and IL‐12 in stimulated PBMCs through transcriptional regulation of their encoding genes. Reactive oxygen species production was diminished by up to 80% in stimulated neutrophils following apabetalone treatment, corresponding with inhibition of NOX2 transcription. This study elucidates that inhibition of BET proteins by BD2‐selective apabetalone alleviates inflammatory processes and oxidative stress in innate immune cells in general and in FD. These results suggest potential benefit of BD2‐selective apabetalone in controlling inflammation and oxidative stress in FD, which will be further investigated in clinical trials.
Collapse
Affiliation(s)
- Li Fu
- Resverlogix Corp, Calgary, AB, Canada
| | | | | | | | | | | | | | | | - Connie M Mohan
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aneal Khan
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Metabolics and Genetics in Calgary (M.A.G.I.C.) Clinic Ltd., Calgary, AB, Canada
| | | |
Collapse
|
32
|
Fuertes Kenneally L, García-Álvarez MI, Feliu Rey E, García Barrios A, Climent-Payá V. Fabry Disease Cardiomyopathy: A Review of the Role of Cardiac Imaging from Diagnosis to Treatment. Rev Cardiovasc Med 2022; 23:192. [PMID: 39077169 PMCID: PMC11273868 DOI: 10.31083/j.rcm2306192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 07/31/2024] Open
Abstract
Fabry disease is a rare X-linked inherited lysosomal storage disorder caused by the absence or reduction of alfa-galactosidase A activity in lysosomes, resulting in accumulation of glycosphingolipids in various tissues. The main organ affected is the heart, which frequently manifests as left ventricular hypertrophy and can ultimately lead to cardiac fibrosis, heart failure, valve disease, cardiac conduction abnormalities and sudden cardiac death. Today we know that myocyte damage starts before these signs and symptoms are detectable on routine studies, during the designated pre-clinical phase of Fabry disease. The initiation of specific therapy for Fabry disease during the early stages of the disease has a great impact on the prognosis of these patients avoiding progression to irreversible fibrosis and preventing cardiovascular complications. Cardiac imaging has become an essential tool in the management of Fabry disease as it can help physicians suspect the disorder, diagnose patients in the early stages and improve outcomes. The recent development of novel imaging techniques makes necessary an update on the subject. This review discusses the role of multimodal imaging in the diagnosis, staging, patient selection for treatment and prognosis of Fabry disease and discusses recent advances in imaging techniques that provide new insights into the pathogenesis of the disorder and the possibility of novel treatment targets.
Collapse
Affiliation(s)
- Laura Fuertes Kenneally
- Heart Failure and Inherited Cardiac Diseases Unit, Cardiology Department, Hospital General Universitario Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | - María Isabel García-Álvarez
- Heart Failure and Inherited Cardiac Diseases Unit, Cardiology Department, Hospital General Universitario Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | - Eloísa Feliu Rey
- Radiology Department, Hospital General Universitario Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | - Ana García Barrios
- Heart Failure and Inherited Cardiac Diseases Unit, Cardiology Department, Hospital General Universitario Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| | - Vicente Climent-Payá
- Heart Failure and Inherited Cardiac Diseases Unit, Cardiology Department, Hospital General Universitario Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
33
|
Pieroni M, Ciabatti M, Graziani F, Camporeale A, Saletti E, Lillo R, Figliozzi S, Bolognese L. The Heart in Fabry Disease: Mechanisms Beyond Storage and Forthcoming Therapies. Rev Cardiovasc Med 2022; 23:196. [PMID: 39077177 PMCID: PMC11273771 DOI: 10.31083/j.rcm2306196] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/29/2022] [Accepted: 04/13/2022] [Indexed: 07/31/2024] Open
Abstract
In patients with Fabry disease (FD), cardiovascular involvement is the main cause of death and reduction of quality of life. Left ventricular hypertrophy mimicking hypertrophic cardiomyopathy is the main feature of FD cardiac involvement although glycolipid storage occurs in all cardiac cellular types. Accumulation of lysosomal globotriasylceramide represents the main mechanism of cardiac damage in early stages, but secondary pathways of cellular and tissue damage, triggered by lysosomal storage, and including altered energy production, inflammation and cell death, contribute to cardiac damage and disease progression. These mechanisms appear prominent in more advanced stages, hampering and reducing the efficacy of FD-specific treatments. Therefore, additional cardiovascular therapies are important to manage cardiovascular symptoms and reduce cardiovascular events. Although new therapies targeting lysosomal storage are in development, a better definition and comprehension of the complex pathophysiology of cardiac damage in FD, may lead to identify new therapeutic targets beyond storage and new therapeutic strategies.
Collapse
Affiliation(s)
- Maurizio Pieroni
- Cardiovascular Department, San Donato Hospital, 52100 Arezzo, Italy
| | - Michele Ciabatti
- Cardiovascular Department, San Donato Hospital, 52100 Arezzo, Italy
| | - Francesca Graziani
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonia Camporeale
- Multimodality Cardiac Imaging Unit, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Elisa Saletti
- Cardiovascular Department, San Donato Hospital, 52100 Arezzo, Italy
| | - Rosa Lillo
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Stefano Figliozzi
- Clinical Echocardiography Diagnostic Service, Cardio Center, Humanitas Research Hospital IRCCS, 20089 Rozzano, Italy
| | | |
Collapse
|
34
|
Giugliani R, Marques S, Andrade LGMD, Pessoa A, Vaisbich MH, Blum A, Tenório F, Rosa Neto NS. Clinical and diagnostic aspects of Fabry disease management: a narrative review with a particular focus on Brazilian experts’ perspectives. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2022. [DOI: 10.1590/2326-4594-jiems-2021-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Roberto Giugliani
- Universidade Federal do Rio Grande do Sul, Brazil; Hospital de Clínicas de Porto Alegre, Brazil
| | | | | | - André Pessoa
- Hospital Infantil Albert Sabin, Brazil; Universidade Estadual do Ceará, Brazil
| | - Maria H. Vaisbich
- Universidade Federal de São Paulo, Brazil; Universidade de São Paulo (HCFMUSP), Brazil
| | | | | | | |
Collapse
|
35
|
Salamon I, Biagini E, Kunderfranco P, Roncarati R, Ferracin M, Taglieri N, Nardi E, Laprovitera N, Tomasi L, Santostefano M, Ditaranto R, Vitale G, Cavarretta E, Pisani A, Riccio E, Aiello V, Capelli I, La Manna G, Galiè N, Spinelli L, Condorelli G. Circulating miR-184 is a potential predictive biomarker of cardiac damage in Anderson-Fabry disease. Cell Death Dis 2021; 12:1150. [PMID: 34897278 PMCID: PMC8665928 DOI: 10.1038/s41419-021-04438-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022]
Abstract
Enzyme replacement therapy (ERT) is a mainstay of treatment for Anderson-Fabry disease (AFD), a pathology with negative effects on the heart and kidneys. However, no reliable biomarkers are available to monitor its efficacy. Therefore, we tested a panel of four microRNAs linked with cardiac and renal damage in order to identify a novel biomarker associated with AFD and modulated by ERT. To this end, 60 patients with a definite diagnosis of AFD and on chronic ERT, and 29 age- and sex-matched healthy individuals, were enrolled by two Italian university hospitals. Only miR-184 met both conditions: its level discriminated untreated AFD patients from healthy individuals (c-statistic = 0.7522), and it was upregulated upon ERT (P < 0.001). On multivariable analysis, miR-184 was independently and inversely associated with a higher risk of cardiac damage (odds ratio = 0.86; 95% confidence interval [CI] = 0.76-0.98; P = 0.026). Adding miR-184 to a comprehensive clinical model improved the prediction of cardiac damage in terms of global model fit, calibration, discrimination, and classification accuracy (continuous net reclassification improvement = 0.917, P < 0.001; integrated discrimination improvement [IDI] = 0.105, P = 0.017; relative IDI = 0.221, 95% CI = 0.002-0.356). Thus, miR-184 is a circulating biomarker of AFD that changes after ERT. Assessment of its level in plasma could be clinically valuable in improving the prediction of cardiac damage in AFD patients.
Collapse
Affiliation(s)
- Irene Salamon
- Humanitas Research Hospital - IRCCS, 20089, Rozzano, (MI), Italy
- Department of Biomedical Sciences, Humanitas University, 20090, Pieve Emanuele, (MI), Italy
| | - Elena Biagini
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | | | - Roberta Roncarati
- Institute of Genetics and Biomedical Research - Milan Unit, National Research Council of Italy, 20089, Rozzano, (MI), Italy
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121, Ferrara, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Nevio Taglieri
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Elena Nardi
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Noemi Laprovitera
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Luciana Tomasi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Marisa Santostefano
- Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Raffaello Ditaranto
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Giovanni Vitale
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Elena Cavarretta
- Department of Medico-Surgical Sciences and Biotechnologies, University of Rome Sapienza, 04100, Latina, Italy
- Mediterranea Cardiocentro, 80122, Naples, Italy
| | - Antonio Pisani
- Department of Public Health - Nephrology Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Eleonora Riccio
- Department of Public Health - Nephrology Unit, University of Naples Federico II, 80131, Naples, Italy
| | - Valeria Aiello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Irene Capelli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
- Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Nazzareno Galiè
- Cardiology Unit, St. Orsola Hospital, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Letizia Spinelli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy.
| | - Gianluigi Condorelli
- Humanitas Research Hospital - IRCCS, 20089, Rozzano, (MI), Italy.
- Department of Biomedical Sciences, Humanitas University, 20090, Pieve Emanuele, (MI), Italy.
| |
Collapse
|
36
|
Tsai YL, Chou RH, Lu YW, Chang CC, Kuo CS, Huang PH, Chen JW, Lin SJ. Associations between galectin-1, left ventricular diastolic dysfunction, and heart failure with preserved ejection fraction. J Cardiol 2021; 79:371-375. [PMID: 34774386 DOI: 10.1016/j.jjcc.2021.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/19/2021] [Accepted: 09/12/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Galectin-1 is a glycan-binding protein with broad anti-inflammatory properties. Left ventricular diastolic dysfunction (DD) is associated with heart failure and mortality. The pathophysiology of DD is complex and our study aimed to investigate the associations between serum galectin-1 level, DD, and heart failure with preserved ejection fraction (HFpEF). METHODS Patients with symptoms of angina pectoris were enrolled. Serum galectin-1 levels and echocardiography were assessed. The study endpoint was a composite of all-cause mortality or new-onset HFpEF. RESULTS In total, 258 patients were enrolled (63% male; mean age 68±12 years) and grouped into tertiles based on galectin-1 levels. Patients in the highest galectin-1 group had increased left ventricular mass indexes, left atrial diameters, and prevalence of DD compared to those in the lower tertiles (all p<0.05). Moreover, elevated galectin-1 levels were significantly associated with the composite endpoint (p=0.039). After adjusting for confounding factors, high galectin-1 levels remained significantly associated with DD (odds ratio 2.44, p=0.005). The Kaplan-Meier analysis revealed patients in the highest galectin-1 group had lowest cumulative survival of composite endpoint (log rank p=0.043). CONCLUSIONS Elevated serum galectin-1 levels were associated with DD and the composite endpoint of all-cause mortality and incident HFpEF.
Collapse
Affiliation(s)
- Yi-Lin Tsai
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ya-Wen Lu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chin Chang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chin-Sung Kuo
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan; Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
37
|
Ducatez F, Mauhin W, Boullier A, Pilon C, Pereira T, Aubert R, Benveniste O, Marret S, Lidove O, Bekri S, Tebani A. Parsing Fabry Disease Metabolic Plasticity Using Metabolomics. J Pers Med 2021; 11:jpm11090898. [PMID: 34575675 PMCID: PMC8468728 DOI: 10.3390/jpm11090898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Fabry disease (FD) is an X-linked lysosomal disease due to a deficiency in the activity of the lysosomal α-galactosidase A (GalA), a key enzyme in the glycosphingolipid degradation pathway. FD is a complex disease with a poor genotype–phenotype correlation. FD could involve kidney, heart or central nervous system impairment that significantly decreases life expectancy. The advent of omics technologies offers the possibility of a global, integrated and systemic approach well-suited for the exploration of this complex disease. Materials and Methods: Sixty-six plasmas of FD patients from the French Fabry cohort (FFABRY) and 60 control plasmas were analyzed using liquid chromatography and mass spectrometry-based targeted metabolomics (188 metabolites) along with the determination of LysoGb3 concentration and GalA enzymatic activity. Conventional univariate analyses as well as systems biology and machine learning methods were used. Results: The analysis allowed for the identification of discriminating metabolic profiles that unambiguously separate FD patients from control subjects. The analysis identified 86 metabolites that are differentially expressed, including 62 Glycerophospholipids, 8 Acylcarnitines, 6 Sphingomyelins, 5 Aminoacids and 5 Biogenic Amines. Thirteen consensus metabolites were identified through network-based analysis, including 1 biogenic amine, 2 lysophosphatidylcholines and 10 glycerophospholipids. A predictive model using these metabolites showed an AUC-ROC of 0.992 (CI: 0.965–1.000). Conclusion: These results highlight deep metabolic remodeling in FD and confirm the potential of omics-based approaches in lysosomal diseases to reveal clinical and biological associations to generate pathophysiological hypotheses.
Collapse
Affiliation(s)
- Franklin Ducatez
- Department of Metabolic Biochemistry, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France; (F.D.); (C.P.); (R.A.); (S.B.)
- Department of Neonatal Pediatrics, Intensive Care, and Neuropediatrics, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France;
| | - Wladimir Mauhin
- Department of Internal Medicine, Groupe Hospitalier Diaconesses Croix Saint Simon, Site Avron & UMRS 974, 75013 Paris, France; (W.M.); (O.L.)
| | - Agnès Boullier
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Avenue de la Croix Jourdain, 80054 Amiens, France;
- Laboratoire de Biochimie CHU Amiens-Picardie, Avenue de la Croix Jourdain, 80054 Amiens, France
| | - Carine Pilon
- Department of Metabolic Biochemistry, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France; (F.D.); (C.P.); (R.A.); (S.B.)
| | - Tony Pereira
- CHU Rouen, Institut de Biologie Clinique, 76000 Rouen, France;
| | - Raphaël Aubert
- Department of Metabolic Biochemistry, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France; (F.D.); (C.P.); (R.A.); (S.B.)
| | - Olivier Benveniste
- Department of Internal Medicine, Hôpital Pitié-Salpêtrière & INSERM U 974, 75013 Paris, France;
| | - Stéphane Marret
- Department of Neonatal Pediatrics, Intensive Care, and Neuropediatrics, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France;
| | - Olivier Lidove
- Department of Internal Medicine, Groupe Hospitalier Diaconesses Croix Saint Simon, Site Avron & UMRS 974, 75013 Paris, France; (W.M.); (O.L.)
| | - Soumeya Bekri
- Department of Metabolic Biochemistry, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France; (F.D.); (C.P.); (R.A.); (S.B.)
| | - Abdellah Tebani
- Department of Metabolic Biochemistry, Normandie University, UNIROUEN, INSERM U1245, CHU Rouen, 76000 Rouen, France; (F.D.); (C.P.); (R.A.); (S.B.)
- Correspondence:
| |
Collapse
|
38
|
Kanack AJ, Aoki K, Tiemeyer M, Dahms NM. Platelet and myeloid cell phenotypes in a rat model of Fabry disease. FASEB J 2021; 35:e21818. [PMID: 34320241 DOI: 10.1096/fj.202001727rr] [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: 07/10/2020] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/11/2022]
Abstract
Fabry disease results from a deficiency of the lysosomal enzyme ⍺-Galactosidase-A (⍺-Gal A) and is estimated to occur in approximately 1:4100 live births. Characteristic of the disease is the accumulation of α-Gal-A substrates, primarily the glycosphingolipids (GSLs) globotriaosylceramide and globotriaosylsphingosine. Thrombotic events are a significant concern for Fabry patients, with strokes contributing to a significant decrease in overall lifespan. Currently, the mechanisms underlying the increased risk of thrombotic events experienced by Fabry patients are incompletely defined. Using a rat model of Fabry disease, we provide an improved understanding of the mechanisms linking GSL accumulation to thrombotic risk. We found that ⍺-Gal A-deficient rats accumulate myeloid-derived leukocytes at sites of GSL accumulation, including in the bone marrow and circulation, and that myeloid-derived leukocyte and megakaryocyte populations were prominent among cell types that accumulated GSLs. In the circulation, ⍺-Gal A-deficient rats had increases in cytokine-producing cell types and a corresponding elevation of pro-inflammatory cytokines. Lastly, circulating platelets from ⍺-Gal A-deficient rats accumulated a similar set of ⍺-Galactosidase-A substrates as was observed in megakaryocytes in the bone marrow, and exhibited increased platelet binding to fibrinogen in microfluidic and flow cytometric assays.
Collapse
Affiliation(s)
- Adam J Kanack
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kazuhiro Aoki
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
39
|
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: 2.7] [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
|
40
|
Chung S, Son M, Chae Y, Oh S, Koh ES, Kim YK, Shin SJ, Park CW, Jung SC, Kim HS. Fabry disease exacerbates renal interstitial fibrosis after unilateral ureteral obstruction via impaired autophagy and enhanced apoptosis. Kidney Res Clin Pract 2021; 40:208-219. [PMID: 34024086 PMCID: PMC8237117 DOI: 10.23876/j.krcp.20.264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background Fabry disease is a rare X-linked genetic lysosomal disorder caused by mutations in the GLA gene encoding alpha-galactosidase A. Despite some data showing that profibrotic and proinflammatory cytokines and oxidative stress could be involved in Fabry disease-related renal injury, the pathogenic link between metabolic derangement within cells and renal injury remains unclear. Methods Renal fibrosis was triggered by unilateral ureteral obstruction (UUO) in mice with Fabry disease to investigate the pathogenic mechanism leading to fibrosis in diseased kidneys. Results Compared to kidneys of wild-type mice, lamellar inclusion bodies were recognized in proximal tubules of mice with Fabry disease. Sirius red and trichrome staining revealed significantly increased fibrosis in all UUO kidneys, though it was more prominent in obstructed Fabry kidneys. Renal messenger RNA levels of inflammatory cytokines and profibrotic factors were increased in all UUO kidneys compared to sham-operated kidneys but were not significantly different between UUO control and UUO Fabry mice. Protein levels of Nox2, Nox4, NQO1, catalase, SOD1, SOD2, and Nrf2 were not significantly different between UUO control and UUO Fabry kidneys, while the protein contents of LC3-II and LC3-I and expression of Beclin1 were significantly decreased in UUO kidneys of Fabry disease mouse models compared with wild-type mice. Notably, TUNEL-positive cells were elevated in obstructed kidneys of Fabry disease mice compared to wild-type control and UUO mice. Conclusion These findings suggest that impaired autophagy and enhanced apoptosis are probable mechanisms involved in enhanced renal fibrosis under the stimulus of UUO in Fabry disease.
Collapse
Affiliation(s)
- Sungjin Chung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mina Son
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yura Chae
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Songhee Oh
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sil Koh
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yong Kyun Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seok Joon Shin
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol Whee Park
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Chul Jung
- Department of Biochemistry, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Ho-Shik Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
41
|
Fabry Disease and the Heart: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22094434. [PMID: 33922740 PMCID: PMC8123068 DOI: 10.3390/ijms22094434] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder caused by mutations of the GLA gene that result in a deficiency of the enzymatic activity of α-galactosidase A and consequent accumulation of glycosphingolipids in body fluids and lysosomes of the cells throughout the body. GB3 accumulation occurs in virtually all cardiac cells (cardiomyocytes, conduction system cells, fibroblasts, and endothelial and smooth muscle vascular cells), ultimately leading to ventricular hypertrophy and fibrosis, heart failure, valve disease, angina, dysrhythmias, cardiac conduction abnormalities, and sudden death. Despite available therapies and supportive treatment, cardiac involvement carries a major prognostic impact, representing the main cause of death in FD. In the last years, knowledge has substantially evolved on the pathophysiological mechanisms leading to cardiac damage, the natural history of cardiac manifestations, the late-onset phenotypes with predominant cardiac involvement, the early markers of cardiac damage, the role of multimodality cardiac imaging on the diagnosis, management and follow-up of Fabry patients, and the cardiac efficacy of available therapies. Herein, we provide a comprehensive and integrated review on the cardiac involvement of FD, at the pathophysiological, anatomopathological, laboratory, imaging, and clinical levels, as well as on the diagnosis and management of cardiac manifestations, their supportive treatment, and the cardiac efficacy of specific therapies, such as enzyme replacement therapy and migalastat.
Collapse
|
42
|
Carnicer-Cáceres C, Arranz-Amo JA, Cea-Arestin C, Camprodon-Gomez M, Moreno-Martinez D, Lucas-Del-Pozo S, Moltó-Abad M, Tigri-Santiña A, Agraz-Pamplona I, Rodriguez-Palomares JF, Hernández-Vara J, Armengol-Bellapart M, del-Toro-Riera M, Pintos-Morell G. Biomarkers in Fabry Disease. Implications for Clinical Diagnosis and Follow-up. J Clin Med 2021; 10:jcm10081664. [PMID: 33924567 PMCID: PMC8068937 DOI: 10.3390/jcm10081664] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/01/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022] Open
Abstract
Fabry disease (FD) is a lysosomal storage disorder caused by deficient alpha-galactosidase A activity in the lysosome due to mutations in the GLA gene, resulting in gradual accumulation of globotriaosylceramide and other derivatives in different tissues. Substrate accumulation promotes different pathogenic mechanisms in which several mediators could be implicated, inducing multiorgan lesions, mainly in the kidney, heart and nervous system, resulting in clinical manifestations of the disease. Enzyme replacement therapy was shown to delay disease progression, mainly if initiated early. However, a diagnosis in the early stages represents a clinical challenge, especially in patients with a non-classic phenotype, which prompts the search for biomarkers that help detect and predict the evolution of the disease. We have reviewed the mediators involved in different pathogenic mechanisms that were studied as potential biomarkers and can be easily incorporated into clinical practice. Some accumulation biomarkers seem to be useful to detect non-classic forms of the disease and could even improve diagnosis of female patients. The combination of such biomarkers with some response biomarkers, may be useful for early detection of organ injury. The incorporation of some biomarkers into clinical practice may increase the capacity of detection compared to that currently obtained with the established diagnostic markers and provide more information on the progression and prognosis of the disease.
Collapse
Affiliation(s)
- Clara Carnicer-Cáceres
- Laboratory of Inborn Errors of Metabolism, Laboratoris Clínics, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.A.-A.); (C.C.-A.)
- Correspondence:
| | - Jose Antonio Arranz-Amo
- Laboratory of Inborn Errors of Metabolism, Laboratoris Clínics, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.A.-A.); (C.C.-A.)
| | - Cristina Cea-Arestin
- Laboratory of Inborn Errors of Metabolism, Laboratoris Clínics, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (J.A.A.-A.); (C.C.-A.)
| | - Maria Camprodon-Gomez
- Department of Internal Medicine, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.C.-G.); (D.M.-M.)
- Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.T.-S.); (M.d.-T.-R.); (G.P.-M.)
| | - David Moreno-Martinez
- Department of Internal Medicine, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (M.C.-G.); (D.M.-M.)
- Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.T.-S.); (M.d.-T.-R.); (G.P.-M.)
- Lysosomal Storage Disorders Unit, Royal Free Hospital NHS Foundation Trust and University College London, London WC1E 6BT, UK
| | - Sara Lucas-Del-Pozo
- Neurodegenerative Diseases Laboratory, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.L.-D.-P.); (J.H.-V.); (M.A.-B.)
- Department of Neurology, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Marc Moltó-Abad
- Functional Validation & Preclinical Research, Drug Delivery & Targeting Group, CIBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain;
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
| | - Ariadna Tigri-Santiña
- Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.T.-S.); (M.d.-T.-R.); (G.P.-M.)
| | - Irene Agraz-Pamplona
- Department of Nephrology, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| | - Jose F Rodriguez-Palomares
- Department of Cardiology, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| | - Jorge Hernández-Vara
- Neurodegenerative Diseases Laboratory, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.L.-D.-P.); (J.H.-V.); (M.A.-B.)
- Department of Neurology, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Mar Armengol-Bellapart
- Neurodegenerative Diseases Laboratory, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.L.-D.-P.); (J.H.-V.); (M.A.-B.)
- Department of Neurology, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Mireia del-Toro-Riera
- Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.T.-S.); (M.d.-T.-R.); (G.P.-M.)
- Department of Pediatric Neurology, Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, 08035 Barcelona, Spain
| | - Guillem Pintos-Morell
- Unit of Hereditary Metabolic Disorders, Vall d’Hebron Barcelona Hospital Campus, Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (A.T.-S.); (M.d.-T.-R.); (G.P.-M.)
- Functional Validation & Preclinical Research, Drug Delivery & Targeting Group, CIBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035 Barcelona, Spain;
| |
Collapse
|
43
|
The Cardiovascular Phenotype in Fabry Disease: New Findings in the Research Field. Int J Mol Sci 2021; 22:ijms22031331. [PMID: 33572752 PMCID: PMC7865937 DOI: 10.3390/ijms22031331] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Fabry disease (FD) is a lysosomal storage disorder, depending on defects in alpha-galactosidase A (GAL) activity. At the clinical level, FD shows a high phenotype variability. Among them, cardiovascular dysfunction is often recurrent or, in some cases, is the sole symptom (cardiac variant) representing the leading cause of death in Fabry patients. The existing therapies, besides specific symptomatic treatments, are mainly based on the restoration of GAL activity. Indeed, mutations of the galactosidase alpha gene (GLA) cause a reduction or lack of GAL activity leading to globotriaosylceramide (Gb3) accumulation in several organs. However, several other mechanisms are involved in FD’s development and progression that could become useful targets for therapeutics. This review discusses FD’s cardiovascular phenotype and the last findings on molecular mechanisms that accelerate cardiac cell damage.
Collapse
|
44
|
Rosa NS, Bento JCDB, Caparbo VDF, Pereira RMR. Increased Serum Interleukin-6 and Tumor Necrosis Factor Alpha Levels in Fabry Disease: Correlation with Disease Burden. Clinics (Sao Paulo) 2021; 76:e2643. [PMID: 34287477 PMCID: PMC8266164 DOI: 10.6061/clinics/2021/e2643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/01/2021] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVES Fabry disease (FD) is an X-linked lysosomal disease caused by variants of the GLA gene; the formation of defective alpha-galactosidase A contributes to the accumulation of substrates in several organs. Chronic inflammation is thought to contribute to organ damage in FD patients. METHODS In total, 36 classic FD patients (15 men/21 women) and 25 healthy controls (20 men/8 women) were assessed. The Mainz Severity Score Index (MSSI) was established after conducting interviews with the patients and chart review. Serum IL-6, IL-1β, and TNF-α levels were evaluated in both groups. RESULTS The mean age (years) for FD patients was 43.1±15.4 and that for the controls was 47.4±12.2 (p>0.05). Twenty-two patients (59.5%) were treated with enzyme replacement therapy (ERT). Serum IL-6 and TNF-α levels were significantly higher in FD patients than in the controls. Patients treated with ERT had higher serum IL-6 and TNF-α levels than those not treated with ERT. There was no difference in the serum IL-1β levels between patients treated with ERT and those who were not. The MSSI scores in the patients were correlated with serum levels of IL-6 (r=0.60, p<0.001) and TNF-α (r=0.45, p<0.001). CONCLUSION FD was associated with elevated serum levels of IL-6 and TNF-α in this cohort. The FD patients treated with ERT, particularly, women, exhibited higher levels of serum IL-6 and TNF-α than those not treated with ERT; the serum IL-6 and TNF-α levels were correlated with the MSSI scores reflecting greater disease burden.
Collapse
Affiliation(s)
- Nilton Salles Rosa
- Divisao de Reumatologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | | | - Valéria de Falco Caparbo
- Divisao de Reumatologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | |
Collapse
|
45
|
Zhang H, Viveiros A, Nikhanj A, Nguyen Q, Wang K, Wang W, Freed DH, Mullen JC, MacArthur R, Kim DH, Tymchak W, Sergi CM, Kassiri Z, Wang S, Oudit GY. The Human Explanted Heart Program: A translational bridge for cardiovascular medicine. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165995. [PMID: 33141063 PMCID: PMC7581399 DOI: 10.1016/j.bbadis.2020.165995] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/17/2022]
Abstract
The progression of cardiovascular research is often impeded by the lack of reliable disease models that fully recapitulate the pathogenesis in humans. These limitations apply to both in vitro models such as cell-based cultures and in vivo animal models which invariably are limited to simulate the complexity of cardiovascular disease in humans. Implementing human heart tissue in cardiovascular research complements our research strategy using preclinical models. We established the Human Explanted Heart Program (HELP) which integrates clinical, tissue and molecular phenotyping thereby providing a comprehensive evaluation into human heart disease. Our collection and storage of biospecimens allow them to retain key pathogenic findings while providing novel insights into human heart failure. The use of human non-failing control explanted hearts provides a valuable comparison group for the diseased explanted hearts. Using HELP we have been able to create a tissue repository which have been used for genetic, molecular, cellular, and histological studies. This review describes the process of collection and use of explanted human heart specimens encompassing a spectrum of pediatric and adult heart diseases, while highlighting the role of these invaluable specimens in translational research. Furthermore, we highlight the efficient procurement and bio-preservation approaches ensuring analytical quality of heart specimens acquired in the context of heart donation and transplantation.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anish Nikhanj
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Quynh Nguyen
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kaiming Wang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wei Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Darren H Freed
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John C Mullen
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacArthur
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel H Kim
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Wayne Tymchak
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Consolato M Sergi
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Anatomical Pathology, Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Cardiac Surgery, Department of Surgery, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
46
|
Laney DA, Germain DP, Oliveira JP, Burlina AP, Cabrera GH, Hong GR, Hopkin RJ, Niu DM, Thomas M, Trimarchi H, Wilcox WR, Politei JM, Ortiz A. Fabry disease and COVID-19: international expert recommendations for management based on real-world experience. Clin Kidney J 2020; 13:913-925. [PMID: 33391734 PMCID: PMC7769541 DOI: 10.1093/ckj/sfaa227] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Indexed: 12/11/2022] Open
Abstract
The rapid spread of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 has raised questions about Fabry disease (FD) as an independent risk factor for severe COVID-19 symptoms. Available real-world data on 22 patients from an international group of healthcare providers reveals that most patients with FD experience mild-to-moderate COVID-19 symptoms with an additional complication of Fabry pain crises and transient worsening of kidney function in some cases; however, two patients over the age of 55 years with renal or cardiac disease experienced critical COVID-19 complications. These outcomes support the theory that pre-existent tissue injury and inflammation may predispose patients with more advanced FD to a more severe course of COVID-19, while less advanced FD patients do not appear to be more susceptible than the general population. Given these observed risk factors, it is best to reinforce all recommended safety precautions for individuals with advanced FD. Diagnosis of FD should not preclude providing full therapeutic and organ support as needed for patients with FD and severe or critical COVID-19, although a FD-specific safety profile review should always be conducted prior to initiating COVID-19-specific therapies. Continued specific FD therapy with enzyme replacement therapy, chaperone therapy, dialysis, renin-angiotensin blockers or participation to clinical trials during the pandemic is recommended as FD progression will only increase susceptibility to infection. In order to compile outcome data and inform best practices, an international registry for patients affected by Fabry and infected by COVID-19 should be established.
Collapse
Affiliation(s)
- Dawn A Laney
- Division of Medical Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Dominique P Germain
- Division of Medical Genetics, University of Versailles, AP-HP Paris Saclay University, Paris, France
| | - João Paulo Oliveira
- Centro Hospitalar Universitário de São João & Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | | | | | - Geu-Ru Hong
- Department of Cardiology, Yonsei University Severance Hospital, Seoul, Korea
| | - Robert J Hopkin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Dau-Ming Niu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Mark Thomas
- Department of Nephrology, Royal Perth Hospital, Perth, Australia
| | | | - William R Wilcox
- Division of Medical Genetics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Juan Manuel Politei
- Department of Neurology, Fundacion Para el Estudio de Enfermedades Neurometabolicas (FESEN), Buenos Aires, Argentina
| | - Alberto Ortiz
- Unidad de Dialisis, IIS-Fundacion Jimenez Diaz, School of Medicine, UAM, IRSIN and REDINREN, Madrid, Spain
| |
Collapse
|
47
|
Simonetta I, Tuttolomondo A, Daidone M, Pinto A. Biomarkers in Anderson-Fabry Disease. Int J Mol Sci 2020; 21:ijms21218080. [PMID: 33138098 PMCID: PMC7662984 DOI: 10.3390/ijms21218080] [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: 09/27/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022] Open
Abstract
Fabry disease is a rare lysosomal storage disorder caused by a deficiency of α-galactosidase A, resulting in multisystemic involvement. Lyso-Gb3 (globotriaosylsphingosine), the deacylated form of Gb3, is currently measured in plasma as a biomarker of classic Fabry disease. Intensive research of biomarkers has been conducted over the years, in order to detect novel markers that may potentially be used in clinical practice as a screening tool, in the context of the diagnostic process and as an indicator of response to treatment. An interesting field of application of such biomarkers is the management of female heterozygotes who present difficulty in predictable clinical progression. This review aims to summarise the current evidence and knowledge about general and specific markers that are actually measured in subjects with confirmed or suspected Fabry disease; moreover, we report potential novel markers such as microRNAs. Recent proteomic or metabolomic studies are in progress bringing out plasma proteome profiles in Fabry patients: this assessment may be useful to characterize molecular pathology of the disease, to improve diagnostic process, and to monitor response to treatment. The management of Fabry disease may be improved by the identification of biomarkers that reflect clinical course, severity, and the progression of the disease.
Collapse
|
48
|
Belarbi K, Cuvelier E, Bonte MA, Desplanque M, Gressier B, Devos D, Chartier-Harlin MC. Glycosphingolipids and neuroinflammation in Parkinson's disease. Mol Neurodegener 2020; 15:59. [PMID: 33069254 PMCID: PMC7568394 DOI: 10.1186/s13024-020-00408-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by the loss of dopaminergic neurons of the nigrostriatal pathway and the formation of neuronal inclusions known as Lewy bodies. Chronic neuroinflammation, another hallmark of the disease, is thought to play an important role in the neurodegenerative process. Glycosphingolipids are a well-defined subclass of lipids that regulate crucial aspects of the brain function and recently emerged as potent regulators of the inflammatory process. Deregulation in glycosphingolipid metabolism has been reported in Parkinson's disease. However, the interrelationship between glycosphingolipids and neuroinflammation in Parkinson's disease is not well known. This review provides a thorough overview of the links between glycosphingolipid metabolism and immune-mediated mechanisms involved in neuroinflammation in Parkinson's disease. After a brief presentation of the metabolism and function of glycosphingolipids in the brain, it summarizes the evidences supporting that glycosphingolipids (i.e. glucosylceramides or specific gangliosides) are deregulated in Parkinson's disease. Then, the implications of these deregulations for neuroinflammation, based on data from human inherited lysosomal glycosphingolipid storage disorders and gene-engineered animal studies are outlined. Finally, the key molecular mechanisms by which glycosphingolipids could control neuroinflammation in Parkinson's disease are highlighted. These include inflammasome activation and secretion of pro-inflammatory cytokines, altered calcium homeostasis, changes in the blood-brain barrier permeability, recruitment of peripheral immune cells or production of autoantibodies.
Collapse
Affiliation(s)
- Karim Belarbi
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Elodie Cuvelier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Marie-Amandine Bonte
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
| | - Mazarine Desplanque
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - Bernard Gressier
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie de la Faculté de Pharmacie, Univ. Lille, Lille, France
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, 1 Place de Verdun, 59006 Lille, France
- Département de Pharmacologie Médicale, I-SITE ULNE, LiCEND, Lille, France
| | | |
Collapse
|
49
|
Assessment of plasma lyso-Gb 3 for clinical monitoring of treatment response in migalastat-treated patients with Fabry disease. Genet Med 2020; 23:192-201. [PMID: 32994552 PMCID: PMC7790748 DOI: 10.1038/s41436-020-00968-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 11/09/2022] Open
Abstract
PURPOSE To assess the utility of globotriaosylsphingosine (lyso-Gb3) for clinical monitoring of treatment response in patients with Fabry disease receiving migalastat. METHODS A post hoc analysis evaluated data from 97 treatment-naive and enzyme replacement therapy (ERT)-experienced patients with migalastat-amenable GLA variants from FACETS (NCT00925301) and ATTRACT (NCT01218659) and subsequent open-label extension studies. The relationship between plasma lyso-Gb3 and measures of Fabry disease progression (left ventricular mass index [LVMi], estimated glomerular filtration rate [eGFR], and pain) and the relationship between lyso-Gb3 and incidence of Fabry-associated clinical events (FACEs) were assessed in both groups. The relationship between changes in lyso-Gb3 and kidney interstitial capillary (KIC) globotriaosylceramide (Gb3) inclusions was assessed in treatment-naive patients. RESULTS No significant correlations were identified between changes in lyso-Gb3 and changes in LVMi, eGFR, or pain. Neither baseline lyso-Gb3 levels nor the rate of change in lyso-Gb3 levels during treatment predicted FACE occurrences in all patients or those receiving migalastat for ≥24 months. Changes in lyso-Gb3 correlated with changes in KIC Gb3 inclusions in treatment-naive patients. CONCLUSIONS Although used as a pharmacodynamic biomarker in research and clinical studies, plasma lyso-Gb3 may not be a suitable biomarker for monitoring treatment response in migalastat-treated patients.
Collapse
|
50
|
van der Veen SJ, Hollak CEM, van Kuilenburg ABP, Langeveld M. Developments in the treatment of Fabry disease. J Inherit Metab Dis 2020; 43:908-921. [PMID: 32083331 PMCID: PMC7540041 DOI: 10.1002/jimd.12228] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 12/19/2022]
Abstract
Enzyme replacement therapy (ERT) with recombinant α-galactosidase A (r-αGAL A) for the treatment of Fabry disease has been available for over 15 years. Long-term treatment may slow down disease progression, but cardiac, renal, and cerebral complications still develop in most patients. In addition, lifelong intravenous treatment is burdensome. Therefore, several new treatment approaches have been explored over the past decade. Chaperone therapy (Migalastat; 1-deoxygalactonojirimycin) is the only other currently approved therapy for Fabry disease. This oral small molecule aims to improve enzyme activity of mutated α-galactosidase A and can only be used in patients with specific mutations. Treatments currently under evaluation in (pre)clinical trials are second generation enzyme replacement therapies (Pegunigalsidase-alfa, Moss-aGal), substrate reduction therapies (Venglustat and Lucerastat), mRNA- and gene-based therapy. This review summarises the knowledge on currently available and potential future options for the treatment of Fabry disease.
Collapse
Affiliation(s)
- Sanne J. van der Veen
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAZAmsterdamThe Netherlands
| | - Carla E. M. Hollak
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAZAmsterdamThe Netherlands
| | - André B. P. van Kuilenburg
- Department of Clinical Chemistry, Gastroenterology & MetabolismAmsterdam UMC, University of AmsterdamAZAmsterdamThe Netherlands
| | - Mirjam Langeveld
- Department of Endocrinology and MetabolismAmsterdam UMC, University of AmsterdamAZAmsterdamThe Netherlands
| |
Collapse
|