1
|
Smith FM, Kosman DJ. Brain microvascular endothelial cells differentiated from a Friedreich's Ataxia patient iPSC are deficient in tight junction protein expression and paracellularly permeable. Front Mol Neurosci 2025; 18:1511388. [PMID: 40303283 PMCID: PMC12037585 DOI: 10.3389/fnmol.2025.1511388] [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: 10/14/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Friedreich's Ataxia (FA) is a rare, inherited ataxia resulting from GAA triplet expansions in the first intron of the Frataxin (FXN) gene, which encodes a mitochondrial protein involved in the incorporation of iron into iron-sulfur clusters. We previously identified decreased levels of F-actin and tight junction (TJ) proteins, which coincided with paracellular permeability in an FXN shRNA-mediated knockdown immortalized human brain microvascular endothelial cell (BMVEC) model. This premise is underexplored in the FA literature, prompting us to confirm these findings using a patient-derived iPSC model. One line each of FA patient iPSCs and age- and sex-matched apparently healthy iPSCs were differentiated into BMVEC-like cells. We quantified actin glutathionylation, F-actin abundance, TJ expression and organization, and barrier integrity. In the absence of dysregulated F-actin organization, FA iBMVEC exhibited a loss of 50% ZO-1, 63% Occludin, and 19% Claudin-5 protein expression, along with a disruption in the bi-cellular organization of the latter two proteins. Functionally, this correlated with barrier hyperpermeability, delayed barrier maturation, and increased flux of the fluorescent tracer Lucifer Yellow. These data indicate that decreased barrier integrity is a pathophysiological phenotype of FA brain microvascular endothelial cells. Clinically, this may represent a targetable pathway to reduce brain iron accumulation, neuroinflammation, and neurodegeneration profiles in FA. Additionally, an investigation into other barrier systems, such as the blood-nerve barrier, blood-CSF barrier, or cardiac vasculature, may provide insights into the extra-neural symptoms experienced by FA patients.
Collapse
Affiliation(s)
| | - Daniel J. Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
2
|
Baile MG, Jones J, Sahr N, Shankar G. Nomlabofusp, a Fusion Protein of Human Frataxin and a Cell Penetrant Peptide, Delivers Mature and Functional Frataxin into Mitochondria. AAPS J 2025; 27:68. [PMID: 40140196 DOI: 10.1208/s12248-025-01054-5] [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: 01/29/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Friedreich's ataxia is a rare, progressive, genetic disorder, the root cause of which is a significant deficiency in the mitochondrial protein frataxin. Frataxin is ubiquitously expressed, but its deficiency results in a variety of debilitating symptoms, with disease severity, rate of progression and age of onset inversely correlating with tissue frataxin levels. Nomlabofusp is a novel cell penetrant peptide based recombinant fusion protein designed to enter cells and deliver human FXN into the mitochondria. Using immunofluorescence staining and western blot we show that frataxin delivered by nomlabofusp is detected in the mitochondria of H9c2 and SH-SY5Y cells. Also in these cells, and in C2C12 and HEK293 cells, we demonstrate the presence of mature frataxin after nomlabofusp exposure. Finally, using buccal swab tissue samples taken from study subjects in a Phase 1 clinical trial who received nomlabofusp, we show increases in mature frataxin levels along with marked changes in gene expression post-administration suggesting intracellular pharmacodynamic activity. Together, these results demonstrate that nomlabofusp enters the cell and localizes to the mitochondria, releasing mature frataxin that appears to be biologically active and support the use of nomlabofusp as a potential treatment for patients with Friedreich's ataxia.
Collapse
Affiliation(s)
- Matthew G Baile
- Discovery Laboratory, Larimar Therapeutics Inc., King of Prussia, PA, USA
| | - John Jones
- Discovery Laboratory, Larimar Therapeutics Inc., King of Prussia, PA, USA
| | - Natasha Sahr
- Statistics & Quantitative Sciences, Larimar Therapeutics Inc., Bala Cynwyd, PA, USA
| | - Gopi Shankar
- Corporate Office, Larimar Therapeutics Inc., 3 Bala Plaza, Suite 506, Bala Cynwyd, PA, 19004, USA.
| |
Collapse
|
3
|
Lynch DR, Shen M, Wilson RB. Friedreich ataxia: what can we learn from non-GAA repeat mutations? Neurodegener Dis Manag 2025; 15:17-26. [PMID: 39810561 PMCID: PMC11938963 DOI: 10.1080/17582024.2025.2452147] [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: 09/23/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Friedreich ataxia (FRDA) is a slowly progressive neurological disease resulting from decreased levels of the protein frataxin, a small mitochondrial protein that facilitates the synthesis of iron-sulfur clusters in the mitochondrion. It is caused by GAA (guanine-adenine-adenine) repeat expansions in the FXN gene in 96% of patients, with 4% of patients carrying other mutations (missense, nonsense, deletion) in the FXN gene. Compound heterozygote patients with one expanded GAA allele and a non-GAA repeat mutation can have subtle differences in phenotype from typical FRDA, including, in patients with selected missense mutations, both more severe features and less severe features in the same patient. In this review, we propose explanations for such phenotypes based on the potential for activities of frataxin other than enhancement of iron-sulfur cluster synthesis, as well as crucial future experiments for fully understanding the role of frataxin in cells.
Collapse
Affiliation(s)
- David R. Lynch
- Friedreich Ataxia Program, Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - M. Shen
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
| | - Robert B. Wilson
- Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
4
|
Scarabino D, Veneziano L, Nethisinghe S, Mantuano E, Fiore A, Granata G, Solanky N, Zanni G, Cavalcanti F, Corbo RM, Giunti P. Unusual Age-Dependent Behavior of Leukocytes Telomere Length in Friedreich's Ataxia. Mov Disord 2024; 39:2058-2066. [PMID: 39235665 DOI: 10.1002/mds.29976] [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: 03/27/2024] [Revised: 06/15/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by an expanded GAA repeat in the first intron of the FXN gene. OBJECTIVE The aim of this study was to analyze leukocyte telomeres length (LTL) in FRDA to verify the possible relationships between LTL and disease progression. We investigated LTL in a cohort of FRDA biallelic patients (n = 61), heterozygous (n = 29), and age-matched healthy subjects (n = 87). METHODS LTL was measured by real-time polymerase chain reaction quantitative analysis (qPCR). RESULTS The results showed that before 35 years of age, leukocyte telomeres were longer in patients than in controls, whereas the reverse applies in patients above 36 years of age. Interestingly, LTL was greater than controls at any age in heterozygous subjects. This picture mirrors what has been previously observed in vitro in FRDA cultured fibroblasts, showing significantly longer telomeres at early passages because of activation of an alternative lengthening of telomeres (ALT)-like mechanism, but showing accelerated telomere shortening as population doubling increases. GAA1 repeat length is positively correlated with the LTL and negatively correlated with the age at blood sampling. The relationship of LTL with clinical parameters (cardiomyopathy, diabetes, dependence on a wheelchair) was also analyzed. Significantly shorter leukocyte telomeres were associated with the presence of cardiomyopathy, but not with diabetes and the dependence on a wheelchair. CONCLUSIONS Overall, the present study indicates that telomere length analysis in FRDA may be a relevant biomarker for following the stages of the disease. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Daniela Scarabino
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| | - Liana Veneziano
- Institute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
- Human Functional Genomics Laboratory, IRCCS San Raffaele, Rome, Italy
| | - Suran Nethisinghe
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Elide Mantuano
- Institute of Translational Pharmacology, National Research Council (CNR), Rome, Italy
| | - Alessia Fiore
- Department of Biology and Biotechnology, La Sapienza University, Rome, Italy
| | - Giulia Granata
- Department of Biology and Biotechnology, La Sapienza University, Rome, Italy
| | - Nita Solanky
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Ginevra Zanni
- Unit of Muscular and Neurodegenerative Disorders, Unit of Developmental Neurology, Bambino Gesù Children's Research Hospital, IRCCS, Rome, Italy
| | - Francesca Cavalcanti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Mangone, Italy
| | - Rosa Maria Corbo
- Department of Biology and Biotechnology, La Sapienza University, Rome, Italy
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
5
|
Alemany-Perna B, Tamarit J, Cabiscol E, Delaspre F, Miguela A, Huertas-Pons JM, Quiroga-Varela A, Merchan Ruiz M, López Domínguez D, Ramió I Torrentà L, Genís D, Ros J. Calcitriol Treatment Is Safe and Increases Frataxin Levels in Friedreich Ataxia Patients. Mov Disord 2024; 39:1099-1108. [PMID: 38696306 DOI: 10.1002/mds.29808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 05/04/2024] Open
Abstract
BACKGROUND Calcitriol, the active form of vitamin D (also known as 1,25-dihydroxycholecalciferol), improves the phenotype and increases frataxin levels in cell models of Friedreich ataxia (FRDA). OBJECTIVES Based on these results, we aimed measuring the effects of a calcitriol dose of 0.25 mcg/24h in the neurological function and frataxin levels when administered to FRDA patients for a year. METHODS 20 FRDA patients where recluted and 15 patients completed the treatment for a year. Evaluations of neurological function changes (SARA scale, 9-HPT, 8-MWT, PATA test) and quality of life (Barthel Scale and Short Form (36) Health Survey [SF-36] quality of life questionnaire) were performed. Frataxin amounts were measured in isolated platelets obtained from these FRDA patients, from heterozygous FRDA carriers (relatives of the FA patients) and from non-heterozygous sex and age matched controls. RESULTS Although the patients did not experience any observable neurological improvement, there was a statistically significant increase in frataxin levels from initial values, 5.5 to 7.0 pg/μg after 12 months. Differences in frataxin levels referred to total protein levels were observed among sex- and age-matched controls (18.1 pg/μg), relative controls (10.1 pg/μg), and FRDA patients (5.7 pg/μg). The treatment was well tolerated by most patients, and only some of them experienced minor adverse effects at the beginning of the trial. CONCLUSIONS Calcitriol dosage used (0.25 mcg/24 h) is safe for FRDA patients, and it increases frataxin levels. We cannot rule out that higher doses administered longer could yield neurological benefits. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Berta Alemany-Perna
- Ataxia Unit, Neurology Service, ICS/IAS, Hospital Josep Trueta/Hospital Santa Caterina, Girona/Salt, Spain
- Department of Medical Sciences, University of Girona (UdG), Girona, Spain
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida (IRBLleida), Universitat de Lleida, Lleida, Spain
| | - Elisa Cabiscol
- Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida (IRBLleida), Universitat de Lleida, Lleida, Spain
| | - Fabien Delaspre
- Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida (IRBLleida), Universitat de Lleida, Lleida, Spain
| | - Albert Miguela
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Joana Maria Huertas-Pons
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Ana Quiroga-Varela
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Miguel Merchan Ruiz
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Daniel López Domínguez
- Ataxia Unit, Neurology Service, ICS/IAS, Hospital Josep Trueta/Hospital Santa Caterina, Girona/Salt, Spain
- Department of Medical Sciences, University of Girona (UdG), Girona, Spain
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Lluís Ramió I Torrentà
- Department of Medical Sciences, University of Girona (UdG), Girona, Spain
- Neurology Service, ICS/IAS, Hospital Josep Trueta/Hospital Santa Caterina, Girona/Salt, Neurodegeneration and Neuroinflammacion Group (IDIBGI), Girona/Salt, Spain
| | - David Genís
- Neurodegeneration and Neuroinflammacion Group, Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, Institut de Recerca Biomèdica de Lleida (IRBLleida), Universitat de Lleida, Lleida, Spain
| |
Collapse
|
6
|
Dong YN, Mercado-Ayón E, Coulman J, Flatley L, Ngaba LV, Adeshina MW, Lynch DR. The Regulation of the Disease-Causing Gene FXN. Cells 2024; 13:1040. [PMID: 38920668 PMCID: PMC11202134 DOI: 10.3390/cells13121040] [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: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Friedreich's ataxia (FRDA) is a progressive neurodegenerative disease caused in almost all patients by expanded guanine-adenine-adenine (GAA) trinucleotide repeats within intron 1 of the FXN gene. This results in a relative deficiency of frataxin, a small nucleus-encoded mitochondrial protein crucial for iron-sulfur cluster biogenesis. Currently, there is only one medication, omaveloxolone, available for FRDA patients, and it is limited to patients 16 years of age and older. This necessitates the development of new medications. Frataxin restoration is one of the main strategies in potential treatment options as it addresses the root cause of the disease. Comprehending the control of frataxin at the transcriptional, post-transcriptional, and post-translational stages could offer potential therapeutic approaches for addressing the illness. This review aims to provide a general overview of the regulation of frataxin and its implications for a possible therapeutic treatment of FRDA.
Collapse
Affiliation(s)
- Yi Na Dong
- Departments of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Jennifer Coulman
- Departments of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Liam Flatley
- The Wharton School, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lucie Vanessa Ngaba
- Departments of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Miniat W. Adeshina
- Departments of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R. Lynch
- Departments of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Shen MM, Rummey C, Lynch DR. Phenotypic variation of FXN compound heterozygotes in a Friedreich ataxia cohort. Ann Clin Transl Neurol 2024; 11:1110-1121. [PMID: 38396238 PMCID: PMC11093247 DOI: 10.1002/acn3.52027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
OBJECTIVE Most individuals with Friedreich ataxia (FRDA) have homozygous GAA triplet repeat expansions in the FXN gene, correlating with a typical phenotype of ataxia and cardiomyopathy. A minority are compound heterozygotes carrying a GAA expansion on one allele and a mutation on the other. The study aim was to examine phenotypic variation among compound heterozygotes. METHODS Data on FXN mutations were obtained from the Friedreich Ataxia Clinical Outcome Measures Study (FA-COMS). We compared clinical features in a single-site FA-COMS cohort of 51 compound heterozygous and 358 homozygous patients, including quantitative measures of cardiac, neurologic, and visual disease progression. RESULTS Non-GAA repeat mutations were associated with reduced cardiac disease, and patients with minimal/no function mutations otherwise had a typical FRDA phenotype but with significantly more severe progression. The partial function mutation group was characterized by relative sparing of bulbar and upper limb function, as well as particularly low cardiac involvement. Other clinical features in this group, including optic atrophy and diabetes mellitus, varied widely depending on the specific type of partial function mutation. INTERPRETATION These data support that the typical FRDA phenotype is driven by frataxin deficiency, especially severe in compound heterozygotes with minimal/no function mutations, whereas the heterogeneous presentations of those with partial function mutations may indicate other contributing factors to FRDA pathogenesis.
Collapse
Affiliation(s)
- Megan M. Shen
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| | | | - David R. Lynch
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine, University of Pennsylvania.PhiladelphiaPennsylvaniaUSA
| |
Collapse
|
8
|
Gunther K, Lynch DR. Pharmacotherapeutic strategies for Friedreich Ataxia: a review of the available data. Expert Opin Pharmacother 2024; 25:529-539. [PMID: 38622054 DOI: 10.1080/14656566.2024.2343782] [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: 02/05/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Friedreich ataxia (FRDA) is a rare autosomal recessive disease, marked by loss of coordination as well as impaired neurological, endocrine, orthopedic, and cardiac function. There are many symptomatic medications for FRDA, and many clinical trials have been performed, but only one FDA-approved medication exists. AREAS COVERED The relative absence of the frataxin protein (FXN) in FRDA causes mitochondrial dysfunction, resulting in clinical manifestations. Currently, the only approved treatment for FRDA is an Nrf2 activator called omaveloxolone (Skyclarys). Patients with FRDA also rely on various symptomatic medications for treatment. Because there is only one approved medication for FRDA, clinical trials continue to advance in FRDA. Although some trials have not met their endpoints, many current and upcoming clinical trials provide exciting possibilities for the treatment of FRDA. EXPERT OPINION The approval of omaveloxolone provides a major advance in FRDA therapeutics. Although well tolerated, it is not curative. Reversal of deficient frataxin levels with gene therapy, protein replacement, or epigenetic approaches provides the most likely prospect for enduring, disease-modifying therapy in the future.
Collapse
Affiliation(s)
- Katherine Gunther
- Friedreich Ataxia Program, Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David R Lynch
- Friedreich Ataxia Program, Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
9
|
Lynch DR, Rojsajjakul T, Subramony SH, Perlman SL, Keita M, Mesaros C, Blair IA. Frataxin analysis using triple quadrupole mass spectrometry: application to a large heterogeneous clinical cohort. J Neurol 2024; 271:1844-1849. [PMID: 38063871 DOI: 10.1007/s00415-023-12118-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Friedreich ataxia is a progressive multisystem disorder caused by deficiency of the protein frataxin; a small mitochondrial protein involved in iron sulfur cluster synthesis. Two types of frataxin exist: FXN-M, found in most cells, and FXN-E, found almost exclusively in red blood cells. Treatments in clinical trials include frataxin restoration by gene therapy, protein replacement, and epigenetic therapies, all of which necessitate sensitive assays for assessing frataxin levels. METHODS In the present study, we have used a triple quadrupole mass spectrometry-based assay to examine the features of both types of frataxin levels in blood in a large heterogenous cohort of 106 patients with FRDA. RESULTS Frataxin levels (FXN-E and FXN M) were predicted by GAA repeat length in regression models (R2 values = 0.51 and 0.27, respectively), and conversely frataxin levels predicted clinical status as determined by modified Friedreich Ataxia Rating scale scores and by disability status (R2 values = 0.13-0.16). There was no significant change in frataxin levels in individual subjects over time, and apart from start codon mutations, FXN-E and FXN-M levels were roughly equal. Accounting for hemoglobin levels in a smaller sub-cohort improved prediction of both FXN-E and FXN-M levels from R2 values of (0.3-0.38 to 0.20-0.51). CONCLUSION The present data show that assay of FXN-M and FXN-E levels in blood provides an appropriate biofluid for assessing their repletion in particular clinical contexts.
Collapse
Affiliation(s)
- David R Lynch
- Penn/CHOP Friedreich Ataxia Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, 502F Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA, 19104-4318, USA.
| | - Teerapat Rojsajjakul
- Penn/CHOP Friedreich Ataxia Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - S H Subramony
- Department of Neurology, University of Florida, Gainesville, FL, 32608, USA
| | - Susan L Perlman
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Medina Keita
- Penn/CHOP Friedreich Ataxia Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Clementina Mesaros
- Penn/CHOP Friedreich Ataxia Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian A Blair
- Penn/CHOP Friedreich Ataxia Center of Excellence, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
10
|
Chang JC, Ryan MR, Stark MC, Liu S, Purushothaman P, Bolan F, Johnson CA, Champe M, Meng H, Lawlor MW, Halawani S, Ngaba LV, Lynch DR, Davis C, Gonzalo-Gil E, Lutz C, Urbinati F, Medicherla B, Fonck C. AAV8 gene therapy reverses cardiac pathology and prevents early mortality in a mouse model of Friedreich's ataxia. Mol Ther Methods Clin Dev 2024; 32:101193. [PMID: 38352270 PMCID: PMC10862410 DOI: 10.1016/j.omtm.2024.101193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Friedreich's ataxia (FRDA) is an autosomal-recessive disorder primarily attributed to biallelic GAA repeat expansions that reduce expression of the mitochondrial protein frataxin (FXN). FRDA is characterized by progressive neurodegeneration, with many patients developing cardiomyopathy that progresses to heart failure and death. The potential to reverse or prevent progression of the cardiac phenotype of FRDA was investigated in a mouse model of FRDA, using an adeno-associated viral vector (AAV8) containing the coding sequence of the FXN gene. The Fxnflox/null::MCK-Cre conditional knockout mouse (FXN-MCK) has an FXN gene ablation that prevents FXN expression in cardiac and skeletal muscle, leading to cardiac insufficiency, weight loss, and morbidity. FXN-MCK mice received a single intravenous injection of an AAV8 vector containing human (hFXN) or mouse (mFXN) FXN genes under the control of a phosphoglycerate kinase promoter. Compared to vehicle-treated FXN-MCK control mice, AAV-treated FXN-MCK mice displayed increases in body weight, reversal of cardiac deficits, and increases in survival without apparent toxicity in the heart or liver for up to 12 weeks postdose. FXN protein expression in heart tissue was detected in a dose-dependent manner, exhibiting wide distribution throughout the heart similar to wild type, but more speckled. These results support an AAV8-based approach to treat FRDA-associated cardiomyopathy.
Collapse
Affiliation(s)
- Joshua C. Chang
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Molly R. Ryan
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Marie C. Stark
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Su Liu
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | | | - Fria Bolan
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | | | - Mark Champe
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Hui Meng
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
- Medical College of Wisconsin, Department of Pathology and Laboratory Medicine, Milwaukee, WI 53226, USA
| | - Michael W. Lawlor
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
- Medical College of Wisconsin, Department of Pathology and Laboratory Medicine, Milwaukee, WI 53226, USA
| | - Sarah Halawani
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lucie V. Ngaba
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R. Lynch
- Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | - Fabrizia Urbinati
- Formerly of Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Bala Medicherla
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| | - Carlos Fonck
- Astellas Gene Therapies, Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
11
|
Lynch DR, Goldsberry A, Rummey C, Farmer J, Boesch S, Delatycki MB, Giunti P, Hoyle JC, Mariotti C, Mathews KD, Nachbauer W, Perlman S, Subramony S, Wilmot G, Zesiewicz T, Weissfeld L, Meyer C. Propensity matched comparison of omaveloxolone treatment to Friedreich ataxia natural history data. Ann Clin Transl Neurol 2024; 11:4-16. [PMID: 37691319 PMCID: PMC10791025 DOI: 10.1002/acn3.51897] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023] Open
Abstract
OBJECTIVE The natural history of Friedreich ataxia is being investigated in a multi-center longitudinal study designated the Friedreich ataxia Clinical Outcome Measures Study (FACOMS). To understand the utility of this study in analysis of clinical trials, we performed a propensity-matched comparison of data from the open-label MOXIe extension (omaveloxolone) to that from FACOMS. METHODS MOXIe extension patients were matched to FACOMS patients using logistic regression to estimate propensity scores based on multiple covariates: sex, baseline age, age of onset, baseline modified Friedreich Ataxia Rating scale (mFARS) score, and baseline gait score. The change from baseline in mFARS at Year 3 for the MOXIe extension patients compared to the matched FACOMS patients was analyzed as the primary efficacy endpoint using mixed model repeated measures analysis. RESULTS Data from the MOXIe extension show that omaveloxolone provided persistent benefit over 3 years when compared to an untreated, matched cohort from FACOMS. At each year, in all analysis populations, patients in the MOXIe extension experienced a smaller change from baseline in mFARS score than matched FACOMS patients. In the primary pooled population (136 patients in each group) by Year 3, patients in the FACOMS matched set progressed 6.6 points whereas patients treated with omaveloxolone in MOXIe extension progressed 3 points (difference = -3.6; nominal p value = 0.0001). INTERPRETATION These results suggest a meaningful slowing of Friedreich ataxia progression with omaveloxolone, and consequently detail how propensity-matched analysis may contribute to understanding of effects of therapeutic agents. This demonstrates the direct value of natural history studies in clinical trial evaluations.
Collapse
Affiliation(s)
- David R. Lynch
- Departments of Pediatrics and NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | | | - Jennifer Farmer
- Friedreich Ataxia Research AllianceDowningtownPennsylvaniaUSA
| | - Sylvia Boesch
- Department of NeurologyMedical University InnsbruckInnsbruckAustria
| | - Martin B. Delatycki
- Victorian Clinical Genetics ServicesMurdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Paola Giunti
- University College London HospitalBloomsburyLondonUK
| | - J. Chad Hoyle
- Department of NeurologyOhio State University College of MedicineColumbusOhioUSA
| | | | - Katherine D. Mathews
- Department of PediatricsUniversity of Iowa Carver College of MedicineIowa CityIowaUSA
| | | | - Susan Perlman
- Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - S.H. Subramony
- Department of Neurology, McKnight Brain InstituteUniversity of Florida Health SystemGainesvilleFloridaUSA
| | - George Wilmot
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Theresa Zesiewicz
- Department of NeurologyUniversity of South Florida Ataxia Research CenterTampaFloridaUSA
| | | | | |
Collapse
|
12
|
Doni D, Cavion F, Bortolus M, Baschiera E, Muccioli S, Tombesi G, d'Ettorre F, Ottaviani D, Marchesan E, Leanza L, Greggio E, Ziviani E, Russo A, Bellin M, Sartori G, Carbonera D, Salviati L, Costantini P. Human frataxin, the Friedreich ataxia deficient protein, interacts with mitochondrial respiratory chain. Cell Death Dis 2023; 14:805. [PMID: 38062036 PMCID: PMC10703789 DOI: 10.1038/s41419-023-06320-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023]
Abstract
Friedreich ataxia (FRDA) is a rare, inherited neurodegenerative disease caused by an expanded GAA repeat in the first intron of the FXN gene, leading to transcriptional silencing and reduced expression of frataxin. Frataxin participates in the mitochondrial assembly of FeS clusters, redox cofactors of the respiratory complexes I, II and III. To date it is still unclear how frataxin deficiency culminates in the decrease of bioenergetics efficiency in FRDA patients' cells. We previously demonstrated that in healthy cells frataxin is closely attached to the mitochondrial cristae, which contain both the FeS cluster assembly machinery and the respiratory chain complexes, whereas in FRDA patients' cells with impaired respiration the residual frataxin is largely displaced in the matrix. To gain novel insights into the function of frataxin in the mitochondrial pathophysiology, and in the upstream metabolic defects leading to FRDA disease onset and progression, here we explored the potential interaction of frataxin with the FeS cluster-containing respiratory complexes I, II and III. Using healthy cells and different FRDA cellular models we found that frataxin interacts with these three respiratory complexes. Furthermore, by EPR spectroscopy, we observed that in mitochondria from FRDA patients' cells the decreased level of frataxin specifically affects the FeS cluster content of complex I. Remarkably, we also found that the frataxin-like protein Nqo15 from T. thermophilus complex I ameliorates the mitochondrial respiratory phenotype when expressed in FRDA patient's cells. Our data point to a structural and functional interaction of frataxin with complex I and open a perspective to explore therapeutic rationales for FRDA targeted to this respiratory complex.
Collapse
Affiliation(s)
- Davide Doni
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Federica Cavion
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Marco Bortolus
- Department of Chemical Sciences, University of Padova, 35131, Padova, Italy
| | - Elisa Baschiera
- Clinical Genetics Unit, Department of Women's and Children Health, University of Padova, 35128, Padova, Italy
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, 35127, Padova, Italy
| | - Silvia Muccioli
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Giulia Tombesi
- Department of Biology, University of Padova, 35121, Padova, Italy
| | | | | | - Elena Marchesan
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, 35121, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Elena Ziviani
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Antonella Russo
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Milena Bellin
- Department of Biology, University of Padova, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, ZA, Leiden, The Netherlands
| | - Geppo Sartori
- Department of Biomedical Sciences, University of Padova, 35121, Padova, Italy
| | | | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children Health, University of Padova, 35128, Padova, Italy.
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, 35127, Padova, Italy.
| | - Paola Costantini
- Department of Biology, University of Padova, 35121, Padova, Italy.
| |
Collapse
|
13
|
Munoz-Zuluaga C, Gertz M, Yost-Bido M, Greco A, Gorman N, Chen A, Kooner V, Rosenberg JB, De BP, Kaminsky SM, Borczuk A, Ricart Arbona RJ, Martin HR, Monette S, Khanna R, Barth JA, Crystal RG, Sondhi D. Identification of Safe and Effective Intravenous Dose of AAVrh.10hFXN to Treat the Cardiac Manifestations of Friedreich's Ataxia. Hum Gene Ther 2023; 34:605-615. [PMID: 37166361 PMCID: PMC10354731 DOI: 10.1089/hum.2023.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/16/2023] [Indexed: 05/12/2023] Open
Abstract
Friedreich's ataxia (FA) is a life-threatening autosomal recessive disorder characterized by neurological and cardiac dysfunction. Arrhythmias and heart failure are the main cause of premature death. From prior studies in murine models of FA, adeno-associated virus encoding the normal human frataxin gene (AAVrh.10hFXN) effectively treated the cardiac manifestations of the disease. However, the therapeutic dose window is limited by high level of human frataxin (hFXN) gene expression associated with toxicity. As a therapeutic goal, since FA heterozygotes have no clinical manifestations of FA, we estimated the level of frataxin (FXN) necessary to convert the heart of a homozygote to that of a heterozygote. In noncardiac cells, FA heterozygotes have 30-80% of normal FXN levels (17.7-47.2 ng/mg, average 32.5 ng/mg) and FA homozygotes 2-30% normal levels (1.2-17.7 ng/mg, average 9.4 ng/mg). Therefore, an AAV vector would need to augment endogenous in an FA homozygote by >8.3 ng/mg. To determine the required dose of AAVrh.10hFXN, we administered 1.8 × 1011, 5.7 × 1011, or 1.8 × 1012 gc/kg of AAVrh.10hFXN intravenously (IV) to muscle creatine kinase (mck)-Cre conditional knockout Fxn mice, a cardiac and skeletal FXN knockout model. The minimally effective dose was 5.7 × 1011 gc/kg, resulting in cardiac hFXN levels of 6.1 ± 4.2 ng/mg and a mild (p < 0.01 compared with phosphate-buffered saline controls) improvement in mortality. A dose of 1.8 × 1012 gc/kg resulted in cardiac hFXN levels of 33.7 ± 6.4 ng/mg, a significant improvement in ejection fraction and fractional shortening (p < 0.05, both comparisons) and a 21.5% improvement in mortality (p < 0.001). To determine if the significantly effective dose of 1.8 × 1012 gc/kg could achieve human FA heterozygote levels in a large animal, this dose was administered IV to nonhuman primates. After 12 weeks, the vector-expressed FXN in the heart was 17.8 ± 4.9 ng/mg, comparable to the target human levels. These data identify both minimally and significantly effective therapeutic doses that are clinically relevant for the treatment of the cardiac manifestations of FA.
Collapse
Affiliation(s)
| | - Monica Gertz
- Department of Genetic Medicine, New York, New York, USA
| | | | | | | | - Alvin Chen
- Department of Genetic Medicine, New York, New York, USA
| | - Vikrum Kooner
- Department of Genetic Medicine, New York, New York, USA
| | | | - Bishnu P. De
- Department of Genetic Medicine, New York, New York, USA
| | | | - Alain Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, New York, USA
| | - Rodolfo J. Ricart Arbona
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Heather R. Martin
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Sebastien Monette
- Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | | | | | | | - Dolan Sondhi
- Department of Genetic Medicine, New York, New York, USA
| |
Collapse
|
14
|
Fil D, Conley RL, Zuberi AR, Lutz CM, Gemelli T, Napierala M, Napierala JS. Neurobehavioral deficits of mice expressing a low level of G127V mutant frataxin. Neurobiol Dis 2023; 177:105996. [PMID: 36638893 PMCID: PMC9901512 DOI: 10.1016/j.nbd.2023.105996] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/20/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin (FXN). Most FRDA patients are homozygous for large expansions of GAA repeats in intron 1 of FXN, while some are compound heterozygotes with an expanded GAA tract in one allele and a missense or nonsense mutation in the other. A missense mutation, changing a glycine to valine at position 130 (G130V), is prevalent among the clinical variants. We and others have demonstrated that levels of mature FXN protein in FRDA G130V samples are reduced below those detected in samples harboring homozygous repeat expansions. Little is known regarding expression and function of endogenous FXN-G130V protein due to lack of reagents and models that can distinguish the mutant FXN protein from the wild-type FXN produced from the GAA-expanded allele. We aimed to determine the effect of the G130V (murine G127V) mutation on Fxn expression and to define its multi-system impact in vivo. We used CRISPR/Cas9 to introduce the G127V missense mutation in the Fxn coding sequence and generated homozygous mice (FxnG127V/G127V). We also introduced the G127V mutation into a GAA repeat expansion FRDA mouse model (FxnGAA230/KO; KIKO) to generate a compound heterozygous strain (FxnG127V/GAA230). We performed neurobehavioral tests on cohorts of WT and Fxn mutant animals at three-month intervals for one year, and collected tissue samples to analyze molecular changes during that time. The endogenous Fxn G127V protein is detected at much lower levels in all tissues analyzed from FxnG127V/G127V mice compared to age and sex-matched WT mice without differences in Fxn transcript levels. FxnG127V/G127V mice are significantly smaller than WT counterparts, but perform similarly in most neurobehavioral tasks. RNA sequencing analysis revealed reduced expression of genes in oxidative phosphorylation and protein synthesis, underscoring the metabolic consequences in our mouse model expressing extremely low levels of Fxn. Results of these studies provide insight into the unique pathogenic mechanism of the FXN G130V mechanism and the tolerable limit of Fxn/FXN expression in vivo.
Collapse
Affiliation(s)
- Daniel Fil
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robbie L Conley
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Aamir R Zuberi
- Technology Evaluation and Development, JAX Center for Precision Genetics, Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Cathleen M Lutz
- The Rare and Orphan Disease Center, JAX Center for Precision Genetics, Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Terry Gemelli
- Department of Neurology, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jill S Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
15
|
Kadirvelu B, Gavriel C, Nageshwaran S, Chan JPK, Nethisinghe S, Athanasopoulos S, Ricotti V, Voit T, Giunti P, Festenstein R, Faisal AA. A wearable motion capture suit and machine learning predict disease progression in Friedreich's ataxia. Nat Med 2023; 29:86-94. [PMID: 36658420 PMCID: PMC9873563 DOI: 10.1038/s41591-022-02159-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/29/2022] [Indexed: 01/21/2023]
Abstract
Friedreich's ataxia (FA) is caused by a variant of the Frataxin (FXN) gene, leading to its downregulation and progressively impaired cardiac and neurological function. Current gold-standard clinical scales use simplistic behavioral assessments, which require 18- to 24-month-long trials to determine if therapies are beneficial. Here we captured full-body movement kinematics from patients with wearable sensors, enabling us to define digital behavioral features based on the data from nine FA patients (six females and three males) and nine age- and sex-matched controls, who performed the 8-m walk (8-MW) test and 9-hole peg test (9 HPT). We used machine learning to combine these features to longitudinally predict the clinical scores of the FA patients, and compared these with two standard clinical assessments, Spinocerebellar Ataxia Functional Index (SCAFI) and Scale for the Assessment and Rating of Ataxia (SARA). The digital behavioral features enabled longitudinal predictions of personal SARA and SCAFI scores 9 months into the future and were 1.7 and 4 times more precise than longitudinal predictions using only SARA and SCAFI scores, respectively. Unlike the two clinical scales, the digital behavioral features accurately predicted FXN gene expression levels for each FA patient in a cross-sectional manner. Our work demonstrates how data-derived wearable biomarkers can track personal disease trajectories and indicates the potential of such biomarkers for substantially reducing the duration or size of clinical trials testing disease-modifying therapies and for enabling behavioral transcriptomics.
Collapse
Affiliation(s)
- Balasundaram Kadirvelu
- Brain & Behaviour Lab, Department of Bioengineering, Imperial College London, London, UK
- Brain & Behaviour Lab, Department of Computing, Imperial College London, London, UK
| | - Constantinos Gavriel
- Brain & Behaviour Lab, Department of Bioengineering, Imperial College London, London, UK
- Brain & Behaviour Lab, Department of Computing, Imperial College London, London, UK
| | - Sathiji Nageshwaran
- Epigenetic Mechanisms and Disease Group, Department of Brain Sciences, Imperial College London, London, UK
| | - Jackson Ping Kei Chan
- Epigenetic Mechanisms and Disease Group, Department of Brain Sciences, Imperial College London, London, UK
| | - Suran Nethisinghe
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Stavros Athanasopoulos
- Epigenetic Mechanisms and Disease Group, Department of Brain Sciences, Imperial College London, London, UK
| | - Valeria Ricotti
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Paola Giunti
- Institute of Neurology, UCL, National Hospital for Neurology and Neurosurgery (UCLH), London, UK
| | - Richard Festenstein
- Epigenetic Mechanisms and Disease Group, Department of Brain Sciences, Imperial College London, London, UK
- Institute of Neurology, UCL, National Hospital for Neurology and Neurosurgery (UCLH), London, UK
- MRC London Institute of Medical Sciences, London, UK
| | - A Aldo Faisal
- Brain & Behaviour Lab, Department of Bioengineering, Imperial College London, London, UK.
- Brain & Behaviour Lab, Department of Computing, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, London, UK.
- Behaviour Analytics Lab, Data Science Institute, Imperial College London, London, UK.
- Brain & Behaviour Lab, Institute for Artificial and Human Intelligence, University of Bayreuth, Bayreuth, Germany.
- Chair in Digital Health, Faculty of Life Sciences, University of Bayreuth, Bayreuth, Germany.
| |
Collapse
|
16
|
Rummey C, Corben LA, Delatycki M, Wilmot G, Subramony SH, Corti M, Bushara K, Duquette A, Gomez C, Hoyle JC, Roxburgh R, Seeberger L, Yoon G, Mathews K, Zesiewicz T, Perlman S, Lynch DR. Natural History of Friedreich Ataxia: Heterogeneity of Neurologic Progression and Consequences for Clinical Trial Design. Neurology 2022; 99:e1499-e1510. [PMID: 35817567 PMCID: PMC9576299 DOI: 10.1212/wnl.0000000000200913] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The understanding of the natural history of Friedreich ataxia (FRDA) has improved considerably recently, but patterns of neurologic deterioration are not fully clarified, compromising the assessment of the clinical relevance of effects and guidance for study design. The goal of this study was to acknowledge the broad genetic diversity of the population, especially for younger individuals, and to provide analyses stratified by age to guide population selection in future studies. METHODS Based on a large natural history study, the FRDA Clinical Outcome Measures study that at the current data cut enrolled 1,115 participants, followed up for 5,287 yearly visits, we present results from the modified FRDA Rating Scale and its subscores. The secondary outcomes included the patient-reported activities of daily living scale, the timed 25-foot walk, and the 9-hole peg test. Long-term progression was modeled using slope analyses within early-onset, typical-onset, intermediate-onset, and late-onset FRDA. To reflect recruitment in clinical trials, short-term changes were analyzed within age-based subpopulations. All analyses were stratified by ambulation status. RESULTS Long-term progression models stratified by disease severity indicated highly differential disease progression, especially at earlier ages at onset. In the ambulatory phase, decline was driven by axial items assessed by the Upright Stability subscore of the mFARS. The analyses of short-term changes showed slower progression with increasing population age due to decreasing genetic severity. Future clinical studies could reduce population diversity, interpatient variability, and the risk of imbalanced treatment groups by selecting the study population based on the functional capacity (e.g., ambulatory status) and by strict age-based stratification. DISCUSSION The understanding of the diversity within FRDA populations and their patterns of functional decline provides an essential foundation for future clinical trial design including patient selection and facilitates the interpretation of the clinical relevance of progression detected in FRDA.
Collapse
Affiliation(s)
- Christian Rummey
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Louise A Corben
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Martin Delatycki
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - George Wilmot
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Sub H Subramony
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Manuela Corti
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Khalaf Bushara
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Antoine Duquette
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Christopher Gomez
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - J Chad Hoyle
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Richard Roxburgh
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Lauren Seeberger
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Grace Yoon
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Katherine Mathews
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Theresa Zesiewicz
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Susan Perlman
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - David R Lynch
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA.
| |
Collapse
|
17
|
Keita M, McIntyre K, Rodden LN, Schadt K, Lynch DR. Friedreich ataxia: clinical features and new developments. Neurodegener Dis Manag 2022; 12:267-283. [PMID: 35766110 PMCID: PMC9517959 DOI: 10.2217/nmt-2022-0011] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022] Open
Abstract
Friedreich's ataxia (FRDA), a neurodegenerative disease characterized by ataxia and other neurological features, affects 1 in 50,000-100,000 individuals in the USA. However, FRDA also includes cardiac, orthopedic and endocrine dysfunction, giving rise to many secondary disease characteristics. The multifaceted approach for clinical care has necessitated the development of disease-specific clinical care guidelines. New developments in FRDA include the advancement of clinical drug trials targeting the NRF2 pathway and frataxin restoration. Additionally, a novel understanding of gene silencing in FRDA, reflecting a variegated silencing pattern, will have applications to current and future therapeutic interventions. Finally, new perspectives on the neuroanatomy of FRDA and its developmental features will refine the time course and anatomical targeting of novel approaches.
Collapse
Affiliation(s)
- Medina Keita
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kellie McIntyre
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Layne N Rodden
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kim Schadt
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Pediatrics & Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Masnovo C, Lobo AF, Mirkin SM. Replication dependent and independent mechanisms of GAA repeat instability. DNA Repair (Amst) 2022; 118:103385. [PMID: 35952488 PMCID: PMC9675320 DOI: 10.1016/j.dnarep.2022.103385] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/20/2022]
Abstract
Trinucleotide repeat instability is a driver of human disease. Large expansions of (GAA)n repeats in the first intron of the FXN gene are the cause Friedreich's ataxia (FRDA), a progressive degenerative disorder which cannot yet be prevented or treated. (GAA)n repeat instability arises during both replication-dependent processes, such as cell division and intergenerational transmission, as well as in terminally differentiated somatic tissues. Here, we provide a brief historical overview on the discovery of (GAA)n repeat expansions and their association to FRDA, followed by recent advances in the identification of triplex H-DNA formation and replication fork stalling. The main body of this review focuses on the last decade of progress in understanding the mechanism of (GAA)n repeat instability during DNA replication and/or DNA repair. We propose that the discovery of additional mechanisms of (GAA)n repeat instability can be achieved via both comparative approaches to other repeat expansion diseases and genome-wide association studies. Finally, we discuss the advances towards FRDA prevention or amelioration that specifically target (GAA)n repeat expansions.
Collapse
Affiliation(s)
- Chiara Masnovo
- Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Ayesha F Lobo
- Department of Biology, Tufts University, Medford, MA 02155, USA
| | - Sergei M Mirkin
- Department of Biology, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
19
|
Wang D, Ho ES, Cotticelli MG, Xu P, Napierala JS, Hauser LA, Napierala M, Himes BE, Wilson RB, Lynch DR, Mesaros C. Skin fibroblast metabolomic profiling reveals that lipid dysfunction predicts the severity of Friedreich's ataxia. J Lipid Res 2022; 63:100255. [PMID: 35850241 PMCID: PMC9399481 DOI: 10.1016/j.jlr.2022.100255] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/26/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by a triplet guanine-adenine-adenine (GAA) repeat expansion in intron 1 of the FXN gene, which leads to decreased levels of the frataxin protein. Frataxin is involved in the formation of iron-sulfur (Fe-S) cluster prosthetic groups for various metabolic enzymes. To provide a better understanding of the metabolic status of patients with FRDA, here we used patient-derived fibroblast cells as a surrogate tissue for metabolic and lipidomic profiling by liquid chromatography-high resolution mass spectrometry. We found elevated HMG-CoA and β-hydroxybutyrate-CoA levels, implying dysregulated fatty acid oxidation, which was further demonstrated by elevated acyl-carnitine levels. Lipidomic profiling identified dysregulated levels of several lipid classes in FRDA fibroblast cells when compared with non-FRDA fibroblast cells. For example, levels of several ceramides were significantly increased in FRDA fibroblast cells; these results positively correlated with the GAA repeat length and negatively correlated with the frataxin protein levels. Furthermore, stable isotope tracing experiments indicated increased ceramide synthesis, especially for long-chain fatty acid-ceramides, in FRDA fibroblast cells compared with ceramide synthesis in healthy control fibroblast cells. In addition, PUFA-containing triglycerides and phosphatidylglycerols were enriched in FRDA fibroblast cells and negatively correlated with frataxin levels, suggesting lipid remodeling as a result of FXN deficiency. Altogether, we demonstrate patient-derived fibroblast cells exhibited dysregulated metabolic capabilities, and their lipid dysfunction predicted the severity of FRDA, making them a useful surrogate to study the metabolic status in FRDA.
Collapse
Affiliation(s)
- Dezhen Wang
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elaine S Ho
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M Grazia Cotticelli
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Peining Xu
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jill S Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, Birmingham, Alabama, USA
| | - Lauren A Hauser
- Department of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research, Philadelphia, Pennsylvania, USA
| | - Marek Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, Birmingham, Alabama, USA
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert B Wilson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David R Lynch
- Department of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research, Philadelphia, Pennsylvania, USA
| | - Clementina Mesaros
- Center for Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
20
|
Dong YNA, Mesaros C, Xu P, Mercado-Ayón E, Halawani S, Ngaba LV, Warren N, Sleiman P, Rodden LN, Schadt KA, Blair IA, Lynch DR. Frataxin controls ketone body metabolism through regulation of OXCT1. PNAS NEXUS 2022; 1:pgac142. [PMID: 36016708 PMCID: PMC9396447 DOI: 10.1093/pnasnexus/pgac142] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/21/2022] [Indexed: 02/05/2023]
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by the deficiency of mitochondrial protein frataxin, which plays a crucial role in iron-sulphur cluster formation and ATP production. The cellular function of frataxin is not entirely known. Here, we demonstrate that frataxin controls ketone body metabolism through regulation of 3-Oxoacid CoA-Transferase 1 (OXCT1), a rate limiting enzyme catalyzing the conversion of ketone bodies to acetoacetyl-CoA that is then fed into the Krebs cycle. Biochemical studies show a physical interaction between frataxin and OXCT1 both in vivo and in vitro. Frataxin overexpression also increases OXCT1 protein levels in human skin fibroblasts while frataxin deficiency decreases OXCT1 in multiple cell types including cerebellum and skeletal muscle both acutely and chronically, suggesting that frataxin directly regulates OXCT1. This regulation is mediated by frataxin-dependent suppression of ubiquitin-proteasome system (UPS)-dependent OXCT1 degradation. Concomitantly, plasma ketone bodies are significantly elevated in frataxin deficient knock-in/knockout (KIKO) mice with no change in the levels of other enzymes involved in ketone body production. In addition, ketone bodies fail to be metabolized to acetyl-CoA accompanied by increased succinyl-CoA in vitro in frataxin deficient cells, suggesting that ketone body elevation is caused by frataxin-dependent reduction of OXCT1 leading to deficits in tissue utilization of ketone bodies. Considering the potential role of metabolic abnormalities and deficiency of ATP production in FRDA, our results suggest a new role for frataxin in ketone body metabolism and also suggest modulation of OXCT1 may be a potential therapeutic approach for FRDA.
Collapse
Affiliation(s)
- Yi NA Dong
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peining Xu
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sarah Halawani
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lucie Vanessa Ngaba
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nathan Warren
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Patrick Sleiman
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Layne N Rodden
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kimberly A Schadt
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
21
|
Rodden LN, Rummey C, Dong YN, Lynch DR. Clinical Evidence for Variegated Silencing in Patients With Friedreich Ataxia. Neurol Genet 2022; 8:e683. [PMID: 35620135 PMCID: PMC9128033 DOI: 10.1212/nxg.0000000000000683] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/30/2022] [Indexed: 11/15/2022]
Abstract
Background and Objectives Friedreich ataxia (FRDA) is a neurodegenerative disease caused by a GAA triplet repeat (GAA-TR) expansion in intron 1 of the FXN gene. Patients have 100-1,300 GAA triplets compared with less than 30 in healthy controls. The GAA-TR expansion leads to FXN silencing, and consequent frataxin protein deficiency results in progressive ataxia, scoliosis, cardiomyopathy, and diabetes. The overt heterogeneity in age at onset and disease severity is explained partly by the length of the GAA-TR, in which shorter repeats correlate with milder disease. Evidence of variegated silencing in FRDA suggests that patients with shorter repeats retain a significant proportion of cells with FXN genes that have escaped GAA-TR expansion-induced silencing, explaining the less severe frataxin deficiency in this subpopulation. In ex vivo experiments, the proportion of spared cells negatively correlates with GAA-TR length until it plateaus at 500 triplets, an indication that the maximal number of silenced cells has been reached. In this study, we assessed whether an analogous ceiling effect occurs in severity of clinical features of FRDA by analyzing clinical outcome data. Methods The FRDA Clinical Outcome Measures Study database was used for a cross-sectional analysis of 1,000 patients with FRDA. Frataxin levels were determined by lateral flow immunoassays. Results The length of the GAA-TR in our cohort predicted frataxin level (R2 = 0.38, p < 0.0001) and age at onset (R2 = 0.46, p < 0.0001) but only with GAA-TRs with ≤700 triplets. Age and disease duration predicted performance on clinical outcome measures, and such predictions in linear regression models statistically improved in the subcohort of patients with >700 GAA triplets. The prevalence of cardiomyopathy and scoliosis increased as GAA-TR length increased up to 700 GAA triplets where prevalence plateaued. Discussion Our data suggest that there is a ceiling effect on the clinical consequences of GAA-TR length in FRDA, as would be predicted by variegated silencing. Patients with GAA-TRs of >700 triplets represent a subgroup in which the severity of clinical manifestations based on GAA-TR length have reached maximal levels and therefore display limited clinical variability in disease progression.
Collapse
Affiliation(s)
- Layne N. Rodden
- From the Departments of Pediatrics and Neurology (L.N.R., Y.N.D., D.R.L.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; and Clinical Data Science GmbH (C.R.), Basel, Switzerland
| | - Christian Rummey
- From the Departments of Pediatrics and Neurology (L.N.R., Y.N.D., D.R.L.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; and Clinical Data Science GmbH (C.R.), Basel, Switzerland
| | - Yi Na Dong
- From the Departments of Pediatrics and Neurology (L.N.R., Y.N.D., D.R.L.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; and Clinical Data Science GmbH (C.R.), Basel, Switzerland
| | - David R. Lynch
- From the Departments of Pediatrics and Neurology (L.N.R., Y.N.D., D.R.L.), Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; and Clinical Data Science GmbH (C.R.), Basel, Switzerland
| |
Collapse
|
22
|
Wang Q, Laboureur L, Weng L, Eskenazi NM, Hauser LA, Mesaros C, Lynch DR, Blair IA. Simultaneous Quantification of Mitochondrial Mature Frataxin and Extra-Mitochondrial Frataxin Isoform E in Friedreich’s Ataxia Blood. Front Neurosci 2022; 16:874768. [PMID: 35573317 PMCID: PMC9098139 DOI: 10.3389/fnins.2022.874768] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is an autosomal recessive disease caused by an intronic guanine-adenine-adenine (GAA) triplet expansion in the frataxin (FXN) gene, which leads to reduced expression of full-length frataxin (1–210) also known as isoform 1. Full-length frataxin has a mitochondrial targeting sequence, which facilitates its translocation into mitochondria where it is processed through cleavage at G41-L42 and K80-S81 by mitochondrial processing (MPP) to release mitochondrial mature frataxin (81–210). Alternative splicing of FXN also leads to expression of N-terminally acetylated extra-mitochondrial frataxin (76–210) named isoform E because it was discovered in erythrocytes. Frataxin isoforms are undetectable in serum or plasma, and originally whole blood could not be used as a biomarker in brief therapeutic trials because it is present in erythrocytes, which have a half-life of 115-days and so frataxin levels would remain unaltered. Therefore, an assay was developed for analyzing frataxin in platelets, which have a half-life of only 10-days. However, our discovery that isoform E is only present in erythrocytes, whereas, mature frataxin is present primarily in short-lived peripheral blood mononuclear cells (PBMCs), granulocytes, and platelets, meant that both proteins could be quantified in whole blood samples. We now report a quantitative assay for frataxin proteoforms in whole blood from healthy controls and FRDA patients. The assay is based on stable isotope dilution coupled with immunoprecipitation (IP) and two-dimensional-nano-ultrahigh performance liquid chromatography/parallel reaction monitoring/high resolution mass spectrometry (2D-nano-UHPLC-PRM/HRMS). The lower limit of quantification was 0.5 ng/mL for each proteoform and the assays had 100% sensitivity and specificity for discriminating between healthy controls (n = 11) and FRDA cases (N = 100 in year-1, N = 22 in year-2,3). The mean levels of mature frataxin in whole blood from healthy controls and homozygous FRDA patients were significantly different (p < 0.0001) at 7.5 ± 1.5 ng/mL and 2.1 ± 1.2 ng/mL, respectively. The mean levels of isoform E in whole blood from healthy controls and homozygous FRDA patients were significantly different (p < 0.0001) at 26.8 ± 4.1 ng/mL and 4.7 ± 3.3 ng/mL, respectively. The mean levels of total frataxin in whole blood from healthy controls and homozygous FRDA patients were significantly different (p < 0.0001) at 34.2 ± 4.3 ng/mL and 6.8 ± 4.0 ng/mL, respectively. The assay will make it possible to rigorously monitor the natural history of the disease and explore the potential role of isoform E in etiology of the disease. It will also facilitate the assessment of therapeutic interventions (including gene therapy approaches) that attempt to increase frataxin protein expression as a treatment for this devastating disease.
Collapse
Affiliation(s)
- Qingqing Wang
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
| | - Laurent Laboureur
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
| | - Liwei Weng
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
| | - Nicolas M. Eskenazi
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
| | - Lauren A. Hauser
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
- Departments of Pediatrics and Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Departments of Pediatrics and Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
| | - David R. Lynch
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
- Departments of Pediatrics and Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Departments of Pediatrics and Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ian A. Blair
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Penn/CHOP Center of Excellence in Friedreich’s Ataxia, Philadelphia, PA, United States
- *Correspondence: Ian A. Blair,
| |
Collapse
|
23
|
Monfort B, Want K, Gervason S, D’Autréaux B. Recent Advances in the Elucidation of Frataxin Biochemical Function Open Novel Perspectives for the Treatment of Friedreich’s Ataxia. Front Neurosci 2022; 16:838335. [PMID: 35310092 PMCID: PMC8924461 DOI: 10.3389/fnins.2022.838335] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is the most prevalent autosomic recessive ataxia and is associated with a severe cardiac hypertrophy and less frequently diabetes. It is caused by mutations in the gene encoding frataxin (FXN), a small mitochondrial protein. The primary consequence is a defective expression of FXN, with basal protein levels decreased by 70–98%, which foremost affects the cerebellum, dorsal root ganglia, heart and liver. FXN is a mitochondrial protein involved in iron metabolism but its exact function has remained elusive and highly debated since its discovery. At the cellular level, FRDA is characterized by a general deficit in the biosynthesis of iron-sulfur (Fe-S) clusters and heme, iron accumulation and deposition in mitochondria, and sensitivity to oxidative stress. Based on these phenotypes and the proposed ability of FXN to bind iron, a role as an iron storage protein providing iron for Fe-S cluster and heme biosynthesis was initially proposed. However, this model was challenged by several other studies and it is now widely accepted that FXN functions primarily in Fe-S cluster biosynthesis, with iron accumulation, heme deficiency and oxidative stress sensitivity appearing later on as secondary defects. Nonetheless, the biochemical function of FXN in Fe-S cluster biosynthesis is still debated. Several roles have been proposed for FXN: iron chaperone, gate-keeper of detrimental Fe-S cluster biosynthesis, sulfide production stimulator and sulfur transfer accelerator. A picture is now emerging which points toward a unique function of FXN as an accelerator of a key step of sulfur transfer between two components of the Fe-S cluster biosynthetic complex. These findings should foster the development of new strategies for the treatment of FRDA. We will review here the latest discoveries on the biochemical function of frataxin and the implication for a potential therapeutic treatment of FRDA.
Collapse
|
24
|
Abstract
INTRODUCTION Friedreich ataxia (FRDA) is an autosomal recessive disorder caused by deficiency of frataxin, an essential mitochondrial protein involved in iron sulfur cluster biogenesis, oxidative phosphorylation and other processes. FRDA most notably affects the heart, sensory neurons, spinal cord, cerebellum, and other brain regions, and manifests clinically as ataxia, sensory loss, dysarthria, spasticity, and hypertrophic cardiomyopathy. Therapeutic approaches in FRDA have consisted of two different approaches: (1) augmenting or restoring frataxin production and (2) modulating a variety of downstream processes related to mitochondrial dysfunction, including reactive oxygen species production, ferroptosis, or Nrf2 activation. AREAS COVERED In this review, we summarize data from major phase II clinical trials in FRDA published between 2015 and 2020, which includes A0001/EPI743, Omaveloxolone, RT001, and Actimmune. EXPERT OPINION A growing number of drug candidates are being tested in phase II clinical trials for FRDA; however, most have not met their primary endpoints, and none have received FDA approval. In this review, we aim to summarize completed phase II clinical trials in FRDA, outlining critical lessons that have been learned and that should be incorporated into future trial design to ultimately optimize drug development in FRDA.
Collapse
|
25
|
Correlation of Visual Quality of Life With Clinical and Visual Status in Friedreich Ataxia. J Neuroophthalmol 2021; 40:213-217. [PMID: 31977662 DOI: 10.1097/wno.0000000000000878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The primary objective was to determine the association of patient-reported vision-specific quality of life to disease status and visual function in patients with Friedreich's ataxia (FRDA). METHODS Patients with FRDA were assessed with the 25-Item National Eye Institute Visual Functioning Questionnaire (NEI-VFQ-25) along with measures of disease status (ataxia stage) and visual function (low- and high-contrast letter acuity scores). The relations of NEI-VFQ-25 scores to those for disease status and visual function were examined. RESULTS Scores for the NEI-VFQ-25 were lower in patients with FRDA (n = 99) compared with published disease-free controls, particularly reduced in a subgroup of FRDA patients with features of early onset, older age, and abnormal visual function. CONCLUSIONS The NEI-VFQ-25 captures the subjective component of visual function in patients with FRDA.
Collapse
|
26
|
Lynch DR, Schadt K, Kichula E, McCormack S, Lin KY. Friedreich Ataxia: Multidisciplinary Clinical Care. J Multidiscip Healthc 2021; 14:1645-1658. [PMID: 34234452 PMCID: PMC8253929 DOI: 10.2147/jmdh.s292945] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/04/2021] [Indexed: 12/17/2022] Open
Abstract
Friedreich ataxia (FRDA) is a multisystem disorder affecting 1 in 50,000-100,000 person in the United States. Traditionally viewed as a neurodegenerative disease, FRDA patients also develop cardiomyopathy, scoliosis, diabetes and other manifestation. Although it usually presents in childhood, it continues throughout life, thus requiring expertise from both pediatric and adult subspecialist in order to provide optimal management. The phenotype of FRDA is unique, giving rise to specific loss of neuronal pathways, a unique form of cardiomyopathy with early hypertrophy and later fibrosis, and diabetes incorporating components of both type I and type II disease. Vision loss, hearing loss, urinary dysfunction and depression also occur in FRDA. Many agents are reaching Phase III trials; if successful, these will provide a variety of new treatments for FRDA that will require many specialists who are not familiar with FRDA to provide clinical therapy. This review provides a summary of the diverse manifestation of FRDA, existing symptomatic therapies, and approaches for integrative care for future therapy in FRDA.
Collapse
Affiliation(s)
- David R Lynch
- Division of Neurology, Departments of Pediatrics and Neurology, Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kim Schadt
- Division of Neurology, Departments of Pediatrics and Neurology, Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Elizabeth Kichula
- Division of Neurology, Departments of Pediatrics and Neurology, Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Shana McCormack
- Division of Endocrinology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Kimberly Y Lin
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, Philadelphia, PA, 19104, USA
| |
Collapse
|
27
|
Napierala JS, Rajapakshe K, Clark A, Chen YY, Huang S, Mesaros C, Xu P, Blair IA, Hauser LA, Farmer J, Lynch DR, Edwards DP, Coarfa C, Napierala M. Reverse Phase Protein Array Reveals Correlation of Retinoic Acid Metabolism With Cardiomyopathy in Friedreich's Ataxia. Mol Cell Proteomics 2021; 20:100094. [PMID: 33991687 PMCID: PMC8214145 DOI: 10.1016/j.mcpro.2021.100094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022] Open
Abstract
Identifying biomarkers is important for assessment of disease progression, prediction of symptom development, and determination of treatment effectiveness. While unbiased analyses of differential gene expression using next-generation sequencing methods are now routinely conducted, proteomics studies are more challenging because of traditional methods predominantly being low throughput and offering a limited dynamic range for simultaneous detection of hundreds of proteins that drastically differ in their intracellular abundance. We utilized a sensitive and high-throughput proteomic technique, reverse phase protein array (RPPA), to attain protein expression profiles of primary fibroblasts obtained from patients with Friedreich's ataxia (FRDA) and unaffected controls (CTRLs). The RPPA was designed to detect 217 proteins or phosphorylated proteins by individual antibody, and the specificity of each antibody was validated prior to the experiment. Among 62 fibroblast samples (44 FRDA and 18 CTRLs) analyzed, 30 proteins/phosphoproteins were significantly changed in FRDA fibroblasts compared with CTRL cells (p < 0.05), mostly representing signaling molecules and metabolic enzymes. As expected, frataxin was significantly downregulated in FRDA samples, thus serving as an internal CTRL for assay integrity. Extensive bioinformatics analyses were conducted to correlate differentially expressed proteins with critical disease parameters (e.g., selected symptoms, age of onset, guanine-adenine-adenine sizes, frataxin levels, and Functional Assessment Rating Scale scores). Members of the integrin family of proteins specifically associated with hearing loss in FRDA. Also, RPPA data, combined with results of transcriptome profiling, uncovered defects in the retinoic acid metabolism pathway in FRDA samples. Moreover, expression of aldehyde dehydrogenase family 1 member A3 differed significantly between cardiomyopathy-positive and cardiomyopathy-negative FRDA cohorts, demonstrating that metabolites such as retinol, retinal, or retinoic acid could become potential predictive biomarkers of cardiac presentation in FRDA.
Collapse
Affiliation(s)
- Jill S Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Amanda Clark
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yu-Yun Chen
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Clementina Mesaros
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peining Xu
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian A Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren A Hauser
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer Farmer
- Friedreich's Ataxia Research Alliance, Downingtown, Pennsylvania, USA
| | - David R Lynch
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
28
|
Moreno-Lorite J, Pérez-Luz S, Katsu-Jiménez Y, Oberdoerfer D, Díaz-Nido J. DNA repair pathways are altered in neural cell models of frataxin deficiency. Mol Cell Neurosci 2021; 111:103587. [PMID: 33418083 DOI: 10.1016/j.mcn.2020.103587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/20/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a hereditary and predominantly neurodegenerative disease caused by a deficiency of the protein frataxin (FXN). As part of the overall efforts to understand the molecular basis of neurodegeneration in FRDA, a new human neural cell line with doxycycline-induced FXN knockdown was established. This cell line, hereafter referred to as iFKD-SY, is derived from the human neuroblastoma SH-SY5Y and retains the ability to differentiate into mature neuron-like cells. In both proliferating and differentiated iFKD-SY cells, the induction of FXN deficiency is accompanied by increases in oxidative stress and DNA damage, reduced aconitase enzyme activity, higher levels of p53 and p21, activation of caspase-3, and subsequent apoptosis. More interestingly, FXN-deficient iFKD-SY cells exhibit an important transcriptional deregulation in many of the genes implicated in DNA repair pathways. The levels of some crucial proteins involved in DNA repair appear notably diminished. Furthermore, similar changes are found in two additional neural cell models of FXN deficit: primary cultures of FXN-deficient mouse neurons and human olfactory mucosa stem cells obtained from biopsies of FRDA patients. These results suggest that the deficiency of FXN leads to a down-regulation of DNA repair pathways that synergizes with oxidative stress to provoke DNA damage, which may be involved in the pathogenesis of FRDA. Thus, a failure in DNA repair may be considered a shared common molecular mechanism contributing to neurodegeneration in a number of hereditary ataxias including FRDA.
Collapse
Affiliation(s)
- Jara Moreno-Lorite
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM), Spain
| | - Sara Pérez-Luz
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM), Spain; Molecular Genetics Unit, Institute of Rare Diseases Research, Institute of Health Carlos III (ISCIII), Ctra. Majadahonda-Pozuelo Km2.200, 28220 Madrid, Spain.
| | - Yurika Katsu-Jiménez
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM), Spain
| | - Daniel Oberdoerfer
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM), Spain
| | - Javier Díaz-Nido
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda (IDIPHIM), Spain
| |
Collapse
|
29
|
Doni D, Rigoni G, Palumbo E, Baschiera E, Peruzzo R, De Rosa E, Caicci F, Passerini L, Bettio D, Russo A, Szabò I, Soriano ME, Salviati L, Costantini P. The displacement of frataxin from the mitochondrial cristae correlates with abnormal respiratory supercomplexes formation and bioenergetic defects in cells of Friedreich ataxia patients. FASEB J 2021; 35:e21362. [PMID: 33629768 DOI: 10.1096/fj.202000524rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 12/09/2020] [Accepted: 12/28/2020] [Indexed: 12/23/2022]
Abstract
Friedreich ataxia (FRDA) is a neurodegenerative disease resulting from a severe decrease of frataxin (FXN). Most patients carry a GAA repeat expansion in both alleles of the FXN gene, whereas a small fraction of them are compound heterozygous for the expansion and a point mutation in the other allele. FXN is involved in the mitochondrial biogenesis of the FeS-clusters. Distinctive feature of FRDA patient cells is an impaired cellular respiration, likely due to a deficit of key redox cofactors working as electrons shuttles through the respiratory chain. However, a definite relationship between FXN levels, FeS-clusters assembly dysregulation and bioenergetics failure has not been established. In this work, we performed a comparative analysis of the mitochondrial phenotype of cell lines from FRDA patients, either homozygous for the expansion or compound heterozygotes for the G130V mutation. We found that, in healthy cells, FXN and two key proteins of the FeS-cluster assembly machinery are enriched in mitochondrial cristae, the dynamic subcompartment housing the respiratory chain. On the contrary, FXN widely redistributes to the matrix in FRDA cells with defects in respiratory supercomplexes assembly and altered respiratory function. We propose that this could be relevant for the early mitochondrial defects afflicting FRDA cells and that perturbation of mitochondrial morphodynamics could in turn be critical in terms of disease mechanisms.
Collapse
Affiliation(s)
- Davide Doni
- Department of Biology, University of Padova, Padova, Italy
| | | | - Elisa Palumbo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Elisa Baschiera
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, Padova, Italy
| | | | - Edith De Rosa
- Department of Biology, University of Padova, Padova, Italy
| | | | | | - Daniela Bettio
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, Padova, Italy
| | - Antonella Russo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Ildiko Szabò
- Department of Biology, University of Padova, Padova, Italy
| | | | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, Padova, Italy
- Myology Center, University of Padova, Padova, Italy
| | | |
Collapse
|
30
|
Santoro M, Perna A, La Rosa P, Petrillo S, Piemonte F, Rossi S, Riso V, Nicoletti TF, Modoni A, Pomponi MG, Chiurazzi P, Silvestri G. Compound heterozygosity for an expanded (GAA) and a (GAAGGA) repeat at FXN locus: from a diagnostic pitfall to potential clues to the pathogenesis of Friedreich ataxia. Neurogenetics 2020; 21:279-287. [PMID: 32638185 DOI: 10.1007/s10048-020-00620-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/14/2020] [Indexed: 12/23/2022]
Abstract
Friedreich's ataxia (FRDA) is usually due to a homozygous GAA expansion in intron 1 of the frataxin (FXN) gene. Rarely, uncommon molecular rearrangements at the FXN locus can cause pitfalls in the molecular diagnosis of FRDA. Here we describe a family whose proband was affected by late-onset Friedreich's ataxia (LOFA); long-range PCR (LR-PCR) documented two small expanded GAA alleles both in the proband and in her unaffected younger sister, who therefore received a diagnosis of pre-symptomatic LOFA. Later studies, however, revealed that the proband's unaffected sister, as well as their healthy mother, were both carriers of an expanded GAA allele and an uncommon (GAAGGA)66-67 repeat mimicking a GAA expansion at the LR-PCR that was the cause of the wrong initial diagnosis of pre-symptomatic LOFA. Extensive studies in tissues from all the family members, including LR-PCR, assessment of methylation status of FXN locus, MboII restriction analysis and direct sequencing of LR-PCR products, analysis of FXN mRNA, and frataxin protein expression, support the virtual lack of pathogenicity of the rare (GAAGGA)66-67 repeat, also providing significant data about the modulation of epigenetic modifications at the FXN locus. Overall, this report highlights a rare but possible pitfall in FRDA molecular diagnosis, emphasizing the need of further analysis in case of discrepancy between clinical and molecular data.
Collapse
Affiliation(s)
- Massimo Santoro
- IRCCS Fondazione Don Carlo Gnocchi, Piazzale Morandi, 6, 20121, Milan, Italy
| | - Alessia Perna
- Dept of Neuroscience, Faculty of Medicine and Surgery, Università Cattolica del Scaro Cuore, L.go F. Vito 1, 000168, Rome, Italy
| | - Piergiorgio La Rosa
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale San Paolo, 15, 00146, Rome, Italy
| | - Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale San Paolo, 15, 00146, Rome, Italy
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Ospedale Pediatrico Bambino Gesù, IRCCS, Viale San Paolo, 15, 00146, Rome, Italy
| | - Salvatore Rossi
- Dept of Neuroscience, Faculty of Medicine and Surgery, Università Cattolica del Scaro Cuore, L.go F. Vito 1, 000168, Rome, Italy
| | - Vittorio Riso
- Dept of Neuroscience, Faculty of Medicine and Surgery, Università Cattolica del Scaro Cuore, L.go F. Vito 1, 000168, Rome, Italy
- Institute of Neurology, Neuroscience Area, Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy
| | - Tommaso Filippo Nicoletti
- Dept of Neuroscience, Faculty of Medicine and Surgery, Università Cattolica del Scaro Cuore, L.go F. Vito 1, 000168, Rome, Italy
- Institute of Neurology, Neuroscience Area, Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy
| | - Anna Modoni
- Institute of Neurology, Neuroscience Area, Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy
| | - Maria Grazia Pomponi
- Institute of Genomic Medicine, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy
| | - Pietro Chiurazzi
- Institute of Genomic Medicine, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy
| | - Gabriella Silvestri
- Dept of Neuroscience, Faculty of Medicine and Surgery, Università Cattolica del Scaro Cuore, L.go F. Vito 1, 000168, Rome, Italy.
- Institute of Neurology, Neuroscience Area, Neurology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168, Rome, Italy.
| |
Collapse
|
31
|
Clay A, Obrochta KM, Soon RK, Russell CB, Lynch DR. Neurofilament light chain as a potential biomarker of disease status in Friedreich ataxia. J Neurol 2020; 267:2594-2598. [PMID: 32385683 DOI: 10.1007/s00415-020-09868-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND The present study evaluates serum neurofilament light chain (NfL) as a biomarker of disease features in Friedreich's ataxia (FRDA). METHODS NfL levels from serum of 117 subjects (85 FRDA patients, 13 carriers, and 19 controls) were assayed and correlated with disease features such as smaller GAA repeat length (GAA1), age, sex, and level of neurological dysfunction. RESULTS Mean serum NfL levels were higher in FRDA patients than in carriers or unaffected controls in two independent cohorts of subjects. In longitudinal samples from FRDA patients drawn monthly or 1 year apart, values changed minimally. No difference was noted between carriers and controls. NfL levels correlated positively with age in controls and carriers of similar age, (Rs = 0.72, p < 0.0005), whereas NfL levels inversely correlated with age in FRDA patients (Rs = - 0.63, p < 0.001). NfL levels were not associated with sex or GAA1 length in patients, and linear regression revealed a significant relationship between NfL levels in the cohort with age (coefficient = - 0.36, p < 0.001), but not sex (p = 0.64) or GAA1 (p = 0.13). CONCLUSION Because NfL is elevated in patients, but decreases with age and disease progression, our results suggest that age is the critical determinant of NfL in FRDA (rather than clinical or genetic severity).
Collapse
Affiliation(s)
- Alexandra Clay
- Department of Pediatrics and Neurology, The Children's Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kristin M Obrochta
- BioMarin Pharmaceutical Inc, 770 Lindaro Street, San Rafael, CA, 94901, USA
| | - Russell K Soon
- BioMarin Pharmaceutical Inc, 770 Lindaro Street, San Rafael, CA, 94901, USA
| | | | - David R Lynch
- Department of Pediatrics and Neurology, The Children's Hospital of Philadelphia; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
La Rosa P, Russo M, D'Amico J, Petrillo S, Aquilano K, Lettieri-Barbato D, Turchi R, Bertini ES, Piemonte F. Nrf2 Induction Re-establishes a Proper Neuronal Differentiation Program in Friedreich's Ataxia Neural Stem Cells. Front Cell Neurosci 2019; 13:356. [PMID: 31417369 PMCID: PMC6685360 DOI: 10.3389/fncel.2019.00356] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Frataxin deficiency is the pathogenic cause of Friedreich’s Ataxia, an autosomal recessive disease characterized by the increase of oxidative stress and production of free radicals in the cell. Although the onset of the pathology occurs in the second decade of life, cognitive differences and defects in brain structure and functional activation are observed in patients, suggesting developmental defects to take place during fetal neurogenesis. Here, we describe impairments in proliferation, stemness potential and differentiation in neural stem cells (NSCs) isolated from the embryonic cortex of the Frataxin Knockin/Knockout mouse, a disease animal model whose slow-evolving phenotype makes it suitable to study pre-symptomatic defects that may manifest before the clinical onset. We demonstrate that enhancing the expression and activity of the antioxidant response master regulator Nrf2 ameliorates the phenotypic defects observed in NSCs, re-establishing a proper differentiation program.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Marta Russo
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Jessica D'Amico
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Sara Petrillo
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Daniele Lettieri-Barbato
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Riccardo Turchi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Enrico S Bertini
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Fiorella Piemonte
- Unit of Neuromuscular and Neurodegenerative Diseases, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
33
|
Clay A, Hearle P, Schadt K, Lynch DR. New developments in pharmacotherapy for Friedreich ataxia. Expert Opin Pharmacother 2019; 20:1855-1867. [PMID: 31311349 DOI: 10.1080/14656566.2019.1639671] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Friedreich ataxia (FRDA), a rare disease caused by the deficiency of the mitochondrial matrix protein frataxin, affects roughly 1 in 50,000 individuals worldwide. Current and emerging therapies focus on reversing the deleterious effects of such deficiency including mitochondrial augmentation and increasing frataxin levels, providing the possibility of treatment options for this physiologically complex, multisystem disorder. Areas covered: In this review article, the authors discuss the current and prior in vivo and in vitro research studies related to the treatment of FRDA, with a particular interest in future implications of each therapy. Expert opinion: Since the discovery of FXN in 1996, multiple clinical trials have occurred or are currently occurring; at a rapid pace for a rare disease. These trials have been directed at the augmentation of mitochondrial function and/or alleviation of symptoms and are not regarded as potential cures in FRDA. Either a combination of therapies or a drug that replaces or increases the pathologically low levels of frataxin better represent potential cures in FRDA.
Collapse
Affiliation(s)
- Alexandra Clay
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Patrick Hearle
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Kim Schadt
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - David R Lynch
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| |
Collapse
|
34
|
The current state of biomarker research for Friedreich's ataxia: a report from the 2018 FARA biomarker meeting. Future Sci OA 2019; 5:FSO398. [PMID: 31285843 PMCID: PMC6609901 DOI: 10.2144/fsoa-2019-0026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The 2018 FARA Biomarker Meeting highlighted the current state of development of biomarkers for Friedreich’s ataxia. A mass spectroscopy assay to sensitively measure mature frataxin (reduction of which is the root cause of disease) is being developed. Biomarkers to monitor neurological disease progression include imaging, electrophysiological measures and measures of nerve function, which may be measured either in serum and/or through imaging-based technologies. Potential pharmacodynamic biomarkers include metabolic and protein biomarkers and markers of nerve damage. Cardiac imaging and serum biomarkers may reflect cardiac disease progression. Considerable progress has been made in the development of biomarkers for various contexts of use, but further work is needed in terms of larger longitudinal multisite studies, and identification of novel biomarkers for additional use cases Biomarkers are characteristics that can be objectively measured, evaluated and used as indicators of disease progression or the effect of a therapy. Friedreich’s ataxia is a progressive multisystem neuromuscular disease with no treatment. Current clinical measures cannot robustly detect disease progression in less than a year, meaning that clinical trials are long and drug development is slow. The Friedreich’s Ataxia Research Alliance and the scientific community are looking for biomarkers that show change in shorter time frames that can accelerate drug development. The 2018 FARA Biomarker Meeting summarized the exciting findings that represent the current state of the field.
Collapse
|
35
|
Dong YN, McMillan E, Clark EM, Lin H, Lynch DR. GRP75 overexpression rescues frataxin deficiency and mitochondrial phenotypes in Friedreich ataxia cellular models. Hum Mol Genet 2019; 28:1594-1607. [PMID: 30590615 PMCID: PMC6494971 DOI: 10.1093/hmg/ddy448] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 11/19/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022] Open
Abstract
Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein crucial for iron-sulfur cluster biogenesis and adenosine triphosphate (ATP) production. Currently, there is no therapy to slow down the progression of FRDA. Recent evidence indicates that posttranslational regulation of residual frataxin levels can rescue some of the functional deficit of FRDA, raising the possibility of enhancing levels of residual frataxin as a treatment for FRDA. Here, we present evidence that mitochondrial molecular chaperone GRP75, also known as mortalin/mthsp70/PBP74, directly interacts with frataxin both in vivo in mouse cortex and in vitro in cortical neurons. Overexpressing GRP75 increases the levels of both wild-type frataxin and clinically relevant missense frataxin variants in human embryonic kidney 293 cells, while clinical GRP75 variants such as R126W, A476T and P509S impair the binding of GRP75 with frataxin and the effect of GRP75 on frataxin levels. In addition, GRP75 overexpression rescues frataxin deficiency and abnormal cellular phenotypes such as the abnormal mitochondrial network and decreased ATP levels in FRDA patient-derived cells. The effect of GRP75 on frataxin might be in part mediated by the physical interaction between GRP75 and mitochondrial processing peptidase (MPP), which makes frataxin more accessible to MPP. As GRP75 levels are decreased in multiple cell types of FRDA patients, restoring GRP75 might be effective in treating both typical FRDA patients with two guanine-adenine-adenine repeat expansions and compound heterozygous patients with point mutations.
Collapse
Affiliation(s)
- Yi Na Dong
- Department of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily McMillan
- Department of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elisia M Clark
- Department of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hong Lin
- Department of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David R Lynch
- Department of Pediatrics and Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
36
|
Boesch S, Indelicato E. Erythropoietin and Friedreich Ataxia: Time for a Reappraisal? Front Neurosci 2019; 13:386. [PMID: 31105516 PMCID: PMC6491891 DOI: 10.3389/fnins.2019.00386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/04/2019] [Indexed: 12/24/2022] Open
Abstract
Friedreich ataxia (FRDA) is a rare neurological disorder due to deficiency of the mitochondrial protein frataxin. Frataxin deficiency results in impaired mitochondrial function and iron deposition in affected tissues. Erythropoietin (EPO) is a cytokine which was mostly known as a key regulator of erythropoiesis until cumulative evidence showed additional neurotrophic and neuroprotective properties. These features offered the rationale for advancement of EPO in clinical trials in different neurological disorders in the past years, including FRDA. Several mechanisms of action of EPO may be beneficial in FRDA. First of all, EPO exposure results in frataxin upregulation in vitro and in vivo. By promoting erythropoiesis, EPO influences iron metabolism and induces shifts in iron pool which may ameliorate conditions of free iron excess and iron accumulation. Furthermore, EPO signaling is crucial for mitochondrial gene activation and mitochondrial biogenesis. Up to date nine clinical trials investigated the effects of EPO and derivatives in FRDA. The majority of these studies had a proof-of-concept design. Considering the natural history of FRDA, all of them were too short in duration and not powered for clinical changes. However, these studies addressed significant issues in the treatment with EPO, such as (1) the challenge of the dose finding, (2) stability of frataxin up-regulation, (3) continuous versus intermittent stimulation with EPO/regimen, or (4) tissue changes after EPO exposure in humans in vivo (muscle biopsy, brain imaging). Despite several clinical trials in the past, no treatment is available for the treatment of FRDA. Current lines of research focus on gene therapy, frataxin replacement strategies and on regulation of key metabolic checkpoints such as NrF2. Due to potential crosstalk with all these mechanisms, interventions on the EPO pathway still represent a valuable research field. The recent development of small EPO mimetics which maintain cytoprotective properties without erythropoietic action may open a new era in EPO research for the treatment of FRDA.
Collapse
Affiliation(s)
- Sylvia Boesch
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
37
|
Cortese A, Simone R, Sullivan R, Vandrovcova J, Tariq H, Yau WY, Humphrey J, Jaunmuktane Z, Sivakumar P, Polke J, Ilyas M, Tribollet E, Tomaselli PJ, Devigili G, Callegari I, Versino M, Salpietro V, Efthymiou S, Kaski D, Wood NW, Andrade NS, Buglo E, Rebelo A, Rossor AM, Bronstein A, Fratta P, Marques WJ, Züchner S, Reilly MM, Houlden H. Biallelic expansion of an intronic repeat in RFC1 is a common cause of late-onset ataxia. Nat Genet 2019; 51:649-658. [PMID: 30926972 PMCID: PMC6709527 DOI: 10.1038/s41588-019-0372-4] [Citation(s) in RCA: 355] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/12/2019] [Indexed: 12/12/2022]
Abstract
Late-onset ataxia is common, often idiopathic, and can result from cerebellar, proprioceptive, or vestibular impairment; when in combination, it is also termed cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS). We used non-parametric linkage analysis and genome sequencing to identify a biallelic intronic AAGGG repeat expansion in the replication factor C subunit 1 (RFC1) gene as the cause of familial CANVAS and a frequent cause of late-onset ataxia, particularly if sensory neuronopathy and bilateral vestibular areflexia coexist. The expansion, which occurs in the poly(A) tail of an AluSx3 element and differs in both size and nucleotide sequence from the reference (AAAAG)11 allele, does not affect RFC1 expression in patient peripheral and brain tissue, suggesting no overt loss of function. These data, along with an expansion carrier frequency of 0.7% in Europeans, implies that biallelic AAGGG expansion in RFC1 is a frequent cause of late-onset ataxia.
Collapse
Affiliation(s)
- Andrea Cortese
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
| | - Roberto Simone
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Roisin Sullivan
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Huma Tariq
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Wai Yan Yau
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Jack Humphrey
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Zane Jaunmuktane
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Prasanth Sivakumar
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - James Polke
- Neurogenetics Laboratory, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Muhammad Ilyas
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| | - Eloise Tribollet
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Pedro J Tomaselli
- Department of Neurology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Grazia Devigili
- UO Neurologia I, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan, Italy
| | | | - Maurizio Versino
- IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Vincenzo Salpietro
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Diego Kaski
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Nick W Wood
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Nadja S Andrade
- Department of Psychiatry and Behavioural Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elena Buglo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Adriana Rebelo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alexander M Rossor
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Adolfo Bronstein
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Pietro Fratta
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Wilson J Marques
- Department of Neurology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mary M Reilly
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Henry Houlden
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
- Neurogenetics Laboratory, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
38
|
Clark E, Strawser C, Schadt K, Lynch DR. Identification of a novel missense mutation in Friedreich's ataxia -FXN W 168R. Ann Clin Transl Neurol 2019; 6:812-816. [PMID: 31020006 PMCID: PMC6469249 DOI: 10.1002/acn3.728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/29/2018] [Accepted: 01/03/2019] [Indexed: 11/11/2022] Open
Abstract
Friedreich's ataxia, characterized by decreased expression of frataxin protein, is caused by GAA trinucleotide repeats within intron 1 in 98% of patients. Two percent of patients carry GAA repeats in conjunction with a point mutation. In this work, we find that frataxinW168R, a novel disease-causing missense mutation, is expressed predominantly as the intermediate frataxin42-210 form, with very little expression of mature frataxin81-210 form. Its localization to mitochondria is not impaired. Additionally, increasing frataxinW168R precursor levels do not lead to an increase in mature frataxin levels, suggesting these patients will require alternative approaches to repair frataxin processing in order to treat the disorder in a disease-modifying manner.
Collapse
Affiliation(s)
- Elisia Clark
- University of PennsylvaniaPhiladelphiaPennsylvania
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Cassandra Strawser
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - Kimberly Schadt
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| | - David R. Lynch
- University of PennsylvaniaPhiladelphiaPennsylvania
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvania
| |
Collapse
|
39
|
Nachun D, Gao F, Isaacs C, Strawser C, Yang Z, Dokuru D, Van Berlo V, Sears R, Farmer J, Perlman S, Lynch DR, Coppola G. Peripheral blood gene expression reveals an inflammatory transcriptomic signature in Friedreich's ataxia patients. Hum Mol Genet 2019; 27:2965-2977. [PMID: 29790959 PMCID: PMC6097013 DOI: 10.1093/hmg/ddy198] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Transcriptional changes in Friedreich's ataxia (FRDA), a rare and debilitating recessive Mendelian neurodegenerative disorder, have been studied in affected but inaccessible tissues-such as dorsal root ganglia, sensory neurons and cerebellum-in animal models or small patient series. However, transcriptional changes induced by FRDA in peripheral blood, a readily accessible tissue, have not been characterized in a large sample. We used differential expression, association with disability stage, network analysis and enrichment analysis to characterize the peripheral blood transcriptome and identify genes that were differentially expressed in FRDA patients (n = 418) compared with both heterozygous expansion carriers (n = 228) and controls (n = 93 739 individuals in total), or were associated with disease progression, resulting in a disease signature for FRDA. We identified a transcriptional signature strongly enriched for an inflammatory innate immune response. Future studies should seek to further characterize the role of peripheral inflammation in FRDA pathology and determine its relevance to overall disease progression.
Collapse
Affiliation(s)
- Daniel Nachun
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fuying Gao
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Charles Isaacs
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Zhongan Yang
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Deepika Dokuru
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Victoria Van Berlo
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Renee Sears
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Susan Perlman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - David R Lynch
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giovanni Coppola
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
40
|
Lynch DR, Hauser L, McCormick A, Wells M, Dong YN, McCormack S, Schadt K, Perlman S, Subramony SH, Mathews KD, Brocht A, Ball J, Perdok R, Grahn A, Vescio T, Sherman JW, Farmer JM. Randomized, double-blind, placebo-controlled study of interferon- γ 1b in Friedreich Ataxia. Ann Clin Transl Neurol 2019; 6:546-553. [PMID: 30911578 PMCID: PMC6414489 DOI: 10.1002/acn3.731] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 01/04/2023] Open
Abstract
Objective In vitro, in vivo, and open-label studies suggest that interferon gamma (IFN-γ 1b) may improve clinical features in Friedreich Ataxia through an increase in frataxin levels. The present study evaluates the efficacy and safety of IFN-γ 1b in the treatment of Friedreich Ataxia through a double-blind, multicenter, placebo-controlled trial. Methods Ninety-two subjects with FRDA between 10 and 25 years of age were enrolled. Subjects received either IFN-γ 1b or placebo for 6 months. The primary outcome measure was the modified Friedreich Ataxia Rating Scale (mFARS). Results No difference was noted between the groups after 6 months of treatment in the mFARS or secondary outcome measures. No change was noted in buccal cell or whole blood frataxin levels. However, during an open-label extension period, subjects had a more stable course than expected based on natural history data. Conclusions This study provides no direct evidence for a beneficial effect of IFN-γ1b in FRDA. The modest stabilization compared to natural history data leaves open the possibility that longer studies may demonstrate benefit.
Collapse
Affiliation(s)
- David R. Lynch
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - Lauren Hauser
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - Ashley McCormick
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - McKenzie Wells
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - Yi Na Dong
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - Shana McCormack
- Division of Endocrinology & DiabetesChildren's Hospital of PhiladelphiaPhiladelphia19104
| | - Kim Schadt
- Division of NeurologyChildren's Hospital of Philadelphia502 Abramson Research Center, 3615 Civic Center BlvdPhiladelphiaPennsylvania19104‐4318
| | - Susan Perlman
- Department of NeurologyUniversity of California Los AngelesBox 956975, 1‐167 RNRCLos AngelesCalifornia90095
| | - Sub H. Subramony
- Department of NeurologyUniversity of FloridaRoom L3‐100, McKnight Brain Institute, 1149 Newell DriveGainesvilleFlorida32611
| | - Katherine D. Mathews
- Department of Pediatrics and NeurologyUniversity of Iowa Carver College of MedicineIowa CityIowa
| | - Alicia Brocht
- Department of NeurologyUniversity of RochesterRochesterNew York14620
| | - Julie Ball
- Horizon Pharma, Inc.Lake ForestIllinois60045
| | | | - Amy Grahn
- Horizon Pharma, Inc.Lake ForestIllinois60045
| | - Tom Vescio
- Horizon Pharma, Inc.Lake ForestIllinois60045
| | | | - Jennifer M. Farmer
- Friedreich's Ataxia Research Alliance533 W Uwchlan AveDowningtownPennsylvania19335
| |
Collapse
|
41
|
Lynch DR, Schadt K, Kichula E. Etravirine in Friedreich's ataxia: Lessons from HIV? Mov Disord 2019; 34:305-306. [DOI: 10.1002/mds.27605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- David R. Lynch
- Division of NeurologyChildren's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Kimberly Schadt
- Division of NeurologyChildren's Hospital of Philadelphia Philadelphia Pennsylvania USA
| | - Elizabeth Kichula
- Division of NeurologyChildren's Hospital of Philadelphia Philadelphia Pennsylvania USA
| |
Collapse
|
42
|
Peverill RE, Donelan L, Corben LA, Delatycki MB. Differences in the determinants of right ventricular and regional left ventricular long-axis dysfunction in Friedreich ataxia. PLoS One 2018; 13:e0209410. [PMID: 30596685 PMCID: PMC6312254 DOI: 10.1371/journal.pone.0209410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/05/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative condition which also has effects on the heart. In 96% of affected individuals FRDA is due to homozygosity of a GAA repeat expansion in intron 1 of the frataxin (FXN) gene. The number of GAA repeats have been shown to relate to disease severity in FRDA, this thought to be via an inverse relationship of GAA repeat number and cellular frataxin levels. We investigated the effects of FRDA on regional long axis function of the left and right ventricles, and also the relationship of long axis systolic (s`) and early diastolic (e`) peak velocities with GAA repeat number on the shorter (GAA1) and longer FXN alleles (GAA2). METHODS The study group of 78 adult subjects (age 32±9 years) with FRDA and normal left ventricular (LV) ejection fraction were compared to 54 healthy control subjects of similar age, sex and body size. Tissue Doppler imaging (TDI) signals were recorded at the mitral annulus for measurement of s`and e`of the septal, lateral, anterior and inferior walls and at the tricuspid annulus for measurement of right ventricular (RV) s`and e`. RESULTS All the regional LV s`and e`, and both RV s`and RV e`, were lower in individuals with FRDA compared to controls (p<0.001 for all). On multivariate analysis, which included LV septal wall thickness (SWT), RV s`and RV e`were both inversely correlated with GAA1 (β = -0.32 & -0.33, respectively, p = 0.01), but not with GAA2, whereas anterior and lateral s`were both inversely correlated with GAA2 (β = -0.25 and β = -0.28, p = 0.02) but not with GAA1. Increasing SWT was the most consistent LV structural correlate of lower s`and e`, whereas age was a consistent inverse correlate of e`but not of s`. CONCLUSION There are generalized abnormalities of both LV regional and RV long axis function in FRDA, but there are also regional differences in the association of this dysfunction with the smaller and larger GAA repeats in the FXN gene.
Collapse
Affiliation(s)
- Roger E. Peverill
- Monash Cardiovascular Research Centre, MonashHeart and Department of Medicine (School of Clinical Sciences at Monash Health), Monash University and Monash Health, Clayton, Victoria, Australia
| | - Lesley Donelan
- Monash Cardiovascular Research Centre, MonashHeart and Department of Medicine (School of Clinical Sciences at Monash Health), Monash University and Monash Health, Clayton, Victoria, Australia
| | - Louise A. Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children’s Research Institute, Parkville, Victoria, Australia
| | - Martin B. Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Parkville, Victoria, Australia
| |
Collapse
|
43
|
Guo L, Wang Q, Weng L, Hauser LA, Strawser CJ, Mesaros C, Lynch DR, Blair IA. Characterization of a new N-terminally acetylated extra-mitochondrial isoform of frataxin in human erythrocytes. Sci Rep 2018; 8:17043. [PMID: 30451920 PMCID: PMC6242848 DOI: 10.1038/s41598-018-35346-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/02/2018] [Indexed: 01/18/2023] Open
Abstract
Frataxin is a highly conserved protein encoded by the frataxin (FXN) gene. The full-length 210-amino acid form of protein frataxin (1-210; isoform A) expressed in the cytosol of cells rapidly translocates to the mitochondria, where it is converted to the mature form (81-210) by mitochondrial processing peptidase. Mature frataxin (81-210) is a critically important protein because it facilitates the assembly of mitochondrial iron-sulfur cluster protein complexes such as aconitase, lipoate synthase, and succinate dehydrogenases. Decreased expression of frataxin protein is responsible for the devastating rare genetic disease of Friedreich's ataxia. The mitochondrial form of frataxin has long been thought to be present in erythrocytes even though paradoxically, erythrocytes lack mitochondria. We have discovered that erythrocyte frataxin is in fact a novel isoform of frataxin (isoform E) with 135-amino acids and an N-terminally acetylated methionine residue. There is three times as much isoform E in erythrocytes (20.9 ± 6.4 ng/mL) from the whole blood of healthy volunteers (n = 10) when compared with the mature mitochondrial frataxin present in other blood cells (7.1 ± 1.0 ng/mL). Isoform E lacks a mitochondrial targeting sequence and so is distributed to both cytosol and the nucleus when expressed in cultured cells. When extra-mitochondrial frataxin isoform E is expressed in HEK 293 cells, it is converted to a shorter isoform identical to the mature frataxin found in mitochondria, which raises the possibility that it is involved in disease etiology. The ability to specifically quantify extra-mitochondrial and mitochondrial isoforms of frataxin in whole blood will make it possible to readily follow the natural history of diseases such as Friedreich's ataxia and monitor the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Lili Guo
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Qingqing Wang
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - Liwei Weng
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Lauren A Hauser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Cassandra J Strawser
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Clementina Mesaros
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
| | - David R Lynch
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, United States
- Departments of Pediatrics and Neurology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Ian A Blair
- Penn SRP Center and Center of Excellence in Environmental Toxicology Center, Department of Systems Pharmacology and Translational Therapeutics Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States.
- Penn/CHOP Center of Excellence in Friedreich's ataxia, Philadelphia, PA, 19104, United States.
| |
Collapse
|
44
|
Role of frataxin protein deficiency and metabolic dysfunction in Friedreich ataxia, an autosomal recessive mitochondrial disease. Neuronal Signal 2018; 2:NS20180060. [PMID: 32714592 PMCID: PMC7373238 DOI: 10.1042/ns20180060] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 01/04/2023] Open
Abstract
Friedreich ataxia (FRDA) is a progressive neurodegenerative disease with developmental features caused by a genetic deficiency of frataxin, a small, nuclear-encoded mitochondrial protein. Frataxin deficiency leads to impairment of iron–sulphur cluster synthesis, and consequently, ATP production abnormalities. Based on the involvement of such processes in FRDA, initial pathophysiological hypotheses focused on reactive oxygen species (ROS) production as a key component of the mechanism. With further study, a variety of other events appear to be involved, including abnormalities of mitochondrially related metabolism and dysfunction in mitochondrial biogenesis. Consequently, present therapies focus not only on free radical damage, but also on control of metabolic abnormalities and correction of mitochondrial biogenesis. Understanding the multitude of abnormalities in FRDA thus offers possibilities for treatment of this disorder.
Collapse
|
45
|
Friedreich's Ataxia: Clinical Presentation of a Compound Heterozygote Child with a Rare Nonsense Mutation and Comparison with Previously Published Cases. Case Rep Neurol Med 2018; 2018:8587203. [PMID: 30159187 PMCID: PMC6106966 DOI: 10.1155/2018/8587203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/18/2018] [Indexed: 11/17/2022] Open
Abstract
Friedreich's ataxia is a neurodegenerative disorder associated with a GAA trinucleotide repeat expansion in intron 1 of the frataxin (FXN) gene. It is the most common autosomal recessive cerebellar ataxia, with a mean age of onset at 16 years. Nearly 95-98% of patients are homozygous for a 90-1300 GAA repeat expansion with only 2-5% demonstrating compound heterozygosity. Compound heterozygous individuals have a repeat expansion in one allele and a point mutation/deletion/insertion in the other. Compound heterozygosity and point mutations are very rare causes of Friedreich's ataxia and nonsense mutations are a further rarity among point mutations. We report a rare compound heterozygous Friedrich's ataxia patient who was found to have one expanded GAA FXN allele and a nonsense point mutation in the other. We summarize the four previously published cases of nonsense mutations and compare the phenotype to that of our patient. We compared clinical information from our patient with other nonsense FXN mutations reported in the literature. This nonsense mutation, to our knowledge, has only been described once previously; interestingly the individual was also of Cuban ancestry. A comparison with previously published cases of nonsense mutations demonstrates some common clinical characteristics.
Collapse
|
46
|
Napierala JS, Li Y, Lu Y, Lin K, Hauser LA, Lynch DR, Napierala M. Comprehensive analysis of gene expression patterns in Friedreich's ataxia fibroblasts by RNA sequencing reveals altered levels of protein synthesis factors and solute carriers. Dis Model Mech 2018; 10:1353-1369. [PMID: 29125828 PMCID: PMC5719256 DOI: 10.1242/dmm.030536] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/21/2017] [Indexed: 12/30/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease usually caused by large homozygous expansions of GAA repeat sequences in intron 1 of the frataxin (FXN) gene. FRDA patients homozygous for GAA expansions have low FXN mRNA and protein levels when compared with heterozygous carriers or healthy controls. Frataxin is a mitochondrial protein involved in iron–sulfur cluster synthesis, and many FRDA phenotypes result from deficiencies in cellular metabolism due to lowered expression of FXN. Presently, there is no effective treatment for FRDA, and biomarkers to measure therapeutic trial outcomes and/or to gauge disease progression are lacking. Peripheral tissues, including blood cells, buccal cells and skin fibroblasts, can readily be isolated from FRDA patients and used to define molecular hallmarks of disease pathogenesis. For instance, FXN mRNA and protein levels as well as FXN GAA-repeat tract lengths are routinely determined using all of these cell types. However, because these tissues are not directly involved in disease pathogenesis, their relevance as models of the molecular aspects of the disease is yet to be decided. Herein, we conducted unbiased RNA sequencing to profile the transcriptomes of fibroblast cell lines derived from 18 FRDA patients and 17 unaffected control individuals. Bioinformatic analyses revealed significantly upregulated expression of genes encoding plasma membrane solute carrier proteins in FRDA fibroblasts. Conversely, the expression of genes encoding accessory factors and enzymes involved in cytoplasmic and mitochondrial protein synthesis was consistently decreased in FRDA fibroblasts. Finally, comparison of genes differentially expressed in FRDA fibroblasts to three previously published gene expression signatures defined for FRDA blood cells showed substantial overlap between the independent datasets, including correspondingly deficient expression of antioxidant defense genes. Together, these results indicate that gene expression profiling of cells derived from peripheral tissues can, in fact, consistently reveal novel molecular pathways of the disease. When performed on statistically meaningful sample group sizes, unbiased global profiling analyses utilizing peripheral tissues are critical for the discovery and validation of FRDA disease biomarkers. Summary: Transcriptome profiling of Friedreich's ataxia fibroblasts by RNA sequencing reveals that this peripheral tissue can be used as a disease model for gene expression biomarker discovery.
Collapse
Affiliation(s)
- Jill Sergesketter Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA
| | - Yanjie Li
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA
| | - Yue Lu
- University of Texas MD Anderson Cancer Center, Department of Molecular Carcinogenesis, Center for Cancer Epigenetics, Science Park, Smithville, Texas 78957, USA
| | - Kevin Lin
- University of Texas MD Anderson Cancer Center, Department of Molecular Carcinogenesis, Center for Cancer Epigenetics, Science Park, Smithville, Texas 78957, USA
| | - Lauren A Hauser
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research Center Room 502, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research Center Room 502, Philadelphia, PA 19104, USA
| | - Marek Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA .,Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, 61-704, Poland
| |
Collapse
|
47
|
Zeitlberger A, Ging H, Nethisinghe S, Giunti P. Advances in the understanding of hereditary ataxia – implications for future patients. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1444477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Anna Zeitlberger
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Heather Ging
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Suran Nethisinghe
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Paola Giunti
- Department of Molecular Neuroscience, UCL, Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
48
|
Lin H, Magrane J, Clark EM, Halawani SM, Warren N, Rattelle A, Lynch DR. Early VGLUT1-specific parallel fiber synaptic deficits and dysregulated cerebellar circuit in the KIKO mouse model of Friedreich ataxia. Dis Model Mech 2017; 10:1529-1538. [PMID: 29259026 PMCID: PMC5769605 DOI: 10.1242/dmm.030049] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 10/30/2017] [Indexed: 01/01/2023] Open
Abstract
Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disorder with progressive ataxia that affects both the peripheral and central nervous system (CNS). While later CNS neuropathology involves loss of large principal neurons and glutamatergic and GABAergic synaptic terminals in the cerebellar dentate nucleus, early pathological changes in FRDA cerebellum remain largely uncharacterized. Here, we report early cerebellar VGLUT1 (SLC17A7)-specific parallel fiber (PF) synaptic deficits and dysregulated cerebellar circuit in the frataxin knock-in/knockout (KIKO) FRDA mouse model. At asymptomatic ages, VGLUT1 levels in cerebellar homogenates are significantly decreased, whereas VGLUT2 (SLC17A6) levels are significantly increased, in KIKO mice compared with age-matched controls. Additionally, GAD65 (GAD2) levels are significantly increased, while GAD67 (GAD1) levels remain unaltered. This suggests early VGLUT1-specific synaptic input deficits, and dysregulation of VGLUT2 and GAD65 synaptic inputs, in the cerebellum of asymptomatic KIKO mice. Immunohistochemistry and electron microscopy further show specific reductions of VGLUT1-containing PF presynaptic terminals in the cerebellar molecular layer, demonstrating PF synaptic input deficiency in asymptomatic and symptomatic KIKO mice. Moreover, the parvalbumin levels in cerebellar homogenates and Purkinje neurons are significantly reduced, but preserved in other interneurons of the cerebellar molecular layer, suggesting specific parvalbumin dysregulation in Purkinje neurons of these mice. Furthermore, a moderate loss of large principal neurons is observed in the dentate nucleus of asymptomatic KIKO mice, mimicking that of FRDA patients. Our findings thus identify early VGLUT1-specific PF synaptic input deficits and dysregulated cerebellar circuit as potential mediators of cerebellar dysfunction in KIKO mice, reflecting developmental features of FRDA in this mouse model.
Collapse
Affiliation(s)
- Hong Lin
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jordi Magrane
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Elisia M Clark
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah M Halawani
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nathan Warren
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Amy Rattelle
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Noguera ME, Aran M, Smal C, Vazquez DS, Herrera MG, Roman EA, Alaimo N, Gallo M, Santos J. Insights on the conformational dynamics of human frataxin through modifications of loop-1. Arch Biochem Biophys 2017; 636:123-137. [PMID: 29097312 DOI: 10.1016/j.abb.2017.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 10/20/2017] [Accepted: 10/28/2017] [Indexed: 02/07/2023]
Abstract
Human frataxin (FXN) is a highly conserved mitochondrial protein involved in iron homeostasis and activation of the iron-sulfur cluster assembly. FXN deficiency causes the neurodegenerative disease Friedreich's Ataxia. Here, we investigated the effect of alterations in loop-1, a stretch presumably essential for FXN function, on the conformational stability and dynamics of the native state. We generated four loop-1 variants, carrying substitutions, insertions and deletions. All of them were stable and well-folded proteins. Fast local motions (ps-ns) and slower long-range conformational dynamics (μs-ms) were altered in some mutants as judged by NMR. Particularly, loop-1 modifications impact on the dynamics of a distant region that includes residues from the β-sheet, helix α1 and the C-terminal. Remarkably, all the mutants retain the ability to activate cysteine desulfurase, even when two of them exhibit a strong decrease in iron binding, revealing a differential sensitivity of these functional features to loop-1 perturbation. Consequently, we found that even for a small and relatively rigid protein, engineering a loop segment enables to alter conformational dynamics through a long-range effect, preserving the native-state structure and important aspects of function.
Collapse
Affiliation(s)
- Martín E Noguera
- Instituto de Química y Físico-Química Biológicas, University of Buenos Aires, Junín 956, 1113AAD, Buenos Aires, Argentina
| | - Martín Aran
- The Leloir Institute Foundation and IIBBA-CONICET, Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Clara Smal
- The Leloir Institute Foundation and IIBBA-CONICET, Av. Patricias Argentinas 435, 1405 Buenos Aires, Argentina
| | - Diego S Vazquez
- Instituto de Química y Físico-Química Biológicas, University of Buenos Aires, Junín 956, 1113AAD, Buenos Aires, Argentina
| | - María Georgina Herrera
- Instituto de Química y Físico-Química Biológicas, University of Buenos Aires, Junín 956, 1113AAD, Buenos Aires, Argentina
| | - Ernesto A Roman
- Instituto de Química y Físico-Química Biológicas, University of Buenos Aires, Junín 956, 1113AAD, Buenos Aires, Argentina
| | - Nadine Alaimo
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome "Tor Vergata", Via della Ricerca Scientifica snc, 00133 Roma, Italy
| | - Mariana Gallo
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome "Tor Vergata", Via della Ricerca Scientifica snc, 00133 Roma, Italy; IRBM Science Park S.p.A., Via Pontina km 30,600., 00071 Pomezia (RM), Italy.
| | - Javier Santos
- Instituto de Química y Físico-Química Biológicas, University of Buenos Aires, Junín 956, 1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
50
|
Lin H, Magrane J, Rattelle A, Stepanova A, Galkin A, Clark EM, Dong YN, Halawani SM, Lynch DR. Early cerebellar deficits in mitochondrial biogenesis and respiratory chain complexes in the KIKO mouse model of Friedreich ataxia. Dis Model Mech 2017; 10:1343-1352. [PMID: 29125827 PMCID: PMC5719255 DOI: 10.1242/dmm.030502] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022] Open
Abstract
Friedreich ataxia (FRDA), the most common recessive inherited ataxia, results from deficiency of frataxin, a small mitochondrial protein crucial for iron-sulphur cluster formation and ATP production. Frataxin deficiency is associated with mitochondrial dysfunction in FRDA patients and animal models; however, early mitochondrial pathology in FRDA cerebellum remains elusive. Using frataxin knock-in/knockout (KIKO) mice and KIKO mice carrying the mitoDendra transgene, we show early cerebellar deficits in mitochondrial biogenesis and respiratory chain complexes in this FRDA model. At asymptomatic stages, the levels of PGC-1α (PPARGC1A), the mitochondrial biogenesis master regulator, are significantly decreased in cerebellar homogenates of KIKO mice compared with age-matched controls. Similarly, the levels of the PGC-1α downstream effectors, NRF1 and Tfam, are significantly decreased, suggesting early impaired cerebellar mitochondrial biogenesis pathways. Early mitochondrial deficiency is further supported by significant reduction of the mitochondrial markers GRP75 (HSPA9) and mitofusin-1 in the cerebellar cortex. Moreover, the numbers of Dendra-labeled mitochondria are significantly decreased in cerebellar cortex, confirming asymptomatic cerebellar mitochondrial biogenesis deficits. Functionally, complex I and II enzyme activities are significantly reduced in isolated mitochondria and tissue homogenates from asymptomatic KIKO cerebella. Structurally, levels of the complex I core subunit NUDFB8 and complex II subunits SDHA and SDHB are significantly lower than those in age-matched controls. These results demonstrate complex I and II deficiency in KIKO cerebellum, consistent with defects identified in FRDA patient tissues. Thus, our findings identify early cerebellar mitochondrial biogenesis deficits as a potential mediator of cerebellar dysfunction and ataxia, thereby providing a potential therapeutic target for early intervention of FRDA.
Collapse
Affiliation(s)
- Hong Lin
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jordi Magrane
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amy Rattelle
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anna Stepanova
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
- Queen's University Belfast, School of Biological Sciences, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Alexander Galkin
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
- Queen's University Belfast, School of Biological Sciences, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Elisia M Clark
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Na Dong
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah M Halawani
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|