1
|
Saha S, Corben LA, Selvadurai LP, Harding IH, Georgiou-Karistianis N. Predictive machine learning and multimodal data to develop highly sensitive, composite biomarkers of disease progression in Friedreich ataxia. Sci Rep 2025; 15:17629. [PMID: 40399385 PMCID: PMC12095658 DOI: 10.1038/s41598-025-01047-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025] Open
Abstract
Friedreich ataxia (FRDA) is a rare, inherited progressive movement disorder for which there is currently no cure. The field urgently requires more sensitive, objective, and clinically relevant biomarkers to enhance the evaluation of treatment efficacy in clinical trials and to speed up the process of drug discovery. This study pioneers the development of clinically relevant, multidomain, fully objective composite biomarkers of disease severity and progression, using multimodal neuroimaging and background data (i.e., demographic, disease history, genetics). Data from 31 individuals with FRDA and 31 controls from a longitudinal multimodal natural history study IMAGE-FRDA, were included. Using an elasticnet predictive machine learning (ML) regression model, we derived a weighted combination of background, structural MRI, diffusion MRI, and quantitative susceptibility imaging (QSM) measures that predicted Friedreich ataxia rating scale (FARS) with high accuracy (R2 = 0.79, root mean square error (RMSE) = 13.19). This composite also exhibited strong sensitivity to disease progression over two years (Cohen's d = 1.12), outperforming the sensitivity of the FARS score alone (d = 0.88). The approach was validated using the Scale for the assessment and rating of ataxia (SARA), demonstrating the potential and robustness of ML-derived composites to surpass individual biomarkers and act as complementary or surrogate markers of disease severity and progression. However, further validation, refinement, and the integration of additional data modalities will open up new opportunities for translating these biomarkers into clinical practice and clinical trials for FRDA, as well as other rare neurodegenerative diseases.
Collapse
Affiliation(s)
- Susmita Saha
- School of Psychological Sciences, The Turner Institute for Brain and Mental Health, Monash University, 18 innovation walk, Clayton campus, Clayton, Victoria, Australia
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
| | - Louise A Corben
- School of Psychological Sciences, The Turner Institute for Brain and Mental Health, Monash University, 18 innovation walk, Clayton campus, Clayton, Victoria, Australia
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Louisa P Selvadurai
- School of Psychological Sciences, The Turner Institute for Brain and Mental Health, Monash University, 18 innovation walk, Clayton campus, Clayton, Victoria, Australia
| | - Ian H Harding
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences, The Turner Institute for Brain and Mental Health, Monash University, 18 innovation walk, Clayton campus, Clayton, Victoria, Australia.
| |
Collapse
|
2
|
Zhang Y, Hulsman M, Salazar A, Tesi N, Knoop L, van der Lee S, Wijesekera S, Krizova J, Kamsteeg EJ, Holstege H. Multisample motif discovery and visualization for tandem repeats. Genome Res 2025; 35:850-862. [PMID: 39537359 PMCID: PMC12047238 DOI: 10.1101/gr.279278.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Tandem repeats (TRs) occupy a significant portion of the human genome and are a source of polymorphisms due to variations in sizes and motif compositions. Some of these variations have been associated with various neuropathological disorders, highlighting the clinical importance of assessing the motif structure of TRs. Moreover, assessing the TR motif variation can offer valuable insights into evolutionary dynamics and population structure. Previously, characterizations of TRs were limited by short-read sequencing technology, which lacks the ability to accurately capture the full TR sequences. As long-read sequencing becomes more accessible and can capture the full complexity of TRs, there is now also a need for tools to characterize and analyze TRs using long-read data across multiple samples. In this study, we present MotifScope, a novel algorithm for the characterization and visualization of TRs based on a de novo k-mer approach for motif discovery. Comparative analysis against established tools reveals that MotifScope can identify a greater number of motifs and more accurately represent the underlying repeat sequences. Moreover, MotifScope has been specifically designed to enable motif composition comparisons across assemblies of different individuals, as well as across long-read sequencing reads within an individual, through combined motif discovery and sequence alignment. We showcase potential applications of MotifScope in diverse fields, including population genetics, clinical settings, and forensic analyses.
Collapse
Affiliation(s)
- Yaran Zhang
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Marc Hulsman
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
| | - Alex Salazar
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Niccolò Tesi
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
| | - Lydian Knoop
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081HV Amsterdam, The Netherlands
| | - Sanduni Wijesekera
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Jana Krizova
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands
| | - Henne Holstege
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081HV Amsterdam, The Netherlands;
- Delft Bioinformatics Lab, Delft University of Technology, 2628CD Delft, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
3
|
Joseph DJ, Mercado-Ayon E, Flatley L, Viaene AN, Hordeaux J, Marsh ED, Lynch DR. Functional Characterization of Parallel Fiber-Purkinje Cell Synapses in Two Friedreich's Ataxia Mouse Models. CEREBELLUM (LONDON, ENGLAND) 2025; 24:42. [PMID: 39907933 PMCID: PMC11799031 DOI: 10.1007/s12311-025-01796-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Friedreich ataxia (FRDA) is an autosomal recessive disorder caused by GAA expansions in the FXN gene, which codes for the protein frataxin (FXN). These mutations reduce FXN expression, leading to mitochondrial dysfunction and multisystemic disease. Accumulating evidence suggests that neuronal dysfunction, rather than neuronal death, may drive the neurological phenotypes of FRDA, but the mechanisms underlying such neurological phenotypes remain unclear. To investigate the neural circuit basis of this dysfunction, we employed field recordings to measure Purkinje cell (PC) function and synaptic properties along with western blotting and immunohistochemistry to determine their density and structure in two established FRDA mouse models, the shRNA-frataxin (FRDAkd) and the frataxin knock in-knockout (KIKO) mice. Western blotting demonstrated subtle changes in mitochondrial proteins and only a modest reduction in the density of calbindin positive cells PCs in the cerebellar cortex of the FRDAkd mice, with no change in the density of PCs in the KIKO mice. Though PC density differed slightly in the two models, field recordings of parallel fiber-PC synapses in the molecular layer demonstrated concordant hypo-excitability of basal synaptic transmission and impairments of long-term plasticity using induction protocols associated with both potentiation and depression of synaptic strength. These results indicate that synaptic instability might be a common feature in FRDA mouse models.
Collapse
Affiliation(s)
- Donald J Joseph
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elizabeth Mercado-Ayon
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liam Flatley
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Angela N Viaene
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Juliette Hordeaux
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Eric D Marsh
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David R Lynch
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Pediatrics and Neurology, Perelman School of Medicine, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Dong TR, Li YJ, Jin SY, Yang FL, Xiong RX, Dai YQ, Song XZ, Guan CP. Progress on mitochondria and hair follicle development in androgenetic alopecia: relationships and therapeutic perspectives. Stem Cell Res Ther 2025; 16:44. [PMID: 39901201 PMCID: PMC11792644 DOI: 10.1186/s13287-025-04182-z] [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: 11/13/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
Hair loss has long been a significant concern for many individuals. Recent studies have indicated that mitochondria play a more crucial role in hair loss than previously recognized. This review summarizes the connection between mitochondrial dysfunction and hair follicle development, outlines the links between diseases related to mitochondrial disorders and hair issues, and highlights the influence of mitochondrial dysfunction on androgenetic alopecia. We discuss the cellular and signaling mechanisms associated with hair loss and examine how mitochondrial dysfunction, such as insufficient energy supply, signaling irregularities, protein/gene abnormalities, and programmed cell death, can hinder the normal proliferation, differentiation, and growth of hair follicle cells. Furthermore, we discuss current treatment approaches and potential innovative therapies, including mitochondrion-targeting drugs and advanced techniques that directly target hair follicle cells, providing fresh insights into the crucial role of mitochondria in maintaining hair follicle health and managing hair disorders. Furthermore, this review explores future therapeutic strategies and proposes that mitochondrial research could lead to groundbreaking treatments for hair loss, thus providing optimism and new avenues for the treatment of individuals experiencing hair loss. This review not only underscores the central importance of mitochondria in hair health but also emphasizes the importance of advancing research and treatment in this field.
Collapse
Affiliation(s)
- Ting-Ru Dong
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Yu-Jie Li
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Shi-Yu Jin
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Feng-Lan Yang
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Ren-Xue Xiong
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Ye-Qin Dai
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Xiu-Zu Song
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China
| | - Cui-Ping Guan
- Department of Dermatology, Hangzhou Third Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310009, China.
- Department of Dermatology, Hangzhou Third People's Hospital, No 38 Xihu Rd, Hangzhou, 310009, China.
| |
Collapse
|
5
|
Allowitz K, Taylor J, Harames K, Yoo J, Baloch O, Ramana KV. Oxidative Stress-mediated Lipid Peroxidation-derived Lipid Aldehydes in the Pathophysiology of Neurodegenerative Diseases. Curr Neuropharmacol 2025; 23:671-685. [PMID: 39440770 DOI: 10.2174/011570159x342720241014164650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Neurodegenerative diseases such as Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis cause damage and gradual loss of neurons affecting the central nervous system. Neurodegenerative diseases are most commonly seen in the ageing process. Ageing causes increased reactive oxygen species and decreased mitochondrial ATP generation, resulting in redox imbalance and oxidative stress. Oxidative stress-generated free radicals cause damage to membrane lipids containing polyunsaturated fatty acids, leading to the formation of toxic lipid aldehyde products such as 4- hydroxynonenal and malondialdehyde. Several studies have shown that lipid peroxidation-derived aldehyde products form adducts with cellular proteins, altering their structure and function. Thus, these lipid aldehydes could act as secondary signaling intermediates, modifying important metabolic pathways, and contributing to the pathophysiology of several human diseases, including neurodegenerative disorders. Additionally, they could serve as biomarkers for disease progression. This narrative review article discusses the biological and clinical significance of oxidative stress-mediated lipid peroxidation-derived lipid aldehydes in the pathophysiology of various neurodegenerative diseases.
Collapse
Affiliation(s)
- Kieran Allowitz
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - Justin Taylor
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - Kyra Harames
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - John Yoo
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - Omar Baloch
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| | - Kota V Ramana
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT-84606, USA
| |
Collapse
|
6
|
Marullo C, Croci L, Giupponi I, Rivoletti C, Zuffetti S, Bettegazzi B, Cremona O, Giunti P, Ambrosi A, Casoni F, Consalez GG, Codazzi F. Altered Ca2+ responses and antioxidant properties in Friedreich's ataxia-like cerebellar astrocytes. J Cell Sci 2025; 138:jcs263446. [PMID: 39648860 PMCID: PMC11828468 DOI: 10.1242/jcs.263446] [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: 07/22/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disorder characterized by severe neurological signs, affecting the peripheral and central nervous system, caused by reduced frataxin protein (FXN) levels. Although several studies have highlighted cellular dysfunctions in neurons, there is limited information on the effects of FXN depletion in astrocytes and on the potential non-cell autonomous mechanisms affecting neurons in FRDA. In this study, we generated a model of FRDA cerebellar astrocytes to unveil phenotypic alterations that might contribute to cerebellar atrophy. We treated primary cerebellar astrocytes with an RNA interference-based approach, to achieve a reduction of FXN comparable to that observed in individuals with FRDA. These FRDA-like astrocytes display some typical features of the disease, such as an increase of oxidative stress and a depletion of glutathione content. Moreover, FRDA-like astrocytes exhibit decreased Ca2+ responses to purinergic stimuli. Our findings shed light on cellular changes caused by FXN downregulation in cerebellar astrocytes, likely impairing their complex interaction with neurons. The potentially impaired ability to provide neuronal cells with glutathione or to release neuromodulators in a Ca2+-dependent manner could affect neuronal function, contributing to neurodegeneration.
Collapse
Affiliation(s)
- Chiara Marullo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Croci
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Iris Giupponi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia Rivoletti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sofia Zuffetti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Barbara Bettegazzi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ottavio Cremona
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Paola Giunti
- Ataxia Centre, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alessandro Ambrosi
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Filippo Casoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Gian Giacomo Consalez
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| | - Franca Codazzi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Faculty of Medicine and Surgery, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
7
|
Mosbach V, Puccio H. A multiple animal and cellular models approach to study frataxin deficiency in Friedreich Ataxia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119809. [PMID: 39134123 DOI: 10.1016/j.bbamcr.2024.119809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
Friedreich's ataxia (FA) is one of the most frequent inherited recessive ataxias characterized by a progressive sensory and spinocerebellar ataxia. The main causative mutation is a GAA repeat expansion in the first intron of the frataxin (FXN) gene which leads to a transcriptional silencing of the gene resulting in a deficit in FXN protein. The nature of the mutation (an unstable GAA expansion), as well as the multi-systemic nature of the disease (with neural and non-neural sites affected) make the generation of models for Friedreich's ataxia quite challenging. Over the years, several cellular and animal models for FA have been developed. These models are all complementary and possess their own strengths to investigate different aspects of the disease, such as the epigenetics of the locus or the pathophysiology of the disease, as well as being used to developed novel therapeutic approaches. This review will explore the recent advancements in the different mammalian models developed for FA.
Collapse
Affiliation(s)
- Valentine Mosbach
- Institut NeuroMyoGene-PGNM UCBL-CNRS UMR5261 INSERM U1315, Lyon, France
| | - Hélène Puccio
- Institut NeuroMyoGene-PGNM UCBL-CNRS UMR5261 INSERM U1315, Lyon, France.
| |
Collapse
|
8
|
Menon PJ, Yi TX, Moran S, Walsh RA, Murphy SM, Bogdanova-Mihaylova P. Health-Related Quality of Life in Patients with Inherited Ataxia in Ireland. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1328-1337. [PMID: 38010570 DOI: 10.1007/s12311-023-01640-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Inherited cerebellar ataxias (CA) are heterogeneous progressive neurological conditions associated with significant functional limitations. This study aimed to assess the implications of inherited CA on patients' self-reported quality of life (QoL) and impairments in work and activities. 129 individuals with ataxia responded to a survey focused on QoL. Health-related QoL was measured using the RAND 36-Item Short Form Survey. An adaptation of the validated Work Productivity and Activity Impairment questionnaire was used to assess the effect of health on work productivity and ability to perform activities over the past week. Nine percent of respondents were currently employed. Individuals with inherited ataxia experienced significant activity impairment, and 75% required professional or informal care. Health-related quality of life (HRQoL) was significantly worse in all areas for the individuals with inherited ataxia compared with Irish population normative values. Participants with Friedreich's ataxia (n = 56) demonstrated worse physical functioning then those with undetermined ataxia (n = 55). Female gender, younger age at symptom onset, current employment, retirement due to age or ataxia, and living in a long-term care facility were associated with higher sub-scores in different domains of HRQoL, while disease duration correlated with worse physical functioning sub-scores. This study is the first cross-sectional study on HRQoL in patients with inherited ataxia in Ireland. It highlights high rates of unemployment, difficulty with daily activities and physical functioning limitations, which is worse than comparative international studies. Given the limited therapeutic options currently available, optimising HRQoL is an important aspect of managing ataxia.
Collapse
Affiliation(s)
- Poornima Jayadev Menon
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland.
- School of Postgraduate Studies, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Tan Xin Yi
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sharon Moran
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
| | - Richard A Walsh
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | - Sinéad M Murphy
- Department of Neurology, Tallaght University Hospital, Dublin, Ireland
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | | |
Collapse
|
9
|
Ilg W, Milne S, Schmitz-Hübsch T, Alcock L, Beichert L, Bertini E, Mohamed Ibrahim N, Dawes H, Gomez CM, Hanagasi H, Kinnunen KM, Minnerop M, Németh AH, Newman J, Ng YS, Rentz C, Samanci B, Shah VV, Summa S, Vasco G, McNames J, Horak FB. Quantitative Gait and Balance Outcomes for Ataxia Trials: Consensus Recommendations by the Ataxia Global Initiative Working Group on Digital-Motor Biomarkers. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1566-1592. [PMID: 37955812 PMCID: PMC11269489 DOI: 10.1007/s12311-023-01625-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/14/2023]
Abstract
With disease-modifying drugs on the horizon for degenerative ataxias, ecologically valid, finely granulated, digital health measures are highly warranted to augment clinical and patient-reported outcome measures. Gait and balance disturbances most often present as the first signs of degenerative cerebellar ataxia and are the most reported disabling features in disease progression. Thus, digital gait and balance measures constitute promising and relevant performance outcomes for clinical trials.This narrative review with embedded consensus will describe evidence for the sensitivity of digital gait and balance measures for evaluating ataxia severity and progression, propose a consensus protocol for establishing gait and balance metrics in natural history studies and clinical trials, and discuss relevant issues for their use as performance outcomes.
Collapse
Affiliation(s)
- Winfried Ilg
- Section Computational Sensomotorics, Hertie Institute for Clinical Brain Research, Otfried-Müller-Straße 25, 72076, Tübingen, Germany.
- Centre for Integrative Neuroscience (CIN), Tübingen, Germany.
| | - Sarah Milne
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, Melbourne University, Melbourne, VIC, Australia
- Physiotherapy Department, Monash Health, Clayton, VIC, Australia
- School of Primary and Allied Health Care, Monash University, Frankston, VIC, Australia
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a cooperation of Max-Delbrueck Center for Molecular Medicine and Charité, Universitätsmedizin Berlin, Berlin, Germany
- Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lisa Alcock
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Lukas Beichert
- Department of Neurodegenerative Diseases and Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital, IRCCS, Rome, Italy
| | | | - Helen Dawes
- NIHR Exeter BRC, College of Medicine and Health, University of Exeter, Exeter, UK
| | | | - Hasmet Hanagasi
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | - Martina Minnerop
- Institute of Neuroscience and Medicine (INM-1)), Research Centre Juelich, Juelich, Germany
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Neurology, Center for Movement Disorders and Neuromodulation, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea H Németh
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jane Newman
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, UK
| | - Clara Rentz
- Institute of Neuroscience and Medicine (INM-1)), Research Centre Juelich, Juelich, Germany
| | - Bedia Samanci
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Vrutangkumar V Shah
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- APDM Precision Motion, Clario, Portland, OR, USA
| | - Susanna Summa
- Movement Analysis and Robotics Laboratory (MARLab), Neurorehabilitation Unit, Neurological Science and Neurorehabilitation Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gessica Vasco
- Movement Analysis and Robotics Laboratory (MARLab), Neurorehabilitation Unit, Neurological Science and Neurorehabilitation Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - James McNames
- APDM Precision Motion, Clario, Portland, OR, USA
- Department of Electrical and Computer Engineering, Portland State University, Portland, OR, USA
| | - Fay B Horak
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- APDM Precision Motion, Clario, Portland, OR, USA
| |
Collapse
|
10
|
Lai Y, Diaz N, Armbrister R, Agoulnik I, Liu Y. DNA Base Damage Repair Crosstalks with Chromatin Structures to Contract Expanded GAA Repeats in Friedreich's Ataxia. Biomolecules 2024; 14:809. [PMID: 39062522 PMCID: PMC11274795 DOI: 10.3390/biom14070809] [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: 06/04/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Trinucleotide repeat (TNR) expansion is the cause of over 40 neurodegenerative diseases, including Huntington's disease and Friedreich's ataxia (FRDA). There are no effective treatments for these diseases due to the poor understanding of molecular mechanisms underlying somatic TNR expansion and contraction in neural systems. We and others have found that DNA base excision repair (BER) actively modulates TNR instability, shedding light on the development of effective treatments for the diseases by contracting expanded repeats through DNA repair. In this study, temozolomide (TMZ) was employed as a model DNA base damaging agent to reveal the mechanisms of the BER pathway in modulating GAA repeat instability at the frataxin (FXN) gene in FRDA neural cells and transgenic mouse mice. We found that TMZ induced large GAA repeat contraction in FRDA mouse brain tissue, neurons, and FRDA iPSC-differentiated neural cells, increasing frataxin protein levels in FRDA mouse brain and neural cells. Surprisingly, we found that TMZ could also inhibit H3K9 methyltransferases, leading to open chromatin and increasing ssDNA breaks and recruitment of the key BER enzyme, pol β, on the repeats in FRDA neural cells. We further demonstrated that the H3K9 methyltransferase inhibitor BIX01294 also induced the contraction of the expanded repeats and increased frataxin protein in FRDA neural cells by opening the chromatin and increasing the endogenous ssDNA breaks and recruitment of pol β on the repeats. Our study provides new mechanistic insight illustrating that inhibition of H3K9 methylation can crosstalk with BER to induce GAA repeat contraction in FRDA. Our results will open a new avenue for developing novel gene therapy by targeting histone methylation and the BER pathway for repeat expansion diseases.
Collapse
Affiliation(s)
- Yanhao Lai
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (Y.L.); (N.D.)
| | - Nicole Diaz
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (Y.L.); (N.D.)
| | - Rhyisa Armbrister
- Biochemistry Ph.D. Program, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (R.A.); (I.A.)
| | - Irina Agoulnik
- Biochemistry Ph.D. Program, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (R.A.); (I.A.)
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Yuan Liu
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (Y.L.); (N.D.)
- Biochemistry Ph.D. Program, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (R.A.); (I.A.)
- Biomolecular Sciences Institute, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| |
Collapse
|
11
|
Stovickova L, Hansikova H, Hanzalova J, Musova Z, Semjonov V, Stovicek P, Hadzic H, Novotna L, Simcik M, Strnad P, Serbina A, Karamazovova S, Schwabova Paulasova J, Vyhnalek M, Krsek P, Zumrova A. Exploring mitochondrial biomarkers for Friedreich's ataxia: a multifaceted approach. J Neurol 2024; 271:3439-3454. [PMID: 38520521 PMCID: PMC11136723 DOI: 10.1007/s00415-024-12223-5] [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: 12/12/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/25/2024]
Abstract
This study presents an in-depth analysis of mitochondrial enzyme activities in Friedreich's ataxia (FA) patients, focusing on the Electron Transport Chain complexes I, II, and IV, the Krebs Cycle enzyme Citrate Synthase, and Coenzyme Q10 levels. It examines a cohort of 34 FA patients, comparing their mitochondrial enzyme activities and clinical parameters, including disease duration and cardiac markers, with those of 17 healthy controls. The findings reveal marked reductions in complexes II and, specifically, IV, highlighting mitochondrial impairment in FA. Additionally, elevated Neurofilament Light Chain levels and cardiomarkers were observed in FA patients. This research enhances our understanding of FA pathophysiology and suggests potential biomarkers for monitoring disease progression. The study underscores the need for further clinical trials to validate these findings, emphasizing the critical role of mitochondrial dysfunction in FA assessment and treatment.
Collapse
Affiliation(s)
- Lucie Stovickova
- Department of Paediatric Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, V Uvalu 84, 15006, Prague 5, Czech Republic.
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic.
| | - Hana Hansikova
- Department of Paediatrics and Inherited Metabolic Disorders, First Medical Faculty, Charles University and General University Hospital in Prague, Prague 2, Czech Republic
| | - Jitka Hanzalova
- Department of Immunology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague 5, Czech Republic
| | - Zuzana Musova
- Department of Biology and Medical Genetics, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague 5, Czech Republic
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic
| | - Valerij Semjonov
- Department of Paediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | | - Haris Hadzic
- Second Faculty of Medicine, Charles University, Prague 5, Czech Republic
| | - Ludmila Novotna
- Second Faculty of Medicine, Charles University, Prague 5, Czech Republic
| | - Martin Simcik
- Second Faculty of Medicine, Charles University, Prague 5, Czech Republic
| | - Pavel Strnad
- Second Faculty of Medicine, Charles University, Prague 5, Czech Republic
| | - Anastaziia Serbina
- Second Faculty of Medicine, Charles University, Prague 5, Czech Republic
| | - Simona Karamazovova
- Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague 5, Czech Republic
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic
| | - Jaroslava Schwabova Paulasova
- Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague 5, Czech Republic
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic
| | - Martin Vyhnalek
- Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague 5, Czech Republic
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic
| | - Pavel Krsek
- Department of Paediatric Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, V Uvalu 84, 15006, Prague 5, Czech Republic
| | - Alena Zumrova
- Department of Paediatric Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, V Uvalu 84, 15006, Prague 5, Czech Republic
- Centre of Hereditary Ataxias, Second Faculty of Medicine, An Official EFACTS Site, a Member of European Reference Network for Rare Neurological Diseases (ERN-RND), Charles University, Motol University Hospital, Prague 5, Czech Republic
| |
Collapse
|
12
|
Krut' VG, Kalinichenko AL, Maltsev DI, Jappy D, Shevchenko EK, Podgorny OV, Belousov VV. Optogenetic and chemogenetic approaches for modeling neurological disorders in vivo. Prog Neurobiol 2024; 235:102600. [PMID: 38548126 DOI: 10.1016/j.pneurobio.2024.102600] [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: 08/15/2023] [Revised: 02/26/2024] [Accepted: 03/22/2024] [Indexed: 04/01/2024]
Abstract
Animal models of human neurological disorders provide valuable experimental tools which enable us to study various aspects of disorder pathogeneses, ranging from structural abnormalities and disrupted metabolism and signaling to motor and mental deficits, and allow us to test novel therapies in preclinical studies. To be valid, these animal models should recapitulate complex pathological features at the molecular, cellular, tissue, and behavioral levels as closely as possible to those observed in human subjects. Pathological states resembling known human neurological disorders can be induced in animal species by toxins, genetic factors, lesioning, or exposure to extreme conditions. In recent years, novel animal models recapitulating neuropathologies in humans have been introduced. These animal models are based on synthetic biology approaches: opto- and chemogenetics. In this paper, we review recent opto- and chemogenetics-based animal models of human neurological disorders. These models allow for the creation of pathological states by disrupting specific processes at the cellular level. The artificial pathological states mimic a range of human neurological disorders, such as aging-related dementia, Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, epilepsy, and ataxias. Opto- and chemogenetics provide new opportunities unavailable with other animal models of human neurological disorders. These techniques enable researchers to induce neuropathological states varying in severity and ranging from acute to chronic. We also discuss future directions for the development and application of synthetic biology approaches for modeling neurological disorders.
Collapse
Affiliation(s)
- Viktoriya G Krut'
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Andrei L Kalinichenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Dmitry I Maltsev
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Evgeny K Shevchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia
| | - Oleg V Podgorny
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia.
| | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow 117997, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow 117997, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow 143025, Russia.
| |
Collapse
|
13
|
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
|
14
|
Mishra P, Sivakumar A, Johnson A, Pernaci C, Warden AS, El-Hachem LR, Hansen E, Badell-Grau RA, Khare V, Ramirez G, Gillette S, Solis AB, Guo P, Coufal N, Cherqui S. Gene editing improves endoplasmic reticulum-mitochondrial contacts and unfolded protein response in Friedreich's ataxia iPSC-derived neurons. Front Pharmacol 2024; 15:1323491. [PMID: 38420191 PMCID: PMC10899513 DOI: 10.3389/fphar.2024.1323491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024] Open
Abstract
Friedreich ataxia (FRDA) is a multisystemic, autosomal recessive disorder caused by homozygous GAA expansion mutation in the first intron of frataxin (FXN) gene. FXN is a mitochondrial protein critical for iron-sulfur cluster biosynthesis and deficiency impairs mitochondrial electron transport chain functions and iron homeostasis within the organelle. Currently, there is no effective treatment for FRDA. We have previously demonstrated that single infusion of wild-type hematopoietic stem and progenitor cells (HSPCs) resulted in prevention of neurologic and cardiac complications of FRDA in YG8R mice, and rescue was mediated by FXN transfer from tissue engrafted, HSPC-derived microglia/macrophages to diseased neurons/myocytes. For a future clinical translation, we developed an autologous stem cell transplantation approach using CRISPR/Cas9 for the excision of the GAA repeats in FRDA patients' CD34+ HSPCs; this strategy leading to increased FXN expression and improved mitochondrial functions. The aim of the current study is to validate the efficiency and safety of our gene editing approach in a disease-relevant model. We generated a cohort of FRDA patient-derived iPSCs and isogenic lines that were gene edited with our CRISPR/Cas9 approach. iPSC derived FRDA neurons displayed characteristic apoptotic and mitochondrial phenotype of the disease, such as non-homogenous microtubule staining in neurites, increased caspase-3 expression, mitochondrial superoxide levels, mitochondrial fragmentation, and partial degradation of the cristae compared to healthy controls. These defects were fully prevented in the gene edited neurons. RNASeq analysis of FRDA and gene edited neurons demonstrated striking improvement in gene clusters associated with endoplasmic reticulum (ER) stress in the isogenic lines. Gene edited neurons demonstrated improved ER-calcium release, normalization of ER stress response gene, XBP-1, and significantly increased ER-mitochondrial contacts that are critical for functional homeostasis of both organelles, as compared to FRDA neurons. Ultrastructural analysis for these contact sites displayed severe ER structural damage in FRDA neurons, that was undetected in gene edited neurons. Taken together, these results represent a novel finding for disease pathogenesis showing dramatic ER structural damage in FRDA, validate the efficacy profile of our FXN gene editing approach in a disease relevant model, and support our approach as an effective strategy for therapeutic intervention for Friedreich's ataxia.
Collapse
Affiliation(s)
- Priyanka Mishra
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Anusha Sivakumar
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Avalon Johnson
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Carla Pernaci
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Anna S. Warden
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Lilas Rony El-Hachem
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Emily Hansen
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Rafael A. Badell-Grau
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Veenita Khare
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Gabriela Ramirez
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Sydney Gillette
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Angelyn B. Solis
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Peng Guo
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
| | - Nicole Coufal
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
15
|
Pilotto F, Del Bondio A, Puccio H. Hereditary Ataxias: From Bench to Clinic, Where Do We Stand? Cells 2024; 13:319. [PMID: 38391932 PMCID: PMC10886822 DOI: 10.3390/cells13040319] [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: 12/01/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Cerebellar ataxias are a wide heterogeneous group of movement disorders. Within this broad umbrella of diseases, there are both genetics and sporadic forms. The clinical presentation of these conditions can exhibit a diverse range of symptoms across different age groups, spanning from pure cerebellar manifestations to sensory ataxia and multisystemic diseases. Over the last few decades, advancements in our understanding of genetics and molecular pathophysiology related to both dominant and recessive ataxias have propelled the field forward, paving the way for innovative therapeutic strategies aimed at preventing and arresting the progression of these diseases. Nevertheless, the rarity of certain forms of ataxia continues to pose challenges, leading to limited insights into the etiology of the disease and the identification of target pathways. Additionally, the lack of suitable models hampers efforts to comprehensively understand the molecular foundations of disease's pathophysiology and test novel therapeutic interventions. In the following review, we describe the epidemiology, symptomatology, and pathological progression of hereditary ataxia, including both the prevalent and less common forms of these diseases. Furthermore, we illustrate the diverse molecular pathways and therapeutic approaches currently undergoing investigation in both pre-clinical studies and clinical trials. Finally, we address the existing and anticipated challenges within this field, encompassing both basic research and clinical endeavors.
Collapse
Affiliation(s)
| | | | - Hélène Puccio
- Institut Neuromyogène, Pathophysiology and Genetics of Neuron and Muscle, Inserm U1315, CNRS-Université Claude Bernard Lyon 1 UMR5261, 69008 Lyon, France
| |
Collapse
|
16
|
Doni D, Cavallari E, Noguera ME, Gentili HG, Cavion F, Parisi G, Fornasari MS, Sartori G, Santos J, Bellanda M, Carbonera D, Costantini P, Bortolus M. Searching for Frataxin Function: Exploring the Analogy with Nqo15, the Frataxin-like Protein of Respiratory Complex I from Thermus thermophilus. Int J Mol Sci 2024; 25:1912. [PMID: 38339189 PMCID: PMC10855754 DOI: 10.3390/ijms25031912] [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: 12/23/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
Nqo15 is a subunit of respiratory complex I of the bacterium Thermus thermophilus, with strong structural similarity to human frataxin (FXN), a protein involved in the mitochondrial disease Friedreich's ataxia (FRDA). Recently, we showed that the expression of recombinant Nqo15 can ameliorate the respiratory phenotype of FRDA patients' cells, and this prompted us to further characterize both the Nqo15 solution's behavior and its potential functional overlap with FXN, using a combination of in silico and in vitro techniques. We studied the analogy of Nqo15 and FXN by performing extensive database searches based on sequence and structure. Nqo15's folding and flexibility were investigated by combining nuclear magnetic resonance (NMR), circular dichroism, and coarse-grained molecular dynamics simulations. Nqo15's iron-binding properties were studied using NMR, fluorescence, and specific assays and its desulfurase activation by biochemical assays. We found that the recombinant Nqo15 isolated from complex I is monomeric, stable, folded in solution, and highly dynamic. Nqo15 does not share the iron-binding properties of FXN or its desulfurase activation function.
Collapse
Affiliation(s)
- Davide Doni
- Department of Biology, University of Padova, 35121 Padova, Italy; (D.D.); (F.C.)
| | - Eva Cavallari
- Department of Biology, University of Padova, 35121 Padova, Italy; (D.D.); (F.C.)
- Grenoble Alpes University, CNRS, CEA, INRAE, IRIG-LPCV, 38000 Grenoble, France
| | - Martin Ezequiel Noguera
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Intendente Güiraldes 2160, Buenos Aires C1428EG, Argentina; (M.E.N.); (H.G.G.); (J.S.)
- Institute of Biological Chemistry and Physical Chemistry, Dr Alejandro Paladini (UBA-CONICET), University of Buenos Aires, Junín 956, Buenos Aires 1113AAD, Argentina
- Department of Science and Technology, National University of Quilmes, Roque Saenz Peña 352, Bernal B1876BXD, Argentina; (G.P.); (M.S.F.)
| | - Hernan Gustavo Gentili
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Intendente Güiraldes 2160, Buenos Aires C1428EG, Argentina; (M.E.N.); (H.G.G.); (J.S.)
| | - Federica Cavion
- Department of Biology, University of Padova, 35121 Padova, Italy; (D.D.); (F.C.)
| | - Gustavo Parisi
- Department of Science and Technology, National University of Quilmes, Roque Saenz Peña 352, Bernal B1876BXD, Argentina; (G.P.); (M.S.F.)
| | - Maria Silvina Fornasari
- Department of Science and Technology, National University of Quilmes, Roque Saenz Peña 352, Bernal B1876BXD, Argentina; (G.P.); (M.S.F.)
| | - Geppo Sartori
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy;
| | - Javier Santos
- Department of Physiology and Molecular and Cellular Biology, Institute of Biosciences, Biotechnology and Translational Biology (iB3), Faculty of Exact and Natural Sciences, University of Buenos Aires, Intendente Güiraldes 2160, Buenos Aires C1428EG, Argentina; (M.E.N.); (H.G.G.); (J.S.)
| | - Massimo Bellanda
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy; (M.B.); (D.C.)
- Consiglio Nazionale delle Ricerche Institute of Biomolecular Chemistry, 35131 Padova, Italy
| | - Donatella Carbonera
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy; (M.B.); (D.C.)
| | - Paola Costantini
- Department of Biology, University of Padova, 35121 Padova, Italy; (D.D.); (F.C.)
| | - Marco Bortolus
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy; (M.B.); (D.C.)
| |
Collapse
|
17
|
Perfitt TL, Huichalaf C, Gooch R, Kuperman A, Ahn Y, Chen X, Ullas S, Hirenallur-Shanthappa D, Zhan Y, Otis D, Whiteley LO, Bulawa C, Martelli A. A modified mouse model of Friedreich's ataxia with conditional Fxn allele homozygosity delays onset of cardiomyopathy. Am J Physiol Heart Circ Physiol 2024; 326:H357-H369. [PMID: 38038720 DOI: 10.1152/ajpheart.00496.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023]
Abstract
Friedreich's ataxia (FA) is an autosomal recessive disorder caused by a deficiency in frataxin (FXN), a mitochondrial protein that plays a critical role in the synthesis of iron-sulfur clusters (Fe-S), vital inorganic cofactors necessary for numerous cellular processes. FA is characterized by progressive ataxia and hypertrophic cardiomyopathy, with cardiac dysfunction as the most common cause of mortality in patients. Commonly used cardiac-specific mouse models of FA use the muscle creatine kinase (MCK) promoter to express Cre recombinase in cardiomyocytes and striated muscle cells in mice with one conditional Fxn allele and one floxed-out/null allele. These mice quickly develop cardiomyopathy that becomes fatal by 9-11 wk of age. Here, we generated a cardiac-specific model with floxed Fxn allele homozygosity (MCK-Fxnflox/flox). MCK-Fxnflox/flox mice were phenotypically normal at 9 wk of age, despite no detectable FXN protein expression. Between 13 and 15 wk of age, these mice began to display progressive cardiomyopathy, including decreased ejection fraction and fractional shortening and increased left ventricular mass. MCK-Fxnflox/flox mice began to lose weight around 16 wk of age, characteristically associated with heart failure in other cardiac-specific FA models. By 18 wk of age, MCK-Fxnflox/flox mice displayed elevated markers of Fe-S deficiency, cardiac stress and injury, and cardiac fibrosis. This modified model reproduced important pathophysiological and biochemical features of FA over a longer timescale than previous cardiac-specific mouse models, offering a larger window for studying potential therapeutics.NEW & NOTEWORTHY Previous cardiac-specific frataxin knockout models exhibit rapid and fatal cardiomyopathy by 9 wk of age. This severe phenotype poses challenges for the design and execution of intervention studies. We introduce an alternative cardiac-specific model, MCK-Fxnflox/flox, with increased longevity and delayed onset of all major phenotypes. These phenotypes develop to the same severity as previous models. Thus, this new model provides the same cardiomyopathy-associated mortality with a larger window for potential studies.
Collapse
Affiliation(s)
- Tyler L Perfitt
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Claudia Huichalaf
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Renea Gooch
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Anna Kuperman
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Youngwook Ahn
- Target Sciences, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Xian Chen
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Soumya Ullas
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Dinesh Hirenallur-Shanthappa
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Yutian Zhan
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Diana Otis
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Laurence O Whiteley
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Christine Bulawa
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| | - Alain Martelli
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer, Incorporated, Cambridge, Massachusetts, United States
| |
Collapse
|
18
|
Pilotto F, Chellapandi DM, Puccio H. Omaveloxolone: a groundbreaking milestone as the first FDA-approved drug for Friedreich ataxia. Trends Mol Med 2024; 30:117-125. [PMID: 38272714 DOI: 10.1016/j.molmed.2023.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024]
Abstract
Friedreich ataxia (FA) is an inherited autosomal recessive neurodegenerative disease (NDD) characterized primarily by progressive sensory and spinocerebellar ataxia associated with hypertrophic cardiomyopathy. FA is due to an intronic GAA repeat expansion within the frataxin gene (FXN) leading to reduced levels of frataxin (FXN) which causes mitochondrial dysfunction, production of reactive oxygen species (ROS), and altered iron metabolism. To date there is no resolutive cure for FA; however, the FDA has recently approved omaveloxolone - a potent activator of nuclear factor erythroid 2-related factor 2 (NRF2) - as the first treatment for FA. We discuss herein the urgency to find a resolutive cure for NDDs that will most probably be achieved via combinatorial therapy targeting multiple disease pathways, and how omavaloxolone serves as an example for future treatments.
Collapse
Affiliation(s)
- Federica Pilotto
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1 CNRS UMR 5261, Inserm U1315, Lyon, France
| | - Deepika M Chellapandi
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1 CNRS UMR 5261, Inserm U1315, Lyon, France
| | - Hélène Puccio
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1 CNRS UMR 5261, Inserm U1315, Lyon, France.
| |
Collapse
|
19
|
Geng Z, Wang J, Liu J, Miao J. Bibliometric Analysis of the Development, Current Status, and Trends in Adult Degenerative Scoliosis Research: A Systematic Review from 1998 to 2023. J Pain Res 2024; 17:153-169. [PMID: 38204581 PMCID: PMC10778169 DOI: 10.2147/jpr.s437575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
Purpose Adult degenerative scoliosis (ADS) research lacks bibliometric analysis, despite numerous studies. This study aimed to systematically analyze the development, current status, hot topics, frontier areas, and trends in ADS research. Patients and Methods A systematic literature review was conducted in the Web of Science Core Collection database from January 1998 to June 2023. Information regarding the country, institution, author, journal, and keywords was collected for each article. Bibliometric analysis was performed using VOSviewer and Citespace software. Results The final analysis covered 1695 publications, demonstrating a steady increase in ADS research. The United States was the most prolific and influential country with 684 publications, followed by China and Japan. The University of California System was the most productive institution with 113 publications. Shaffrey, CI (47 publications) and Lenke, LG (41 publications) were top authors. The analysis revealed seven main research clusters: "intervertebral disc", "adult spinal deformity", "lumbar fusion", "minimally invasive surgery", "navigation", "postoperative complications", and "mental retardation". Keywords with strong bursts of activity included degeneration, prevalence, imbalance, classification, lumbar spinal stenosis, and kyphosis. Conclusion In conclusion, in recent years, ADS research has undergone rapid development. This study analyzed its hot topics, advancements, and research directions, making it the latest bibliometric analysis in this field. The findings aim to provide a new perspective and guidance for clinical practitioners and researchers.
Collapse
Affiliation(s)
- Ziming Geng
- Department of Spine Surgery, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Jian Wang
- Department of Spine Surgery, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Jianchao Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin, People’s Republic of China
| | - Jun Miao
- Department of Spine Surgery, Tianjin Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
20
|
Krasilnikova MM, Humphries CL, Shinsky EM. Friedreich's ataxia: new insights. Emerg Top Life Sci 2023; 7:313-323. [PMID: 37698160 DOI: 10.1042/etls20230017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/09/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023]
Abstract
Friedreich ataxia (FRDA) is an inherited disease that is typically caused by GAA repeat expansion within the first intron of the FXN gene coding for frataxin. This results in the frataxin deficiency that affects mostly muscle, nervous, and cardiovascular systems with progressive worsening of the symptoms over the years. This review summarizes recent progress that was achieved in understanding of molecular mechanism of the disease over the last few years and latest treatment strategies focused on overcoming the frataxin deficiency.
Collapse
Affiliation(s)
- Maria M Krasilnikova
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, U.S.A
| | - Casey L Humphries
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, U.S.A
| | - Emily M Shinsky
- Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, U.S.A
| |
Collapse
|
21
|
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: 11] [Impact Index Per Article: 5.5] [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
|
22
|
Gong C, Long Y, Peng XM, Hu H, Chen J, Xiao L, Zhong YB, Wang MY, Luo Y. Efficacy and safety of noninvasive brain stimulation for patients with cerebellar ataxia: a systematic review and meta-analysis of randomized controlled trials. J Neurol 2023; 270:4782-4799. [PMID: 37460852 DOI: 10.1007/s00415-023-11799-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND With the development of noninvasive brain stimulation (NIBS) techniques, many researchers have turned their attention to NIBS as a promising treatment for cerebellar ataxia. Therefore, we conducted a systematic review and meta-analysis to investigate the efficacy and safety of NIBS in treating patients with cerebellar ataxia. METHODS Databases, including PubMed, Embase, Web of Science, Medline, and Cochrane Library, were retrieved for relevant randomized controlled trials (RCTs). Two researchers conducted literature screening, data extraction, literature quality assessment, and heterogeneity analysis between RCTs. According to the magnitude of heterogeneity I2, an appropriate data analysis model was selected for meta-analysis. RESULTS A total of 14 RCTs including 406 patients with cerebellar ataxia met the inclusion criteria. The included RCTs had an overall low-risk bias and an intermediate level of evidence recommendation for key outcome indicators, such as the scale for the assessment and rating of ataxia (SARA) and international cooperative ataxia rating scale (ICARS). The results of meta-analysis showed that cerebellar NIBS, including transcranial direct current stimulation (tDCS) and repetitive transcranial magnetic stimulation (rTMS), was effective in reducing the SARA scores (MD = - 3.45, 95%CI = [- 4.85, - 2.50], P < 0.05) and ICARS scores (MD = - 10.87, 95%CI = [- 14.46, - 7.28], P < 0.05) in patients with cerebellar ataxia compared to controls. Subgroup analysis showed that the efficacy of tDCS and rTMS was statistically different in patients with cerebellar ataxia as assessed by the SARA scores, but not by the ICARS scores. There was statistically significant difference in the efficacy of NIBS for the treatment of cerebellar ataxia caused by different etiologies. As for safety, 8 of 14 included studies documented the adverse effects of NIBS, and only two studies reported the mild adverse events of NIBS. CONCLUSIONS Cerebellar NIBS was safe and effective in improving the motor coordination of patients with cerebellar ataxia, and tDCS was better than rTMS in the treatment of cerebellar ataxia. In addition, the efficacy of NIBS was different in the treatment of different types of cerebellar ataxia.
Collapse
Affiliation(s)
- Cheng Gong
- Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
| | - Yi Long
- Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
| | - Xu-Miao Peng
- Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
| | - Hao Hu
- Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
| | - Jing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, 341400, Jiangxi Province, China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, 341400, Jiangxi Province, China
| | - Li Xiao
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, 341400, Jiangxi Province, China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, 341400, Jiangxi Province, China
| | - Yan-Biao Zhong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, 341400, Jiangxi Province, China.
| | - Mao-Yuan Wang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, 341400, Jiangxi Province, China.
| | - Yun Luo
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, 341400, Jiangxi Province, China.
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou City, 341400, Jiangxi Province, China.
| |
Collapse
|
23
|
Maheshwari S, Vilema-Enríquez G, Wade-Martins R. Patient-derived iPSC models of Friedreich ataxia: a new frontier for understanding disease mechanisms and therapeutic application. Transl Neurodegener 2023; 12:45. [PMID: 37726850 PMCID: PMC10510273 DOI: 10.1186/s40035-023-00376-8] [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: 05/11/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
Friedreich ataxia (FRDA) is a rare genetic multisystem disorder caused by a pathological GAA trinucleotide repeat expansion in the FXN gene. The numerous drawbacks of historical cellular and rodent models of FRDA have caused difficulty in performing effective mechanistic and translational studies to investigate the disease. The recent discovery and subsequent development of induced pluripotent stem cell (iPSC) technology provides an exciting platform to enable enhanced disease modelling for studies of rare genetic diseases. Utilising iPSCs, researchers have created phenotypically relevant and previously inaccessible cellular models of FRDA. These models enable studies of the molecular mechanisms underlying GAA-induced pathology, as well as providing an exciting tool for the screening and testing of novel disease-modifying therapies. This review explores how the use of iPSCs to study FRDA has developed over the past decade, as well as discussing the enormous therapeutic potentials of iPSC-derived models, their current limitations and their future direction within the field of FRDA research.
Collapse
Affiliation(s)
- Saumya Maheshwari
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Gabriela Vilema-Enríquez
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
24
|
Rodden LN, McIntyre K, Keita M, Wells M, Park C, Profeta V, Waldman A, Rummey C, Balcer LJ, Lynch DR. Retinal hypoplasia and degeneration result in vision loss in Friedreich ataxia. Ann Clin Transl Neurol 2023; 10:1397-1406. [PMID: 37334854 PMCID: PMC10424660 DOI: 10.1002/acn3.51830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/16/2023] [Accepted: 05/26/2023] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVE Friedreich ataxia (FRDA) is an inherited condition caused by a GAA triplet repeat (GAA-TR) expansion in the FXN gene. Clinical features of FRDA include ataxia, cardiomyopathy, and in some, vision loss. In this study, we characterize features of vision loss in a large cohort of adults and children with FRDA. METHODS Using optical coherence tomography (OCT), we measured peripapillary retinal nerve fiber layer (RNFL) thickness in 198 people with FRDA, and 77 controls. Sloan letter charts were used to determine visual acuity. RNFL thickness and visual acuity were compared to measures of disease severity obtained from the Friedreich Ataxia Clinical Outcomes Measures Study (FACOMS). RESULTS The majority of patients, including children, had pathologically thin RNFLs (mean = 73 ± 13 μm in FRDA; 98 ± 9 μm in controls) and low-contrast vision deficits early in the disease course. Variability in RNFL thickness in FRDA (range: 36 to 107 μm) was best predicted by disease burden (GAA-TR length X disease duration). Significant deficits in high-contrast visual acuity were apparent in patients with an RNFL thickness of ≤68 μm. RNFL thickness decreased at a rate of -1.2 ± 1.4 μm/year and reached 68 μm at a disease burden of approximately 12,000 GAA years, equivalent to disease duration of 17 years for participants with 700 GAAs. INTERPRETATION These data suggest that both hypoplasia and subsequent degeneration of the RNFL may be responsible for the optic nerve dysfunction in FRDA and support the development of a vision-directed treatment for selected patients early in the disease to prevent RNFL loss from reaching the critical threshold.
Collapse
Affiliation(s)
- Layne N. Rodden
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Kellie McIntyre
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Medina Keita
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Mckenzie Wells
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Courtney Park
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Victoria Profeta
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Amy Waldman
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Laura J. Balcer
- Departments of Neurology, Population Health and OphthalmologyNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - David R. Lynch
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
25
|
Culley MK, Rao RJ, Mehta M, Zhao J, El Khoury W, Harvey LD, Perk D, Tai YY, Tang Y, Shiva S, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency disrupts mitochondrial respiration and pulmonary endothelial cell function. Vascul Pharmacol 2023; 151:107181. [PMID: 37164245 PMCID: PMC10524929 DOI: 10.1016/j.vph.2023.107181] [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: 08/22/2022] [Revised: 04/19/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Deficiency of iron‑sulfur (FeS) clusters promotes metabolic rewiring of the endothelium and the development of pulmonary hypertension (PH) in vivo. Joining a growing number of FeS biogenesis proteins critical to pulmonary endothelial function, recent data highlighted that frataxin (FXN) reduction drives Fe-S-dependent genotoxic stress and senescence across multiple types of pulmonary vascular disease. Trinucleotide repeat mutations in the FXN gene cause Friedreich's ataxia, a disease characterized by cardiomyopathy and neurodegeneration. These tissue-specific phenotypes have historically been attributed to mitochondrial reprogramming and oxidative stress. Whether FXN coordinates both nuclear and mitochondrial processes in the endothelium is unknown. Here, we aim to identify the mitochondria-specific effects of FXN deficiency in the endothelium that predispose to pulmonary hypertension. Our data highlight an Fe-S-driven metabolic shift separate from previously described replication stress whereby FXN knockdown diminished mitochondrial respiration and increased glycolysis and oxidative species production. In turn, FXN-deficient endothelial cells had increased vasoconstrictor production (EDN1) and decreased nitric oxide synthase expression (NOS3). These data were observed in primary pulmonary endothelial cells after pharmacologic inhibition of FXN, mice carrying a genetic endothelial deletion of FXN, and inducible pluripotent stem cell-derived endothelial cells from patients with FXN mutations. Altogether, this study indicates FXN is an upstream driver of pathologic aberrations in metabolism and genomic stability. Moreover, our study highlights FXN-specific vasoconstriction in vivo, prompting future studies to investigate available and novel PH therapies in contexts of FXN deficiency.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rashmi J Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Monica Mehta
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lloyd D Harvey
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dror Perk
- Medical Scientist Training Program, Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, USA
| | - Marlene Rabinovitch
- Stanford Children's Health Betty Irene Moore Children's Heart Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, UMR7275, IPMC, Valbonne, France
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
26
|
Yaméogo P, Gérard C, Majeau N, Tremblay JP. Removal of the GAA repeat in the heart of a Friedreich's ataxia mouse model using CjCas9. Gene Ther 2023; 30:612-619. [PMID: 36781946 DOI: 10.1038/s41434-023-00387-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/15/2023]
Abstract
Most Friedreich ataxia (FRDA) cases are caused by the elongation of the GAA repeat (GAAr) sequence in the first intron of the FXN gene, leading to a decrease of the frataxin protein expression. Deletion of this GAAr with CRISPR/Cas9 technology leads to an increase in frataxin expression in vitro. We are therefore aiming to develop FRDA treatment based on the deletion of GAAr with CRISPR/Cas9 technology using a single AAV expressing a small Cas9 (CjCas9) and two single guide RNAs (sgRNAs) targeting the FXN gene. This AAV was intraperitoneally administrated to YG8sR (250-300 GAAr) and to YG8-800 (800 GAAr) mice. DNA and RNA were extracted from different organs a month later. PCR amplification of part of intron 1 of the FXN gene detected some GAAr deletion in some cells in heart and liver of both mouse models, but the editing rate was not sufficient to cause an increase in frataxin mRNA in the heart. However, the correlation observed between the editing rate and the distribution of AAV suggests a possible therapy based on the removal of the GAAr with a better delivery tool of the CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Pouiré Yaméogo
- Centre de Recherche du CHU de Québec-Université Laval, Québec city, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec city, QC, Canada
| | - Catherine Gérard
- Centre de Recherche du CHU de Québec-Université Laval, Québec city, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec city, QC, Canada
| | - Nathalie Majeau
- Centre de Recherche du CHU de Québec-Université Laval, Québec city, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec city, QC, Canada
| | - Jacques P Tremblay
- Centre de Recherche du CHU de Québec-Université Laval, Québec city, QC, Canada.
- Département de Médecine Moléculaire, Université Laval, Québec city, QC, Canada.
| |
Collapse
|
27
|
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
|
28
|
Nobili G, Botticelli S, La Penna G, Morante S, Rossi G, Salina G. Probing protein stability: towards a computational atomistic, reliable, affordable, and improvable model. Front Mol Biosci 2023; 10:1122269. [PMID: 37325476 PMCID: PMC10267363 DOI: 10.3389/fmolb.2023.1122269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
We present an improved application of a recently proposed computational method designed to evaluate the change of free energy as a function of the average value of a suitably chosen collective variable in proteins. The method is based on a full atomistic description of the protein and its environment. The goal is to understand how the protein melting temperature changes upon single-point mutations, because the sign of the temperature variation will allow us to discriminate stabilizing vs. destabilizing mutations in protein sequences. In this refined application the method is based on altruistic well-tempered metadynamics, a variant of multiple-walkers metadynamics. The resulting metastatistics is then modulated by the maximal constrained entropy principle. The latter turns out to be especially helpful in free-energy calculations as it is able to alleviate the severe limitations of metadynamics in properly sampling folded and unfolded configurations. In this work we apply the computational strategy outlined above in the case of the bovine pancreatic trypsin inhibitor, a well-studied small protein, which is a reference for computer simulations since decades. We compute the variation of the melting temperature characterizing the folding-unfolding process between the wild-type protein and two of its single-point mutations that are seen to have opposite effect on the free energy changes. The same approach is used for free energy difference calculations between a truncated form of frataxin and a set of five of its variants. Simulation data are compared to in vitro experiments. In all cases the sign of the change of melting temperature is reproduced, under the further approximation of using an empirical effective mean-field to average out protein-solvent interactions.
Collapse
Affiliation(s)
- Germano Nobili
- Dipartimento di Fisica, Universitá di Roma Tor Vergata, Roma, Italy
- INFN, Sezione di Roma Tor Vergata, Roma, Italy
| | - Simone Botticelli
- Dipartimento di Fisica, Universitá di Roma Tor Vergata, Roma, Italy
- INFN, Sezione di Roma Tor Vergata, Roma, Italy
| | - Giovanni La Penna
- CNR-Istituto di Chimica Dei Composti Organometallici, Firenze, Italy
- INFN, Sezione di Roma Tor Vergata, Roma, Italy
| | - Silvia Morante
- Dipartimento di Fisica, Universitá di Roma Tor Vergata, Roma, Italy
- INFN, Sezione di Roma Tor Vergata, Roma, Italy
- CNR-Istituto di Chimica Dei Composti Organometallici, Firenze, Italy
| | - Giancarlo Rossi
- Dipartimento di Fisica, Universitá di Roma Tor Vergata, Roma, Italy
- INFN, Sezione di Roma Tor Vergata, Roma, Italy
- Museo Storico della Fisica e Centro Studi e Ricerche E. Fermi, Roma, Italy
| | | |
Collapse
|
29
|
Tiberi J, Segatto M, Fiorenza MT, La Rosa P. Apparent Opportunities and Hidden Pitfalls: The Conflicting Results of Restoring NRF2-Regulated Redox Metabolism in Friedreich's Ataxia Pre-Clinical Models and Clinical Trials. Biomedicines 2023; 11:biomedicines11051293. [PMID: 37238963 DOI: 10.3390/biomedicines11051293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
Friedreich's ataxia (FRDA) is an autosomal, recessive, inherited neurodegenerative disease caused by the loss of activity of the mitochondrial protein frataxin (FXN), which primarily affects dorsal root ganglia, cerebellum, and spinal cord neurons. The genetic defect consists of the trinucleotide GAA expansion in the first intron of FXN gene, which impedes its transcription. The resulting FXN deficiency perturbs iron homeostasis and metabolism, determining mitochondrial dysfunctions and leading to reduced ATP production, increased reactive oxygen species (ROS) formation, and lipid peroxidation. These alterations are exacerbated by the defective functionality of the nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor acting as a key mediator of the cellular redox signalling and antioxidant response. Because oxidative stress represents a major pathophysiological contributor to FRDA onset and progression, a great effort has been dedicated to the attempt to restore the NRF2 signalling axis. Despite this, the beneficial effects of antioxidant therapies in clinical trials only partly reflect the promising results obtained in preclinical studies conducted in cell cultures and animal models. For these reasons, in this critical review, we overview the outcomes obtained with the administration of various antioxidant compounds and critically analyse the aspects that may have contributed to the conflicting results of preclinical and clinical studies.
Collapse
Affiliation(s)
- Jessica Tiberi
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Marco Segatto
- Department of Bioscience and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00179 Rome, Italy
| | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00179 Rome, Italy
| |
Collapse
|
30
|
Johnson KC, Corey DR. RNAi in cell nuclei: potential for a new layer of biological regulation and a new strategy for therapeutic discovery. RNA (NEW YORK, N.Y.) 2023; 29:415-422. [PMID: 36657971 PMCID: PMC10019369 DOI: 10.1261/rna.079500.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
RNA interference is almost always associated with post-transcriptional silencing in the cytoplasm. MicroRNAs (miRNAs) and critical RNAi protein factors like argonaute (AGO) and trinucleotide repeat binding containing 6 protein (TNRC6), however, are also found in cell nuclei, suggesting that nuclear miRNAs may be targets for gene regulation. Designed small duplex RNAs (dsRNAs) can modulate nuclear processes such as transcription and splicing, suggesting that they can also provide leads for therapeutic discovery. The goal of this Perspective is to provide the background on nuclear RNAi necessary to guide discussions on whether nuclear RNAi can play a role in therapeutic development programs.
Collapse
Affiliation(s)
- Krystal C Johnson
- UT Southwestern Medical Center, Departments of Pharmacology and Biochemistry, Dallas, Texas 75205, USA
| | - David R Corey
- UT Southwestern Medical Center, Departments of Pharmacology and Biochemistry, Dallas, Texas 75205, USA
| |
Collapse
|
31
|
Pizcueta P, Vergara C, Emanuele M, Vilalta A, Rodríguez-Pascau L, Martinell M. Development of PPARγ Agonists for the Treatment of Neuroinflammatory and Neurodegenerative Diseases: Leriglitazone as a Promising Candidate. Int J Mol Sci 2023; 24:ijms24043201. [PMID: 36834611 PMCID: PMC9961553 DOI: 10.3390/ijms24043201] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence suggests that the peroxisome proliferator-activated receptor γ (PPARγ), a member of the nuclear receptor superfamily, plays an important role in physiological processes in the central nervous system (CNS) and is involved in cellular metabolism and repair. Cellular damage caused by acute brain injury and long-term neurodegenerative disorders is associated with alterations of these metabolic processes leading to mitochondrial dysfunction, oxidative stress, and neuroinflammation. PPARγ agonists have demonstrated the potential to be effective treatments for CNS diseases in preclinical models, but to date, most drugs have failed to show efficacy in clinical trials of neurodegenerative diseases including amyotrophic lateral sclerosis, Parkinson's disease, and Alzheimer's disease. The most likely explanation for this lack of efficacy is the insufficient brain exposure of these PPARγ agonists. Leriglitazone is a novel, blood-brain barrier (BBB)-penetrant PPARγ agonist that is being developed to treat CNS diseases. Here, we review the main roles of PPARγ in physiology and pathophysiology in the CNS, describe the mechanism of action of PPARγ agonists, and discuss the evidence supporting the use of leriglitazone to treat CNS diseases.
Collapse
Affiliation(s)
- Pilar Pizcueta
- Minoryx Therapeutics SL, 08302 Barcelona, Spain
- Correspondence:
| | | | - Marco Emanuele
- Minoryx Therapeutics BE, Gosselies, 6041 Charleroi, Belgium
| | | | | | - Marc Martinell
- Minoryx Therapeutics SL, 08302 Barcelona, Spain
- Minoryx Therapeutics BE, Gosselies, 6041 Charleroi, Belgium
| |
Collapse
|
32
|
Profeta V, McIntyre K, Wells M, Park C, Lynch DR. Omaveloxolone: an activator of Nrf2 for the treatment of Friedreich ataxia. Expert Opin Investig Drugs 2023; 32:5-16. [PMID: 36708320 DOI: 10.1080/13543784.2023.2173063] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Friedreich ataxia (FRDA) is a rare autosomal recessive degenerative disorder characterized by ataxia, dysarthria, diabetes, cardiomyopathy, scoliosis, and occasionally vision loss in late-stage disease. The discovery of the abnormal gene in FRDA and its product frataxin has provided insight into the pathophysiology and mechanisms of treatment. AREAS COVERED Although the neurologic phenotype of FRDA is well defined, there are currently no established pharmacological treatments. Omaveloxolone, a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, is currently under review by the Food and Drug Administration (FDA) and has the potential to be the first approved treatment for FRDA. In the present report, we have reviewed the basic and clinical literature on Nrf2 deficiency in FRDA, and evidence for the benefit of omaveloxolone. EXPERT OPINION The present perspective suggests that omaveloxolone is a rational and efficacious therapy that is possibly disease modifying in treatment of FRDA.
Collapse
Affiliation(s)
- Victoria Profeta
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kellie McIntyre
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - McKenzie Wells
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Courtney Park
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David R Lynch
- Departments of Pediatrics and Neurology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Milne SC, Kim SH, Murphy A, Larkindale J, Farmer J, Malapira R, Danoudis M, Shaw J, Ramakrishnan T, Rasouli F, Yiu EM, Georgiou-Karistianis N, Tai G, Zesiewicz T, Delatycki MB, Corben LA. The Responsiveness of Gait and Balance Outcomes to Disease Progression in Friedreich Ataxia. CEREBELLUM (LONDON, ENGLAND) 2022; 21:963-975. [PMID: 34855135 DOI: 10.1007/s12311-021-01348-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
To identify gait and balance measures that are responsive to change during the timeline of a clinical trial in Friedreich ataxia (FRDA), we administered a battery of potential measures three times over a 12-month period. Sixty-one ambulant individuals with FRDA underwent assessment of gait and balance at baseline, 6 months and 12 months. Outcomes included GAITRite® spatiotemporal gait parameters; Biodex Balance System Postural Stability Test (PST) and Limits of Stability; Berg Balance Scale (BBS); Timed 25-Foot Walk Test; Dynamic Gait Index (DGI); SenseWear MF Armband step and energy activity; and the Friedreich Ataxia Rating Scale Upright Stability Subscale (FARS USS). The standardised response mean (SRM) or correlation coefficients were reported as effect size indices for comparison of internal responsiveness. Internal responsiveness was also analysed in subgroups. SenseWear Armband daily step count had the largest effect size of all the variables over 6 months (SRM = -0.615), while the PST medial-lateral index had the largest effect size (SRM = 0.829) over 12 months. The FARS USS (SRM = 0.824) and BBS (SRM = -0.720) were the only outcomes able to detect change over 12 months in all subgroups. The DGI was the most responsive outcome in children, detecting a mean change of -2.59 (95% CI -3.52 to -1.66, p < 0.001, SRM = -1.429). In conclusion, the FARS USS and BBS are highly responsive and can detect change in a wide range of ambulant individuals with FRDA. However, therapeutic effects in children may be best measured by the DGI.
Collapse
Affiliation(s)
- Sarah C Milne
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne,, Australia.
- Physiotherapy Department, Monash Health, Melbourne, Australia.
- Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia.
- School of Primary and Allied Health Care, Monash University, Melbourne, Australia.
| | | | - Anna Murphy
- MonARC, Monash Health, Melbourne, Australia
- School of Public Health and Preventative Medicine, Monash University, Melbourne, Australia
| | | | | | | | - Mary Danoudis
- MonARC, Monash Health, Melbourne, Australia
- School of Public Health and Preventative Medicine, Monash University, Melbourne, Australia
| | | | | | | | - Eppie M Yiu
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne,, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- Department of Neurology, The Royal Children's Hospital Melbourne, Melbourne, Australia
| | - Nellie Georgiou-Karistianis
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Geneieve Tai
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne,, Australia
| | | | - Martin B Delatycki
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne,, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- Victorian Clinical Genetics Services, Melbourne, Australia
| | - Louise A Corben
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne,, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- School of Primary and Allied Health Care, Monash University, Melbourne, Australia
| |
Collapse
|
34
|
Hohenfeld C, Terstiege U, Dogan I, Giunti P, Parkinson MH, Mariotti C, Nanetti L, Fichera M, Durr A, Ewenczyk C, Boesch S, Nachbauer W, Klopstock T, Stendel C, Rodríguez de Rivera Garrido FJ, Schöls L, Hayer SN, Klockgether T, Giordano I, Didszun C, Rai M, Pandolfo M, Rauhut H, Schulz JB, Reetz K. Prediction of the disease course in Friedreich ataxia. Sci Rep 2022; 12:19173. [PMID: 36357508 PMCID: PMC9649725 DOI: 10.1038/s41598-022-23666-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
We explored whether disease severity of Friedreich ataxia can be predicted using data from clinical examinations. From the database of the European Friedreich Ataxia Consortium for Translational Studies (EFACTS) data from up to five examinations of 602 patients with genetically confirmed FRDA was included. Clinical instruments and important symptoms of FRDA were identified as targets for prediction, while variables such as genetics, age of disease onset and first symptom of the disease were used as predictors. We used modelling techniques including generalised linear models, support-vector-machines and decision trees. The scale for rating and assessment of ataxia (SARA) and the activities of daily living (ADL) could be predicted with predictive errors quantified by root-mean-squared-errors (RMSE) of 6.49 and 5.83, respectively. Also, we were able to achieve reasonable performance for loss of ambulation (ROC-AUC score of 0.83). However, predictions for the SCA functional assessment (SCAFI) and presence of cardiological symptoms were difficult. In conclusion, we demonstrate that some clinical features of FRDA can be predicted with reasonable error; being a first step towards future clinical applications of predictive modelling. In contrast, targets where predictions were difficult raise the question whether there are yet unknown variables driving the clinical phenotype of FRDA.
Collapse
Affiliation(s)
- Christian Hohenfeld
- grid.1957.a0000 0001 0728 696XDepartment of Neurology, RWTH Aachen University, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XJARA Brain Institute Molecular Neuroscience and Neuroimaging, Research Centre Jülich and RWTH Aachen University, 52056 Aachen, Germany
| | - Ulrich Terstiege
- grid.1957.a0000 0001 0728 696XChair for Mathematics of Information Processing, RWTH Aachen University, 52062 Aachen, Germany
| | - Imis Dogan
- grid.1957.a0000 0001 0728 696XDepartment of Neurology, RWTH Aachen University, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XJARA Brain Institute Molecular Neuroscience and Neuroimaging, Research Centre Jülich and RWTH Aachen University, 52056 Aachen, Germany
| | - Paola Giunti
- grid.83440.3b0000000121901201Department of Clinical and Movement Neurosciences, Ataxia Centre, UCL-Queen Square Institute of Neurology, London, WC1N 3BG UK
| | - Michael H. Parkinson
- grid.83440.3b0000000121901201Department of Clinical and Movement Neurosciences, Ataxia Centre, UCL-Queen Square Institute of Neurology, London, WC1N 3BG UK
| | - Caterina Mariotti
- grid.417894.70000 0001 0707 5492Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Lorenzo Nanetti
- grid.417894.70000 0001 0707 5492Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Mario Fichera
- grid.417894.70000 0001 0707 5492Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy ,grid.7563.70000 0001 2174 1754PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Alexandra Durr
- grid.411439.a0000 0001 2150 9058Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, 75646 Paris, France
| | - Claire Ewenczyk
- grid.411439.a0000 0001 2150 9058Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, 75646 Paris, France
| | - Sylvia Boesch
- grid.5361.10000 0000 8853 2677Department of Neurology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Wolfgang Nachbauer
- grid.5361.10000 0000 8853 2677Department of Neurology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Thomas Klopstock
- grid.5252.00000 0004 1936 973XDepartment of Neurology, Friedrich Baur Institute, University Hospital, LMU, 80336 Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany ,grid.452617.3Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Claudia Stendel
- grid.5252.00000 0004 1936 973XDepartment of Neurology, Friedrich Baur Institute, University Hospital, LMU, 80336 Munich, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | | | - Ludger Schöls
- grid.10392.390000 0001 2190 1447Department of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Stefanie N. Hayer
- grid.10392.390000 0001 2190 1447Department of Neurology and Hertie-Institute for Clinical Brain Research, University of Tübingen, 72076 Tübingen, Germany
| | - Thomas Klockgether
- grid.15090.3d0000 0000 8786 803XDepartment of Neurology, University Hospital of Bonn, 53127 Bonn, Germany ,grid.424247.30000 0004 0438 0426German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Ilaria Giordano
- grid.15090.3d0000 0000 8786 803XDepartment of Neurology, University Hospital of Bonn, 53127 Bonn, Germany
| | - Claire Didszun
- grid.1957.a0000 0001 0728 696XDepartment of Neurology, RWTH Aachen University, 52074 Aachen, Germany
| | - Myriam Rai
- grid.4989.c0000 0001 2348 0746Laboratory of Experimental Neurology, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Massimo Pandolfo
- grid.4989.c0000 0001 2348 0746Laboratory of Experimental Neurology, Université Libre de Bruxelles, 1070 Brussels, Belgium ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4 Canada
| | - Holger Rauhut
- grid.1957.a0000 0001 0728 696XChair for Mathematics of Information Processing, RWTH Aachen University, 52062 Aachen, Germany
| | - Jörg B. Schulz
- grid.1957.a0000 0001 0728 696XDepartment of Neurology, RWTH Aachen University, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XJARA Brain Institute Molecular Neuroscience and Neuroimaging, Research Centre Jülich and RWTH Aachen University, 52056 Aachen, Germany
| | - Kathrin Reetz
- grid.1957.a0000 0001 0728 696XDepartment of Neurology, RWTH Aachen University, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XJARA Brain Institute Molecular Neuroscience and Neuroimaging, Research Centre Jülich and RWTH Aachen University, 52056 Aachen, Germany
| |
Collapse
|
35
|
Beyond Sarcomeric Hypertrophic Cardiomyopathy: How to Diagnose and Manage Phenocopies. Curr Cardiol Rep 2022; 24:1567-1585. [PMID: 36053410 DOI: 10.1007/s11886-022-01778-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW We describe the most common phenocopies of hypertrophic cardiomyopathy, their pathogenesis, and clinical presentation highlighting similarities and differences. We also suggest a step-by-step diagnostic work-up that can guide in differential diagnosis and management. RECENT FINDINGS In the last years, a wider application of genetic testing and the advances in cardiac imaging have significantly changed the diagnostic approach to HCM phenocopies. Different prognosis and management, with an increasing availability of disease-specific therapies, make differential diagnosis mandatory. The HCM phenotype can be the cardiac manifestation of different inherited and acquired disorders presenting different etiology, prognosis, and treatment. Differential diagnosis requires a cardiomyopathic mindset allowing to recognize red flags throughout the diagnostic work-up starting from clinical and family history and ending with advanced imaging and genetic testing. Different prognosis and management, with an increasing availability of disease-specific therapies make differential diagnosis mandatory.
Collapse
|
36
|
Li Y, Li J, Wang J, Zhang S, Giles K, Prakash TP, Rigo F, Napierala JS, Napierala M. Premature transcription termination at the expanded GAA repeats and aberrant alternative polyadenylation contributes to the Frataxin transcriptional deficit in Friedreich's ataxia. Hum Mol Genet 2022; 31:3539-3557. [PMID: 35708503 PMCID: PMC9558844 DOI: 10.1093/hmg/ddac134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 06/05/2022] [Indexed: 11/18/2022] Open
Abstract
Frataxin deficiency in Friedreich's ataxia results from transcriptional downregulation of the FXN gene caused by expansion of the intronic trinucleotide guanine-adenine-adenine (GAA) repeats. We used multiple transcriptomic approaches to determine the molecular mechanism of transcription inhibition caused by long GAAs. We uncovered that transcription of FXN in patient cells is prematurely terminated upstream of the expanded repeats leading to the formation of a novel, truncated and stable RNA. This FXN early terminated transcript (FXN-ett) undergoes alternative, non-productive splicing and does not contribute to the synthesis of functional frataxin. The level the FXN-ett RNA directly correlates with the length of the longer of the two expanded GAA tracts. Targeting GAAs with antisense oligonucleotides or excision of the repeats eliminates the transcription impediment, diminishes expression of the aberrant FXN-ett, while increasing levels of FXN mRNA and frataxin. Non-productive transcription may represent a common phenomenon and attractive therapeutic target in diseases caused by repeat-mediated transcription aberrations.
Collapse
Affiliation(s)
- Yanjie Li
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Jixue Li
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Jun Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Siyuan Zhang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Keith Giles
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
| | - Thazha P Prakash
- Ionis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Frank Rigo
- Ionis Pharmaceuticals Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Jill S Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL 35294, USA
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
37
|
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: 36] [Impact Index Per Article: 12.0] [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
|
38
|
Antoine JC. Sensory neuronopathies, diagnostic criteria and causes. Curr Opin Neurol 2022; 35:553-561. [PMID: 35950727 DOI: 10.1097/wco.0000000000001105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To stress on the diagnostic strategy of sensory neuronopathies (SNN), including new genes and antibodies. RECENT FINDING SNN involve paraneoplastic, dysimmune, toxic, viral and genetic mechanisms. About one-third remains idiopathic. Recently, new antibodies and genes have reduced this proportion. Anti-FGFR3 and anti-AGO antibodies are not specific of SNN, although SNN is predominant and may occur with systemic autoimmune diseases. These antibodies are the only marker of an underlying dysimmune context in two-thirds (anti-FGFR3 antibodies) and one-third of the cases (anti-AGO antibodies), respectively. Patients with anti-AGO antibodies may improve with treatment, which is less clear with anti-FGFR3 antibodies. A biallelic expansion in the RFC1 gene is responsible for the cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) in which SNN is a predominant manifestation. Most of the patients have an adult onset and are sporadic. The RFC1 mutation may represent one-third of idiopathic sensory neuropathies. Finally, the criteria for the diagnosis of paraneoplastic SNN have recently been updated. SUMMARY The diagnostic of SNN relies on criteria distinguishing SNN from other neuropathies. The strategy in search of their cause now needs to include these recent findings.
Collapse
Affiliation(s)
- Jean-Christophe Antoine
- University Hospital of Saint-Etienne, European Reference Network for Rare Diseases- Euro-NMD, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
39
|
Kelekçi S, Yıldız AB, Sevinç K, Çimen DU, Önder T. Perspectives on current models of Friedreich’s ataxia. Front Cell Dev Biol 2022; 10:958398. [PMID: 36036008 PMCID: PMC9403045 DOI: 10.3389/fcell.2022.958398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Friedreich’s ataxia (FRDA, OMIM#229300) is the most common hereditary ataxia, resulting from the reduction of frataxin protein levels due to the expansion of GAA repeats in the first intron of the FXN gene. Why the triplet repeat expansion causes a decrease in Frataxin protein levels is not entirely known. Generation of effective FRDA disease models is crucial for answering questions regarding the pathophysiology of this disease. There have been considerable efforts to generate in vitro and in vivo models of FRDA. In this perspective article, we highlight studies conducted using FRDA animal models, patient-derived materials, and particularly induced pluripotent stem cell (iPSC)-derived models. We discuss the current challenges in using FRDA animal models and patient-derived cells. Additionally, we provide a brief overview of how iPSC-based models of FRDA were used to investigate the main pathways involved in disease progression and to screen for potential therapeutic agents for FRDA. The specific focus of this perspective article is to discuss the outlook and the remaining challenges in the context of FRDA iPSC-based models.
Collapse
Affiliation(s)
| | | | | | | | - Tamer Önder
- *Correspondence: Simge Kelekçi, , ; Tamer Önder,
| |
Collapse
|
40
|
O'Connell TM, Logsdon DL, Payne RM. Metabolomics analysis reveals dysregulation in one carbon metabolism in Friedreich Ataxia. Mol Genet Metab 2022; 136:306-314. [PMID: 35798654 DOI: 10.1016/j.ymgme.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/14/2022] [Accepted: 06/05/2022] [Indexed: 11/30/2022]
Abstract
Friedreich Ataxia (FA) is a rare and often fatal autosomal recessive disease in which a mitochondrial protein, frataxin (FXN), is severely reduced in all tissues. With loss of FXN, mitochondrial metabolism is severely disrupted. Multiple therapeutic approaches are in development, but a key limitation is the lack of biomarkers reflecting the activity of FXN in a timely fashion. We predicted this dysregulated metabolism would present a unique metabolite profile in blood of FA patients versus Controls (Con). Plasma from 10 FA and 11 age and sex matched Con subjects was analyzed by targeted mass spectrometry and untargeted NMR. This combined approach yielded quantitative measurements for 540 metabolites and found 59 unique metabolites (55 from MS and 4 from NMR) that were significantly different between cohorts. Correlation-based network analysis revealed several clusters of pathway related metabolites including a cluster associated with one‑carbon (1C) metabolism composed of formate, sarcosine, hypoxanthine, and homocysteine. Receiver operator characteristics analyses demonstrated an excellent ability to discriminate between Con and FA with AUC values >0.95. These results are the first reported metabolomic analyses of human patients with FA. The metabolic perturbations, especially those related to 1C metabolism, may serve as a valuable biomarker panel of disease progression and response to therapy. The identification of dysregulated 1C metabolism may also inform the search for new therapeutic targets related to this pathway.
Collapse
Affiliation(s)
- Thomas M O'Connell
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, United States of America; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - David L Logsdon
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - R Mark Payne
- Department of Pediatrics, Division of Cardiology, and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States of America
| |
Collapse
|
41
|
Gene Therapy for Mitochondrial Diseases: Current Status and Future Perspective. Pharmaceutics 2022; 14:pharmaceutics14061287. [PMID: 35745859 PMCID: PMC9231068 DOI: 10.3390/pharmaceutics14061287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial diseases (MDs) are a group of severe genetic disorders caused by mutations in the nuclear or mitochondrial genome encoding proteins involved in the oxidative phosphorylation (OXPHOS) system. MDs have a wide range of symptoms, ranging from organ-specific to multisystemic dysfunctions, with different clinical outcomes. The lack of natural history information, the limits of currently available preclinical models, and the wide range of phenotypic presentations seen in MD patients have all hampered the development of effective therapies. The growing number of pre-clinical and clinical trials over the last decade has shown that gene therapy is a viable precision medicine option for treating MD. However, several obstacles must be overcome, including vector design, targeted tissue tropism and efficient delivery, transgene expression, and immunotoxicity. This manuscript offers a comprehensive overview of the state of the art of gene therapy in MD, addressing the main challenges, the most feasible solutions, and the future perspectives of the field.
Collapse
|
42
|
Affiliation(s)
- R. Mark Payne
- Indiana University School of Medicine, Wells Center for Pediatric Research, 1044 West Walnut, R4302b, Indianapolis, IN 46202, USA
- Corresponding author R. Mark Payne, MD, Indiana University School of Medicine, Wells Center for Pediatric Research, 1044 West Walnut, R4302b, Indianapolis, IN 46202, USA.
| |
Collapse
|
43
|
Sivakumar A, Cherqui S. Advantages and Limitations of Gene Therapy and Gene Editing for Friedreich's Ataxia. Front Genome Ed 2022; 4:903139. [PMID: 35663795 PMCID: PMC9157421 DOI: 10.3389/fgeed.2022.903139] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an inherited, multisystemic disorder predominantly caused by GAA hyper expansion in intron 1 of frataxin (FXN) gene. This expansion mutation transcriptionally represses FXN, a mitochondrial protein that is required for iron metabolism and mitochondrial homeostasis, leading to neurodegerative and cardiac dysfunction. Current therapeutic options for FRDA are focused on improving mitochondrial function and increasing frataxin expression through pharmacological interventions but are not effective in delaying or preventing the neurodegeneration in clinical trials. Recent research on in vivo and ex vivo gene therapy methods in FRDA animal and cell models showcase its promise as a one-time therapy for FRDA. In this review, we provide an overview on the current and emerging prospects of gene therapy for FRDA, with specific focus on advantages of CRISPR/Cas9-mediated gene editing of FXN as a viable option to restore endogenous frataxin expression. We also assess the potential of ex vivo gene editing in hematopoietic stem and progenitor cells as a potential autologous transplantation therapeutic option and discuss its advantages in tackling FRDA-specific safety aspects for clinical translation.
Collapse
Affiliation(s)
| | - Stephanie Cherqui
- Division of Genetics, Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
44
|
Botticelli S, La Penna G, Nobili G, Rossi G, Stellato F, Morante S. Modelling Protein Plasticity: The Example of Frataxin and Its Variants. Molecules 2022; 27:1955. [PMID: 35335316 PMCID: PMC8950120 DOI: 10.3390/molecules27061955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 12/20/2022] Open
Abstract
Frataxin (FXN) is a protein involved in storage and delivery of iron in the mitochondria. Single-point mutations in the FXN gene lead to reduced production of functional frataxin, with the consequent dyshomeostasis of iron. FXN variants are at the basis of neurological impairment (the Friedreich's ataxia) and several types of cancer. By using altruistic metadynamics in conjunction with the maximal constrained entropy principle, we estimate the change of free energy in the protein unfolding of frataxin and of some of its pathological mutants. The sampled configurations highlight differences between the wild-type and mutated sequences in the stability of the folded state. In partial agreement with thermodynamic experiments, where most of the analyzed variants are characterized by lower thermal stability compared to wild type, the D104G variant is found with a stability comparable to the wild-type sequence and a lower water-accessible surface area. These observations, obtained with the new approach we propose in our work, point to a functional switch, affected by single-point mutations, of frataxin from iron storage to iron release. The method is suitable to investigate wide structural changes in proteins in general, after a proper tuning of the chosen collective variable used to perform the transition.
Collapse
Affiliation(s)
- Simone Botticelli
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
| | - Giovanni La Penna
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
- Istituto di Chimica dei Composti Organometallici, Consiglio Nazionale delle Ricerche, Via Madonna del Piano 10, I-50019 Firenze, Italy
| | - Germano Nobili
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
| | - Giancarlo Rossi
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
- Centro Fermi—Museo Storico della Fisica e Centro Studi e Ricerche E. Fermi, I-00184 Roma, Italy
| | - Francesco Stellato
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
- Centro Fermi—Museo Storico della Fisica e Centro Studi e Ricerche E. Fermi, I-00184 Roma, Italy
| | - Silvia Morante
- Dipartimento di Fisica, Università di Roma Tor Vergata and Sezione di Roma Tor Vergata, INFN, Via della Ricerca Scientifica 1, I-00133 Roma, Italy; (S.B.); (G.N.); (G.R.); (F.S.); (S.M.)
| |
Collapse
|
45
|
Huichalaf C, Perfitt TL, Kuperman A, Gooch R, Kovi RC, Brenneman KA, Chen X, Hirenallur-Shanthappa D, Ma T, Assaf BT, Pardo I, Franks T, Monarski L, Cheng TW, Le K, Su C, Somanathan S, Whiteley LO, Bulawa C, Pregel MJ, Martelli A. In vivo overexpression of frataxin causes toxicity mediated by iron-sulfur cluster deficiency. Mol Ther Methods Clin Dev 2022; 24:367-378. [PMID: 35252470 PMCID: PMC8866050 DOI: 10.1016/j.omtm.2022.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/04/2022] [Indexed: 12/25/2022]
Abstract
Friedreich's ataxia is a rare disorder resulting from deficiency of frataxin, a mitochondrial protein implicated in the synthesis of iron-sulfur clusters. Preclinical studies in mice have shown that gene therapy is a promising approach to treat individuals with Friedreich's ataxia. However, a recent report provided evidence that AAVrh10-mediated overexpression of frataxin could lead to cardiotoxicity associated with mitochondrial dysfunction. While evaluating an AAV9-based frataxin gene therapy using a chicken β-actin promoter, we showed that toxic overexpression of frataxin could be reached in mouse liver and heart with doses between 1 × 1013 and 1 × 1014 vg/kg. In a mouse model of cardiac disease, these doses only corrected cardiac dysfunction partially and transiently and led to adverse findings associated with iron-sulfur cluster deficiency in liver. We demonstrated that toxicity required frataxin's primary function by using a frataxin construct bearing the N146K mutation, which impairs binding to the iron-sulfur cluster core complex. At the lowest tested dose, we observed moderate liver toxicity that was accompanied by progressive loss of transgene expression and liver regeneration. Together, our data provide insights into the toxicity of frataxin overexpression that should be considered in the development of a gene therapy approach for Friedreich's ataxia.
Collapse
Affiliation(s)
- Claudia Huichalaf
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Tyler L Perfitt
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Anna Kuperman
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Renea Gooch
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Ramesh C Kovi
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Karrie A Brenneman
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Xian Chen
- Comparative Medicine, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | | | - Tiffany Ma
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Basel T Assaf
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Ingrid Pardo
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Tania Franks
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Laura Monarski
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Ting-Wen Cheng
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Kevin Le
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Chunyan Su
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Suryanarayan Somanathan
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Laurence O Whiteley
- Drug Safety Research and Development, Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Christine Bulawa
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Marko J Pregel
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| | - Alain Martelli
- Rare Disease Research Unit, Worldwide Research, Development and Medical, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA
| |
Collapse
|
46
|
Mitochondrial De Novo Assembly of Iron–Sulfur Clusters in Mammals: Complex Matters in a Complex That Matters. INORGANICS 2022. [DOI: 10.3390/inorganics10030031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022] Open
Abstract
Iron–sulfur clusters (Fe–S or ISC) are essential cofactors that function in a wide range of biological pathways. In mammalian cells, Fe–S biosynthesis primarily relies on mitochondria and involves a concerted group of evolutionary-conserved proteins forming the ISC pathway. In the early stage of the ISC pathway, the Fe–S core complex is required for de novo assembly of Fe–S. In humans, the Fe–S core complex comprises the cysteine desulfurase NFS1, the scaffold protein ISCU2, frataxin (FXN), the ferredoxin FDX2, and regulatory/accessory proteins ISD11 and Acyl Carrier Protein (ACP). In recent years, the field has made significant advances in unraveling the structure of the Fe–S core complex and the mechanism underlying its function. Herein, we review the key recent findings related to the Fe–S core complex and its components. We highlight some of the unanswered questions and provide a model of the Fe–S assembly within the complex. In addition, we briefly touch on the genetic diseases associated with mutations in the Fe–S core complex components.
Collapse
|
47
|
Schreiber AM, Li Y, Chen YH, Napierala JS, Napierala M. Selected Histone Deacetylase Inhibitors Reverse the Frataxin Transcriptional Defect in a Novel Friedreich's Ataxia Induced Pluripotent Stem Cell-Derived Neuronal Reporter System. Front Neurosci 2022; 16:836476. [PMID: 35281493 PMCID: PMC8904878 DOI: 10.3389/fnins.2022.836476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/12/2022] [Indexed: 11/23/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a neurodegenerative disorder caused by the expansion of guanine-adenine-adenine repeats within the first intron of the frataxin (FXN) gene. The location and nature of the expansion have been proven to contribute to transcriptional repression of FXN by decreasing the rate of polymerase II (RNA polymerase II) progression and increasing the presence of histone modifications associated with a heterochromatin-like state. Targeting impaired FXN transcription appears as a feasible option for therapeutic intervention, while no cure currently exists. We created a novel reporter cell line containing an FXN-Nanoluciferase (FXN-NLuc) fusion in induced pluripotent stem cells (iPSCs) reprogrammed from the fibroblasts of patients with FRDA, thus allowing quantification of endogenous FXN expression. The use of iPSCs provides the opportunity to differentiate these cells into disease-relevant neural progenitor cells (NPCs). NPCs derived from the FXN-NLuc line responded to treatments with a known FXN inducer, RG109. Results were validated by quantitative PCR and Western blot in multiple FRDA NPC lines. We then screened a commercially available library of compounds consisting of molecules targeting various enzymes and pathways critical for silencing or activation of gene expression. Only selected histone deacetylase inhibitors were capable of partial reactivation of FXN expression. This endogenous, FRDA iPSC-derived reporter can be utilized for high-throughput campaigns performed in cells most relevant to disease pathology in search of FXN transcription activators.
Collapse
Affiliation(s)
- Anna M. Schreiber
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yanjie Li
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yi-Hsien Chen
- Genome Engineering and iPSC Center, Washington University, St. Louis, MO, United States
| | - Jill S. Napierala
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
48
|
Shah S, Dooms MM, Amaral-Garcia S, Igoillo-Esteve M. Current Drug Repurposing Strategies for Rare Neurodegenerative Disorders. Front Pharmacol 2022; 12:768023. [PMID: 34992533 PMCID: PMC8724568 DOI: 10.3389/fphar.2021.768023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Rare diseases are life-threatening or chronically debilitating low-prevalent disorders caused by pathogenic mutations or particular environmental insults. Due to their high complexity and low frequency, important gaps still exist in their prevention, diagnosis, and treatment. Since new drug discovery is a very costly and time-consuming process, leading pharmaceutical companies show relatively low interest in orphan drug research and development due to the high cost of investments compared to the low market return of the product. Drug repurposing–based approaches appear then as cost- and time-saving strategies for the development of therapeutic opportunities for rare diseases. In this article, we discuss the scientific, regulatory, and economic aspects of the development of repurposed drugs for the treatment of rare neurodegenerative disorders with a particular focus on Huntington’s disease, Friedreich’s ataxia, Wolfram syndrome, and amyotrophic lateral sclerosis. The role of academia, pharmaceutical companies, patient associations, and foundations in the identification of candidate compounds and their preclinical and clinical evaluation will also be discussed.
Collapse
Affiliation(s)
- Sweta Shah
- Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | |
Collapse
|
49
|
Kilikevicius A, Wang J, Shen X, Rigo F, Prakash TP, Napierala M, Corey DR. Difficulties translating antisense-mediated activation of Frataxin expression from cell culture to mice. RNA Biol 2021; 19:364-372. [PMID: 35289725 PMCID: PMC8928816 DOI: 10.1080/15476286.2022.2043650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/11/2022] [Indexed: 11/09/2022] Open
Abstract
Friedreich's ataxia (FA) is an inherited neurodegenerative disorder caused by decreased expression of frataxin (FXN) protein. Previous studies have shown that antisense oligonucleotides (ASOs) and single-stranded silencing RNAs can be used to increase expression of frataxin in cultured patient-derived cells. In this study, we investigate the potential for oligonucleotides to increase frataxin expression in a mouse model for FA. After confirming successful in vivo delivery of oligonucleotides using a benchmark gapmer targeting the nuclear noncoding RNA Malat1, we tested anti-FXN oligonucleotides designed to function by various mechanisms. None of these strategies yielded enhanced expression of FXN in the model mice. Our inability to translate activation of FXN expression from cell culture to mice may be due to inadequate potency of our compounds or differences in the molecular mechanisms governing FXN gene repression and activation in FA model mice.
Collapse
Affiliation(s)
- Audrius Kilikevicius
- Department of Pharmacology and Biochemistry, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Jun Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, Alabama, USA
| | - Xiulong Shen
- Department of Pharmacology and Biochemistry, UT Southwestern Medical Center, Dallas, Texas, United States
| | - Frank Rigo
- Ionis Pharmaceuticals, Medicinal Chemistry and Antisense Research, Carlsbad, California, USA
| | - Thahza P. Prakash
- Ionis Pharmaceuticals, Medicinal Chemistry and Antisense Research, Carlsbad, California, USA
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, Alabama, USA
- Department of Neurology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - David R. Corey
- Department of Pharmacology and Biochemistry, UT Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
50
|
Seminotti B, Grings M, Tucci P, Leipnitz G, Saso L. Nuclear Factor Erythroid-2-Related Factor 2 Signaling in the Neuropathophysiology of Inherited Metabolic Disorders. Front Cell Neurosci 2021; 15:785057. [PMID: 34955754 PMCID: PMC8693715 DOI: 10.3389/fncel.2021.785057] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/05/2021] [Indexed: 01/14/2023] Open
Abstract
Inherited metabolic disorders (IMDs) are rare genetic conditions that affect multiple organs, predominantly the central nervous system. Since treatment for a large number of IMDs is limited, there is an urgent need to find novel therapeutical targets. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a transcription factor that has a key role in controlling the intracellular redox environment by regulating the expression of antioxidant enzymes and several important genes related to redox homeostasis. Considering that oxidative stress along with antioxidant system alterations is a mechanism involved in the neuropathophysiology of many IMDs, this review focuses on the current knowledge about Nrf2 signaling dysregulation observed in this group of disorders characterized by neurological dysfunction. We review here Nrf2 signaling alterations observed in X-linked adrenoleukodystrophy, glutaric acidemia type I, hyperhomocysteinemia, and Friedreich’s ataxia. Additionally, beneficial effects of different Nrf2 activators are shown, identifying a promising target for treatment of patients with these disorders. We expect that this article stimulates research into the investigation of Nrf2 pathway involvement in IMDs and the use of potential pharmacological modulators of this transcription factor to counteract oxidative stress and exert neuroprotection.
Collapse
Affiliation(s)
- Bianca Seminotti
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mateus Grings
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Guilhian Leipnitz
- Postgraduate Program in Biological Sciences: Biochemistry, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.,Postgraduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|