1
|
Ercanbrack WS, Ramirez M, Dungan A, Gaul E, Ercanbrack SJ, Wingert RA. Frataxin deficiency and the pathology of Friedreich's Ataxia across tissues. Tissue Barriers 2025:2462357. [PMID: 39981684 DOI: 10.1080/21688370.2025.2462357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 02/22/2025] Open
Abstract
Friedreich's Ataxia (FRDA) is a neurodegenerative disease that affects a variety of different organ systems. The disease is caused by GAA repeat expansions in intron 1 of the Frataxin gene (FXN), which results in a decrease in the expression of the FXN protein. FXN is needed for the biogenesis of iron-sulfur clusters (ISC) which are required by key metabolic processes in the mitochondria. Without ISCs those processes do not occur properly. As a result, reactive oxygen species accumulate, and the mitochondria cease to function. Iron is also thought to accumulate in the cells of certain tissue types. These processes are thought to be intimately related to the pathologies affecting a myriad of tissues in FRDA. Most FRDA patients suffer from loss of motor control, cardiomyopathy, scoliosis, foot deformities, and diabetes. In this review, we discuss the known features of FRDA pathology and the current understanding about the basis of these alterations.
Collapse
Affiliation(s)
- Wesley S Ercanbrack
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Mateo Ramirez
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Austin Dungan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Ella Gaul
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Sarah J Ercanbrack
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
2
|
Harding IH, Nur Karim MI, Selvadurai LP, Corben LA, Delatycki MB, Monti S, Saccà F, Georgiou-Karistianis N, Cocozza S, Egan GF. Localized Changes in Dentate Nucleus Shape and Magnetic Susceptibility in Friedreich Ataxia. Mov Disord 2024; 39:1109-1118. [PMID: 38644761 DOI: 10.1002/mds.29816] [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: 11/13/2023] [Revised: 03/07/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND The dentate nuclei of the cerebellum are key sites of neuropathology in Friedreich ataxia (FRDA). Reduced dentate nucleus volume and increased mean magnetic susceptibility, a proxy of iron concentration, have been reported by magnetic resonance imaging studies in people with FRDA. Here, we investigate whether these changes are regionally heterogeneous. METHODS Quantitative susceptibility mapping data were acquired from 49 people with FRDA and 46 healthy controls. The dentate nuclei were manually segmented and analyzed using three dimensional vertex-based shape modeling and voxel-based assessments to identify regional changes in morphometry and susceptibility, respectively. RESULTS Individuals with FRDA, relative to healthy controls, showed significant bilateral surface contraction most strongly at the rostral and caudal boundaries of the dentate nuclei. The magnitude of this surface contraction correlated with disease duration, and to a lesser extent, ataxia severity. Significantly greater susceptibility was also evident in the FRDA cohort relative to controls, but was instead localized to bilateral dorsomedial areas, and also correlated with disease duration and ataxia severity. CONCLUSIONS Changes in the structure of the dentate nuclei in FRDA are not spatially uniform. Atrophy is greatest in areas with high gray matter density, whereas increases in susceptibility-reflecting iron concentration, demyelination, and/or gliosis-predominate in the medial white matter. These findings converge with established histological reports and indicate that regional measures of dentate nucleus substructure are more sensitive measures of disease expression than full-structure averages. Biomarker development and therapeutic strategies that directly target the dentate nuclei, such as gene therapies, may be optimized by targeting these areas of maximal pathology. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ian H Harding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Muhammad Ikhsan Nur Karim
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Louisa P Selvadurai
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Louise A Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Pediatrics, University of Melbourne, Parkville, Australia
- Turner Institute for Brain and Mental Health and School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Martin B Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Australia
- Department of Pediatrics, University of Melbourne, Parkville, Australia
| | - Serena Monti
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Francesco Saccà
- Neurosciences and Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Nellie Georgiou-Karistianis
- Turner Institute for Brain and Mental Health and School of Psychological Sciences, Monash University, Melbourne, Australia
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Melbourne, Australia
| |
Collapse
|
3
|
Koka M, Li H, Akther R, Perlman S, Wong D, Fogel BL, Lynch DR, Chandran V. Long non-coding RNA TUG1 is downregulated in Friedreich's ataxia. Brain Commun 2024; 6:fcae170. [PMID: 38846537 PMCID: PMC11154142 DOI: 10.1093/braincomms/fcae170] [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: 10/02/2023] [Revised: 03/25/2024] [Accepted: 05/14/2024] [Indexed: 06/09/2024] Open
Abstract
Friedreich's ataxia is a neurodegenerative disorder caused by reduced frataxin levels. It leads to motor and sensory impairments and has a median life expectancy of around 35 years. As the most common inherited form of ataxia, Friedreich's ataxia lacks reliable, non-invasive biomarkers, prolonging and inflating the cost of clinical trials. This study proposes TUG1, a long non-coding RNA, as a promising blood-based biomarker for Friedreich's ataxia, which is known to regulate various cellular processes. In a previous study using a frataxin knockdown mouse model, we observed several hallmark Friedreich's ataxia symptoms. Building on this, we hypothesized that a dual-source approach-comparing the data from peripheral blood samples from Friedreich's ataxia patients with tissue samples from affected areas in Friedreich's ataxia knockdown mice, tissues usually unattainable from patients-would effectively identify robust biomarkers. A comprehensive reanalysis was conducted on gene expression data from 183 age- and sex-matched peripheral blood samples of Friedreich's ataxia patients, carriers and controls and 192 tissue data sets from Friedreich's ataxia knockdown mice. Blood and tissue samples underwent RNA isolation and quantitative reverse transcription polymerase chain reaction, and frataxin knockdown was confirmed through enzyme-linked immunosorbent assays. Tug1 RNA interaction was explored via RNA pull-down assays. Validation was performed in serum samples on an independent set of 45 controls and 45 Friedreich's ataxia patients and in blood samples from 66 heterozygous carriers and 72 Friedreich's ataxia patients. Tug1 and Slc40a1 emerged as potential blood-based biomarkers, confirmed in the Friedreich's ataxia knockdown mouse model (one-way ANOVA, P ≤ 0.05). Tug1 was consistently downregulated after Fxn knockdown and correlated strongly with Fxn levels (R 2 = 0.71 during depletion, R 2 = 0.74 during rescue). Slc40a1 showed a similar but tissue-specific pattern. Further validation of Tug1's downstream targets strengthened its biomarker candidacy. In additional human samples, TUG1 levels were significantly downregulated in both whole blood and serum of Friedreich's ataxia patients compared with controls (Wilcoxon signed-rank test, P < 0.05). Regression analyses revealed a negative correlation between TUG1 fold-change and disease onset (P < 0.0037) and positive correlations with disease duration and functional disability stage score (P < 0.04). This suggests that elevated TUG1 levels correlate with earlier onset and more severe cases. This study identifies TUG1 as a potential blood-based biomarker for Friedreich's ataxia, showing consistent expression variance in human and mouse tissues related to disease severity and key Friedreich's ataxia pathways. It correlates with frataxin levels, indicating its promise as an early, non-invasive marker. TUG1 holds potential for Friedreich's ataxia monitoring and therapeutic development, meriting additional research.
Collapse
Affiliation(s)
- Mert Koka
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Hui Li
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rumana Akther
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Susan Perlman
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Darice Wong
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Clinical Neurogenomics Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Brent L Fogel
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Clinical Neurogenomics Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - David R Lynch
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Vijayendran Chandran
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Neuroscience, College of Medicine, University of Florida, and McKnight Brain Institute, Gainesville, FL 32610, USA
| |
Collapse
|
4
|
Pandolfo M, Reetz K, Darling A, Rodriguez de Rivera FJ, Henry PG, Joers J, Lenglet C, Adanyeguh I, Deelchand D, Mochel F, Pousset F, Pascual S, Van den Eede D, Martin-Ugarte I, Vilà-Brau A, Mantilla A, Pascual M, Martinell M, Meya U, Durr A. Efficacy and Safety of Leriglitazone in Patients With Friedreich Ataxia. Neurol Genet 2022; 8:e200034. [DOI: 10.1212/nxg.0000000000200034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/18/2022] [Indexed: 11/06/2022]
Abstract
Background and ObjectivesFriedreich ataxia (FRDA) is an autosomal recessive ataxia with no approved treatments. Leriglitazone is a selective peroxisome proliferator–activated receptor γ agonist that crosses the blood-brain barrier and, in preclinical models, improved mitochondrial function and energy production. We assessed effects of leriglitazone in patients with FRDA in a proof-of-concept study.MethodsIn this double-blind, randomized controlled trial, eligible participants (age 12–60 years) had genetically confirmed FRDA, a Scale for the Assessment and Rating of Ataxia (SARA) total score <25, and a SARA item 1 score of 2–6, inclusive. Key exclusion criteria were age at FRDA onset ≥25 years and history of cardiac dysfunction. Participants were randomly assigned (2:1) to receive a daily, oral, individualized dose of leriglitazone or placebo for 48 weeks. The primary endpoint was the change from baseline to week 48 in spinal cord area (C2-C3) (measured by MRI). Secondary endpoints included the change from baseline to week 48 in iron accumulation in the dentate nucleus (quantitative susceptibility mapping) and totalN-acetylaspartate to myo-inositol (tNAA/mIns) ratio.ResultsOverall, 39 patients were enrolled (mean age 24 years; 43.6% women; mean time since symptom onset 10.5 years): 26 patients received leriglitazone (20 completed) and 13 received placebo (12 completed). There was no difference between groups in spinal cord area from baseline to week 48 (least-squares [LS] mean change [standard error (SE)]: leriglitazone, −0.39 [0.55] mm2; placebo, 0.08 [0.72] mm2;p= 0.61). Iron accumulation in the dentate nucleus was greater with placebo (LS mean change [SE]: leriglitazone, 0.10 [1.33] ppb; placebo, 4.86 [1.84] ppb;p= 0.05), and a numerical difference was seen in tNAA/mIns ratio (LS mean change [SE]: leriglitazone, 0.03 [0.02]; placebo, −0.02 [0.03];p= 0.25). The most frequent adverse event was peripheral edema (leriglitazone 73.1%, placebo 0%).DiscussionThe primary endpoint of change in spinal cord area was not met. Secondary endpoints provide evidence supporting proof of concept for leriglitazone mode of action and, with acceptable safety data, support larger studies in patients with FRDA.Trial Registration InformationClinicalTrials.gov:NCT03917225; EudraCT: 2018-004405-64; submitted April 17, 2019; first patient enrolled April 2, 2019.clinicaltrials.gov/ct2/show/NCT03917225?term=NCT03917225&draw=2&rank=1.Classification of EvidenceThis study provides Class I evidence that individualized dosing of leriglitazone, compared with placebo, is not associated with changes in spinal cord area in patients with FRDA.
Collapse
|
5
|
Vavla M, Arrigoni F, Peruzzo D, Montanaro D, Frijia F, Pizzighello S, De Luca A, Della Libera E, Tessarotto F, Guerra P, Harding IH, Martinuzzi A. Functional MRI Studies in Friedreich's Ataxia: A Systematic Review. Front Neurol 2022; 12:802496. [PMID: 35360279 PMCID: PMC8960250 DOI: 10.3389/fneur.2021.802496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an inherited neurodegenerative movement disorder with early onset, widespread cerebral and cerebellar pathology, and no cure still available. Functional MRI (fMRI) studies, although currently limited in number, have provided a better understanding of brain changes in people with FRDA. This systematic review aimed to provide a critical overview of the findings and methodologies of all fMRI studies conducted in genetically confirmed FRDA so far, and to offer recommendations for future study designs. About 12 cross-sectional and longitudinal fMRI studies, included 198 FRDA children and young adult patients and, 205 healthy controls (HCs), according to the inclusion criteria. Details regarding GAA triplet expansion and demographic and clinical severity measures were widely reported. fMRI designs included motor and cognitive task paradigms, and resting-state studies, with widespread changes in functionally activated areas and extensive variability in study methodologies. These studies highlight a mixed picture of both hypoactivation and hyperactivation in different cerebral and cerebellar brain regions depending on fMRI design and cohort characteristics. Functional changes often correlate with clinical variables. In aggregate, the findings provide support for cerebro-cerebellar loop damage and the compensatory mechanism hypothesis. Current literature indicates that fMRI is a valuable tool for gaining in vivo insights into FRDA pathology, but addressing that its limitations would be a key to improving the design, interpretation, and generalizability of studies in the future.
Collapse
Affiliation(s)
- Marinela Vavla
- Department of Neurorehabilitation, Pieve di Soligo, Scientific Institute, IRCCS E. Medea, Pieve di Soligo, Italy
- *Correspondence: Marinela Vavla ;
| | - Filippo Arrigoni
- Neuroimaging Lab, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Denis Peruzzo
- Neuroimaging Lab, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Domenico Montanaro
- U.O.C. Risonanza Magnetica Specialistica e Neuroradiologia, Fondazione CNR/Regione Toscana G. Monasterio, Pisa, Italy
- U.O.S.D. Servizio Autonomo di Risonanza Magnetica, Dipartimento Clinico di Neuroscienze dell'Età Evolutiva - IRCCS Fondazione Stella Maris - Pisa, Italy
| | - Francesca Frijia
- U.O.C. Bioingegneria e Ing. Clinica, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Silvia Pizzighello
- Department of Neurorehabilitation, Pieve di Soligo, Scientific Institute, IRCCS E. Medea, Pieve di Soligo, Italy
| | - Alberto De Luca
- Department of Neurology, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, Netherlands
| | | | - Federica Tessarotto
- Department of Neurorehabilitation, Pieve di Soligo, Scientific Institute, IRCCS E. Medea, Pieve di Soligo, Italy
| | - Paola Guerra
- Department of Neurorehabilitation, Pieve di Soligo, Scientific Institute, IRCCS E. Medea, Pieve di Soligo, Italy
| | - Ian H. Harding
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Andrea Martinuzzi
- Department of Neurorehabilitation, Pieve di Soligo, Scientific Institute, IRCCS E. Medea, Pieve di Soligo, Italy
| |
Collapse
|
6
|
Deistung A, Jäschke D, Draganova R, Pfaffenrot V, Hulst T, Steiner KM, Thieme A, Giordano IA, Klockgether T, Tunc S, Münchau A, Minnerop M, Göricke SL, Reichenbach JR, Timmann D. Quantitative susceptibility mapping reveals alterations of dentate nuclei in common types of degenerative cerebellar ataxias. Brain Commun 2022; 4:fcab306. [PMID: 35291442 PMCID: PMC8914888 DOI: 10.1093/braincomms/fcab306] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 10/28/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
The cerebellar nuclei are a brain region with high iron content. Surprisingly,
little is known about iron content in the cerebellar nuclei and its possible
contribution to pathology in cerebellar ataxias, with the only exception of
Friedreich’s ataxia. In the present exploratory cross-sectional study,
quantitative susceptibility mapping was used to investigate volume, iron
concentration and total iron content of the dentate nuclei in common types of
hereditary and non-hereditary degenerative ataxias. Seventy-nine patients with
spinocerebellar ataxias of types 1, 2, 3 and 6; 15 patients with
Friedreich’s ataxia; 18 patients with multiple system atrophy, cerebellar
type and 111 healthy controls were also included. All underwent 3 T MRI
and clinical assessments. For each specific ataxia subtype, voxel-based and
volumes-of-interest-based group analyses were performed in comparison with a
corresponding age- and sex-matched control group, both for volume, magnetic
susceptiblity (indicating iron concentration) and susceptibility mass
(indicating total iron content) of the dentate nuclei. Spinocerebellar ataxia of
type 1 and multiple system atrophy, cerebellar type patients showed higher
susceptibilities in large parts of the dentate nucleus but unaltered
susceptibility masses compared with controls. Friedreich’s ataxia
patients and, only on a trend level, spinocerebellar ataxia of type 2 patients
showed higher susceptibilities in more circumscribed parts of the dentate. In
contrast, spinocerebellar ataxia of type 6 patients revealed lower
susceptibilities and susceptibility masses compared with controls throughout the
dentate nucleus. Spinocerebellar ataxia of type 3 patients showed no significant
changes in susceptibility and susceptibility mass. Lower volume of the dentate
nuclei was found to varying degrees in all ataxia types. It was most pronounced
in spinocerebellar ataxia of type 6 patients and least prominent in
spinocerebellar ataxia of type 3 patients. The findings show that alterations in
susceptibility revealed by quantitative susceptibility mapping are common in the
dentate nuclei in different types of cerebellar ataxias. The most striking
changes in susceptibility were found in spinocerebellar ataxia of type 1,
multiple system atrophy, cerebellar type and spinocerebellar ataxia of type 6.
Because iron content is known to be high in glial cells but not in neurons of
the cerebellar nuclei, the higher susceptibility in spinocerebellar ataxia of
type 1 and multiple system atrophy, cerebellar type may be explained by a
reduction of neurons (increase in iron concentration) and/or an increase in
iron-rich glial cells, e.g. microgliosis. Hypomyelination also leads to higher
susceptibility and could also contribute. The lower susceptibility in SCA6
suggests a loss of iron-rich glial cells. Quantitative susceptibility maps
warrant future studies of iron content and iron-rich cells in ataxias to gain a
more comprehensive understanding of the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Andreas Deistung
- University Clinic and Outpatient Clinic for Radiology, Department for Radiation Medicine, University Hospital Halle (Saale), Halle (Saale), Germany
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| | - Dominik Jäschke
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| | - Rossitza Draganova
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| | - Viktor Pfaffenrot
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, University Duisburg-Essen, Essen, Germany
| | - Thomas Hulst
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
- Erasmus University College, Erasmus School of Social and Behavioural Sciences, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Katharina M. Steiner
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| | - Ilaria A. Giordano
- Department of Neurology, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thomas Klockgether
- Department of Neurology, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sinem Tunc
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Alexander Münchau
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Martina Minnerop
- Institute of Neuroscience and Medicine (INM-1), Research Center Juelich, Juelich, Germany
- Department of Neurology, Center for Movement Disorders and Neuromodulation, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Duesseldorf, Germany
| | - Sophia L. Göricke
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, Essen University Hospital, Essen, Germany
| | - Jürgen R. Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Dagmar Timmann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Essen University Hospital, Essen, Germany
| |
Collapse
|
7
|
Salegio EA, Cukrov M, Lortz R, Green A, Lambert E, Copeland S, Gonzalez M, Stockinger DE, Yeung JM, Hwa GGC. Feasibility of Targeted Delivery of AAV5-GFP into the Cerebellum of Nonhuman Primates Following a Single Convection-Enhanced Delivery Infusion. Hum Gene Ther 2022; 33:86-93. [PMID: 34779239 DOI: 10.1089/hum.2021.163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In this study, we built upon our previous work to demonstrate the distribution and transport of AAV5-green fluorescent protein (GFP) following a single convection-enhanced delivery infusion into the nonhuman primate cerebellum, with no untoward side effects noted. Dosing under magnetic resonance imaging guidance revealed a sixfold larger volume of distribution compared with the volume of infusion, with no evidence of reflux underscoring the convective properties of the cerebellum and step design of the cannula. Postmortem tissue analysis, 4 weeks post-adeno-associated viral (AAV) delivery, revealed the robust presence of the transgene in situ, with GFP detection in secondary regions not directly targeted by the infusion, denoting distal transport of the vector. Irrespective of tropism, a twofold larger area of transgene expression was found and was corroborated against the presence of contrast on T1-weighted images. Different levels of transduction were detected between animals, which were negatively correlated with the level of antibody titer against the GFP construct, whereby the higher the antibody titer, the lower the level of transgene expression. These findings support the use of the posterior fossa as a potential target site for direct delivery of gene-based therapeutics for cerebellar diseases.
Collapse
Affiliation(s)
| | - Mira Cukrov
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Rachel Lortz
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Abigail Green
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Emily Lambert
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | | - Marc Gonzalez
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | | - Jeremy M Yeung
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | |
Collapse
|
8
|
Abstract
INTRODUCTION Friedreich ataxia (FRDA) is an autosomal recessive disorder caused by deficiency of frataxin, an essential mitochondrial protein involved in iron sulfur cluster biogenesis, oxidative phosphorylation and other processes. FRDA most notably affects the heart, sensory neurons, spinal cord, cerebellum, and other brain regions, and manifests clinically as ataxia, sensory loss, dysarthria, spasticity, and hypertrophic cardiomyopathy. Therapeutic approaches in FRDA have consisted of two different approaches: (1) augmenting or restoring frataxin production and (2) modulating a variety of downstream processes related to mitochondrial dysfunction, including reactive oxygen species production, ferroptosis, or Nrf2 activation. AREAS COVERED In this review, we summarize data from major phase II clinical trials in FRDA published between 2015 and 2020, which includes A0001/EPI743, Omaveloxolone, RT001, and Actimmune. EXPERT OPINION A growing number of drug candidates are being tested in phase II clinical trials for FRDA; however, most have not met their primary endpoints, and none have received FDA approval. In this review, we aim to summarize completed phase II clinical trials in FRDA, outlining critical lessons that have been learned and that should be incorporated into future trial design to ultimately optimize drug development in FRDA.
Collapse
|
9
|
Abstract
OBJECTIVE Friedreich's ataxia (FRDA) is the most common hereditary ataxia. It is a neurodegenerative disorder, characterized by progressive ataxia. FRDA is also associated with cognitive impairments. To date, the evolution of cognitive functioning is unknown. Our aim was to investigate the changes in the cognitive functioning of FRDA patients over an average eight-year timeframe. In addition, we aimed to study the relationship between cognitive changes and clinical variables. METHODS Twenty-nine FRDA patients who had been part of the sample of a previous study participated in the present study. The mean average time between the two assessments was 8.24 years. The participants completed an extensive battery of neuropsychological tests chosen to examine cognitive functioning in various cognitive domains: processing speed, attention, working memory, executive functions, verbal and visual memory, visuoperceptive and visuospatial skills, visuoconstructive functions and language. RESULTS At follow-up, cerebellar symptoms had worsened, and patients presented greater disability. Differences between baseline and follow-up were observed in motor and cognitive reaction times, several trials of the Stroop test, semantic fluency, and block designs. No other cognitive changes were observed. Deterioration in simple cognitive reactions times and block designs performance correlated with the progression of cerebellar symptoms. CONCLUSIONS Our study has demonstrated for the first time that patients with FRDA experience a significant decline over time in several cognitive domains. Specifically, after an eight-year period, FRDA patients worsened in processing speed, fluency, and visuoconstructive skills. This progression is unlikely to be due to greater motor or speech impairment.
Collapse
|
10
|
Cocozza S, Pontillo G, De Michele G, Di Stasi M, Guerriero E, Perillo T, Pane C, De Rosa A, Ugga L, Brunetti A. Conventional MRI findings in hereditary degenerative ataxias: a pictorial review. Neuroradiology 2021; 63:983-999. [PMID: 33733696 PMCID: PMC8213578 DOI: 10.1007/s00234-021-02682-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022]
Abstract
Purpose Cerebellar ataxias are a large and heterogeneous group of disorders. The evaluation of brain parenchyma via MRI plays a central role in the diagnostic assessment of these conditions, being mandatory to exclude the presence of other underlying causes in determining the clinical phenotype. Once these possible causes are ruled out, the diagnosis is usually researched in the wide range of hereditary or sporadic ataxias. Methods We here propose a review of the main clinical and conventional imaging findings of the most common hereditary degenerative ataxias, to help neuroradiologists in the evaluation of these patients. Results Hereditary degenerative ataxias are all usually characterized from a neuroimaging standpoint by the presence, in almost all cases, of cerebellar atrophy. Nevertheless, a proper assessment of imaging data, extending beyond the mere evaluation of cerebellar atrophy, evaluating also the pattern of volume loss as well as concomitant MRI signs, is crucial to achieve a proper diagnosis. Conclusion The integration of typical neuroradiological characteristics, along with patient’s clinical history and laboratory data, could allow the neuroradiologist to identify some conditions and exclude others, addressing the neurologist to the more appropriate genetic testing.
Collapse
Affiliation(s)
- Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy.
| | - Giuseppe Pontillo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy.,Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Naples, Italy
| | - Giovanna De Michele
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Martina Di Stasi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Elvira Guerriero
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Teresa Perillo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Chiara Pane
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Anna De Rosa
- Department of Neurosciences and Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| |
Collapse
|
11
|
Straub S, Mangesius S, Emmerich J, Indelicato E, Nachbauer W, Degenhardt KS, Ladd ME, Boesch S, Gizewski ER. Toward quantitative neuroimaging biomarkers for Friedreich's ataxia at 7 Tesla: Susceptibility mapping, diffusion imaging, R 2 and R 1 relaxometry. J Neurosci Res 2020; 98:2219-2231. [PMID: 32731306 PMCID: PMC7590084 DOI: 10.1002/jnr.24701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/12/2020] [Accepted: 07/08/2020] [Indexed: 01/21/2023]
Abstract
Friedreich's ataxia (FRDA) is a rare genetic disorder leading to degenerative processes. So far, no effective treatment has been found. Therefore, it is important to assist the development of medication with imaging biomarkers reflecting disease status and progress. Ten FRDA patients (mean age 37 ± 14 years; four female) and 10 age- and sex-matched controls were included. Acquisition of magnetic resonance imaging (MRI) data for quantitative susceptibility mapping, R1 , R2 relaxometry and diffusion imaging was performed at 7 Tesla. Results of volume of interest (VOI)-based analyses of the quantitative data were compared with a voxel-based morphometry (VBM) evaluation. Differences between patients and controls were assessed using the analysis of covariance (ANCOVA; p < 0.01) with age and sex as covariates, effect size of group differences, and correlations with disease characteristics with Spearman correlation coefficient. For the VBM analysis, a statistical threshold of 0.001 for uncorrected and 0.05 for corrected p-values was used. Statistically significant differences between FRDA patients and controls were found in five out of twelve investigated structures, and statistically significant correlations with disease characteristics were revealed. Moreover, VBM revealed significant white matter atrophy within regions of the brainstem, and the cerebellum. These regions overlapped partially with brain regions for which significant differences between healthy controls and patients were found in the VOI-based quantitative MRI evaluation. It was shown that two independent analyses provided overlapping results. Moreover, positive results on correlations with disease characteristics were found, indicating that these quantitative MRI parameters could provide more detailed information and assist the search for effective treatments.
Collapse
Affiliation(s)
- Sina Straub
- Division of Medical Physics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Mangesius
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria.,Neuroimaging Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| | - Julian Emmerich
- Division of Medical Physics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | | | - Wolfgang Nachbauer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katja S Degenhardt
- Division of Medical Physics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Mark E Ladd
- Division of Medical Physics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Physics and Astronomy, Heidelberg University, Heidelberg, Germany.,Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Sylvia Boesch
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke R Gizewski
- Department of Neuroradiology, Medical University of Innsbruck, Innsbruck, Austria.,Neuroimaging Core Facility, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
12
|
Kim Y, Connor JR. The roles of iron and HFE genotype in neurological diseases. Mol Aspects Med 2020; 75:100867. [PMID: 32654761 DOI: 10.1016/j.mam.2020.100867] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Iron accumulation is a recurring pathological phenomenon in many neurological diseases including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and others. Iron is essential for normal development and functions of the brain; however, excess redox-active iron can also lead to oxidative damage and cell death. Especially for terminally differentiated cells like neurons, regulation of reactive oxygen species is critical for cell viability. As a result, cellular iron level is tightly regulated. Although iron accumulation related to neurological diseases has been well documented, the pathoetiological contributions of the homeostatic iron regulator (HFE), which controls cellular iron uptake, is less understood. Furthermore, a common HFE variant, H63D HFE, has been identified as a modifier of multiple neurological diseases. This review will discuss the roles of iron and HFE in the brain as well as their impact on various disease processes.
Collapse
Affiliation(s)
- Yunsung Kim
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA
| | - James R Connor
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA.
| |
Collapse
|
13
|
Integrated analysis of the molecular pathogenesis of FDXR-associated disease. Cell Death Dis 2020; 11:423. [PMID: 32499495 PMCID: PMC7272433 DOI: 10.1038/s41419-020-2637-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
The mitochondrial flavoprotein ferredoxin reductase (FDXR) is required for biogenesis of iron-sulfur clusters and for steroidogenesis. Iron-sulfur (Fe-S) clusters are ubiquitous cofactors essential to various cellular processes, and an increasing number of disorders are associated with disruptions in the synthesis of Fe-S clusters. Our previous studies have demonstrated that hypomorphic mutations in FDXR cause a novel mitochondriopathy and optic atrophy in humans and mice, attributed in part to reduced function of the electron transport chain (ETC) as well as elevated production of reactive oxygen species (ROS). Inflammation and peripheral neuropathy are also hallmarks of this disease. In this paper, we demonstrate that FDXR mutation leads to significant optic transport defects that are likely to underlie optic atrophy, a major clinical presentation in FDXR patients, as well as a neurodegenerative loss of cells in the central nervous system (CNS). Molecular analysis indicates that FDXR mutation also leads to mitochondrial iron overload and an associated depolarization of the mitochondrial membrane, further supporting the hypothesis that FDXR mutations cause neurodegeneration by affecting FDXR's critical role in iron homeostasis.
Collapse
|
14
|
Pattern of Cerebellar Atrophy in Friedreich's Ataxia-Using the SUIT Template. THE CEREBELLUM 2019; 18:435-447. [PMID: 30771164 DOI: 10.1007/s12311-019-1008-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Whole-brain voxel-based morphometry (VBM) studies revealed patterns of patchy atrophy within the cerebellum of Friedreich's ataxia patients, missing clear clinico-anatomic correlations. Studies so far are lacking an appropriate registration to the infratentorial space. To circumvent these limitations, we applied a high-resolution atlas template of the human cerebellum and brainstem (SUIT template) to characterize regional cerebellar atrophy in Friedreich's ataxia (FRDA) on 3-T MRI data. We used a spatially unbiased voxel-based morphometry approach together with T2-based manual segmentation, T2 histogram analysis, and atlas generation of the dentate nuclei in a representative cohort of 18 FRDA patients and matched healthy controls. We demonstrate that the cerebellar volume in FRDA is generally not significantly different from healthy controls but mild lobular atrophy develops beyond normal aging. The medial parts of lobule VI, housing the somatotopic representation of tongue and lips, are the major site of this lobular atrophy, which possibly reflects speech impairment. Extended white matter affection correlates with disease severity across and beyond the cerebellar inflow and outflow tracts. The dentate nucleus, as a major site of cerebellar degeneration, shows a mean volume loss of about 30%. Remarkably, not the atrophy but the T2 signal decrease of the dentate nuclei highly correlates with disease duration and severity.
Collapse
|
15
|
Cerebellum and cognition in Friedreich ataxia: a voxel-based morphometry and volumetric MRI study. J Neurol 2019; 267:350-358. [PMID: 31641877 DOI: 10.1007/s00415-019-09582-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/25/2019] [Accepted: 10/14/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recent studies have suggested the presence of a significant atrophy affecting the cerebellar cortex in Friedreich ataxia (FRDA) patients, an area of the brain long considered to be relatively spared by neurodegenerative phenomena. Cognitive deficits, which occur in FRDA patients, have been associated with cerebellar volume loss in other conditions. The aim of this study was to investigate the correlation between cerebellar volume and cognition in FRDA. METHODS Nineteen FRDA patients and 20 healthy controls (HC) were included in this study and evaluated via a neuropsychological examination. Cerebellar global and lobular volumes were computed using the Spatially Unbiased Infratentorial Toolbox (SUIT). Furthermore, a cerebellar voxel-based morphometry (VBM) analysis was also carried out. Correlations between MRI metrics and clinical data were tested via partial correlation analysis. RESULTS FRDA patients showed a significant reduction of the total cerebellar volume (p = 0.004), significantly affecting the Lobule IX (p = 0.001). At the VBM analysis, we found a cluster of significant reduced GM density encompassing the entire lobule IX (p = 0.003). When correlations were probed, we found a direct correlation between Lobule IX volume and impaired visuo-spatial functions (r = 0.58, p = 0.02), with a similar correlation that was found between the same altered function and results obtained at the VBM (r = 0.52; p = 0.03). CONCLUSIONS With two different image analysis techniques, we confirmed the presence of cerebellar volume loss in FRDA, mainly affecting the posterior lobe. In particular, Lobule IX atrophy correlated with worse visuo-spatial abilities, further expanding our knowledge about the physiopathology of cognitive impairment in FRDA.
Collapse
|
16
|
Bermudez Noguera C, Bao S, Petersen KJ, Lopez AM, Reid J, Plassard AJ, Zald DH, Claassen DO, Dawant BM, Landman BA. Using deep learning for a diffusion-based segmentation of the dentate nucleus and its benefits over atlas-based methods. J Med Imaging (Bellingham) 2019; 6:044007. [PMID: 31824980 PMCID: PMC6895566 DOI: 10.1117/1.jmi.6.4.044007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/18/2019] [Indexed: 01/17/2023] Open
Abstract
The dentate nucleus (DN) is a gray matter structure deep in the cerebellum involved in motor coordination, sensory input integration, executive planning, language, and visuospatial function. The DN is an emerging biomarker of disease, informing studies that advance pathophysiologic understanding of neurodegenerative and related disorders. The main challenge in defining the DN radiologically is that, like many deep gray matter structures, it has poor contrast in T1-weighted magnetic resonance (MR) images and therefore requires specialized MR acquisitions for visualization. Manual tracing of the DN across multiple acquisitions is resource-intensive and does not scale well to large datasets. We describe a technique that automatically segments the DN using deep learning (DL) on common imaging sequences, such as T1-weighted, T2-weighted, and diffusion MR imaging. We trained a DL algorithm that can automatically delineate the DN and provide an estimate of its volume. The automatic segmentation achieved higher agreement to the manual labels compared to template registration, which is the current common practice in DN segmentation or multiatlas segmentation of manual labels. Across all sequences, the FA maps achieved the highest mean Dice similarity coefficient (DSC) of 0.83 compared to T1 imaging ( DSC = 0.76 ), T2 imaging ( DSC = 0.79 ), or a multisequence approach ( DSC = 0.80 ). A single atlas registration approach using the spatially unbiased atlas template of the cerebellum and brainstem template achieved a DSC of 0.23, and multi-atlas segmentation achieved a DSC of 0.33. Overall, we propose a method of delineating the DN on clinical imaging that can reproduce manual labels with higher accuracy than current atlas-based tools.
Collapse
Affiliation(s)
- Camilo Bermudez Noguera
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Shunxing Bao
- Vanderbilt University, Department of Electrical Engineering and Computer Science, Nashville, Tennessee, United States
| | - Kalen J. Petersen
- Vanderbilt University, Department of Neurology, Nashville, Tennessee, United States
| | - Alexander M. Lopez
- Vanderbilt University, Department of Neurology, Nashville, Tennessee, United States
| | - Jacqueline Reid
- Vanderbilt University, Department of Neurology, Nashville, Tennessee, United States
| | - Andrew J. Plassard
- Vanderbilt University, Department of Electrical Engineering and Computer Science, Nashville, Tennessee, United States
| | - David H. Zald
- Vanderbilt University, Department of Psychology and Psychiatry, Nashville, Tennessee, United States
| | - Daniel O. Claassen
- Vanderbilt University, Department of Neurology, Nashville, Tennessee, United States
| | - Benoit M. Dawant
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical Engineering and Computer Science, Nashville, Tennessee, United States
| | - Bennett A. Landman
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical Engineering and Computer Science, Nashville, Tennessee, United States
- Vanderbilt University, Department of Psychology and Psychiatry, Nashville, Tennessee, United States
| |
Collapse
|
17
|
Llorens JV, Soriano S, Calap-Quintana P, Gonzalez-Cabo P, Moltó MD. The Role of Iron in Friedreich's Ataxia: Insights From Studies in Human Tissues and Cellular and Animal Models. Front Neurosci 2019; 13:75. [PMID: 30833885 PMCID: PMC6387962 DOI: 10.3389/fnins.2019.00075] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2019] [Indexed: 12/12/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a rare early-onset degenerative disease that affects both the central and peripheral nervous systems, and other extraneural tissues, mainly the heart and endocrine pancreas. This disorder progresses as a mixed sensory and cerebellar ataxia, primarily disturbing the proprioceptive pathways in the spinal cord, peripheral nerves and nuclei of the cerebellum. FRDA is an inherited disease with an autosomal recessive pattern caused by an insufficient amount of the nuclear-encoded mitochondrial protein frataxin, which is an essential and highly evolutionary conserved protein whose deficit results in iron metabolism dysregulation and mitochondrial dysfunction. The first experimental evidence connecting frataxin with iron homeostasis came from Saccharomyces cerevisiae; iron accumulates in the mitochondria of yeast with deletion of the frataxin ortholog gene. This finding was soon linked to previous observations of iron deposits in the hearts of FRDA patients and was later reported in animal models of the disease. Despite advances made in the understanding of FRDA pathophysiology, the role of iron in this disease has not yet been completely clarified. Some of the questions still unresolved include the molecular mechanisms responsible for the iron accumulation and iron-mediated toxicity. Here, we review the contribution of the cellular and animal models of FRDA and relevance of the studies using FRDA patient samples to gain knowledge about these issues. Mechanisms of mitochondrial iron overload are discussed considering the potential roles of frataxin in the major mitochondrial metabolic pathways that use iron. We also analyzed the effect of iron toxicity on neuronal degeneration in FRDA by reactive oxygen species (ROS)-dependent and ROS-independent mechanisms. Finally, therapeutic strategies based on the control of iron toxicity are considered.
Collapse
Affiliation(s)
- José Vicente Llorens
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Sirena Soriano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Pablo Calap-Quintana
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
- Center of Biomedical Network Research on Rare Diseases CIBERER, Valencia, Spain
- Associated Unit for Rare Diseases INCLIVA-CIPF, Biomedical Research Institute INCLIVA, Valencia, Spain
| | - María Dolores Moltó
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, Valencia, Spain
- Unit for Psychiatry and Neurodegenerative Diseases, Biomedical Research Institute INCLIVA, Valencia, Spain
- Center of Biomedical Network Research on Mental Health CIBERSAM, Valencia, Spain
| |
Collapse
|
18
|
Ward PGD, Harding IH, Close TG, Corben LA, Delatycki MB, Storey E, Georgiou-Karistianis N, Egan GF. Longitudinal evaluation of iron concentration and atrophy in the dentate nuclei in friedreich ataxia. Mov Disord 2019; 34:335-343. [PMID: 30624809 DOI: 10.1002/mds.27606] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/26/2018] [Accepted: 12/06/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Friedreich ataxia is a recessively inherited, progressive neurological disease characterized by impaired mitochondrial iron metabolism. The dentate nuclei of the cerebellum are characteristic sites of neurodegeneration in the disease, but little is known of the longitudinal progression of abnormalities in these structures. METHODS Using in vivo magnetic resonance imaging, including quantitative susceptibility mapping, we investigated changes in iron concentration and volume in the dentate nuclei in individuals with Friedreich ataxia (n = 20) and healthy controls (n = 18) over a 2-year period. RESULTS The longitudinal rate of iron concentration was significantly elevated bilaterally in participants with Friedreich ataxia relative to healthy controls. Atrophy rates did not differ significantly between groups. Change in iron concentration and atrophy both correlated with baseline disease severity or duration, indicating sensitivity of these measures to disease stage. Specifically, atrophy was maximal in individuals early in the disease course, whereas the rate of iron concentration increased with disease progression. CONCLUSIONS Progressive dentate nucleus abnormalities are evident in vivo in Friedreich ataxia, and the rates of change of iron concentration and atrophy in these structures are sensitive to the disease stage. The findings are consistent with an increased rate of iron concentration and atrophy early in the disease, followed by iron accumulation and stable volume in later stages. This pattern suggests that iron dysregulation persists after loss of the vulnerable neurons in the dentate. The significant changes observed over a 2-year period highlight the utility of quantitative susceptibility mapping as a longitudinal biomarker and staging tool. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Phillip G D Ward
- Monash Biomedical Imaging, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne, Australia
| | - Ian H Harding
- School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| | - Thomas G Close
- Monash Biomedical Imaging, Monash University, Melbourne, Australia
| | - Louise A Corben
- School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Martin B Delatycki
- School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Australia.,Victorian Clinical Genetics Service, Parkville, Australia
| | - Elsdon Storey
- Department of Medicine, Monash University, Melbourne, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Melbourne, Australia.,School of Psychological Sciences and Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Melbourne, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne, Australia
| |
Collapse
|
19
|
A new MRI marker of ataxia with oculomotor apraxia. Eur J Radiol 2018; 110:187-192. [PMID: 30599859 DOI: 10.1016/j.ejrad.2018.11.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/23/2018] [Accepted: 11/28/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Evaluate the specificity and sensitivity of disappearance of susceptibility weighted imaging (SWI) dentate nuclei (DN) hypointensity in oculomotor apraxia patients (AOA). METHOD In this prospective study, 27 patients with autosomal genetic ataxia (AOA (n = 11), Friedreich ataxia and ataxia with vitamin E deficit (n = 4), and dominant genetic ataxia (n = 12)) were included along with fifteen healthy controls. MRIs were qualitatively classified for the presence or absence of DN hypointensity on FLAIR and SWI sequences. The MRIs were then quantitatively studied, with measurement of a ratio of DN over brainstem white matter signal intensity through manual delineation. The institutional review board approved this study, and written informed consent was obtained. In the cross-sectional analysis, the Mann-Whitney test was applied. RESULTS Qualitatively, the eleven AOA patients presented absence of both DN SWI and FLAIR hyposignals; three dominant genetic ataxia patients had moderate SWI DN hyposignal and absent FLAIR hyposignal; the thirteen remaining subjects presented normal SWI and FLAIR DN hyposignal. Absence of DN SWI hypointensity was 100% sensitive and specific to AOA. Quantitative signal intensity ratio (mean ± standard deviation) of the AOA group (98·96 ± 5·37%) was significantly higher than in control subjects group (76.40 ± 8.34%; p < 0.001), dominant genetic ataxia group (81·15 ± 9·94%; p < 0·001), and Friedreich ataxia and ataxia with vitamin E deficit group (87·56 ± 2·78%; p < 0·02). CONCLUSION This small study shows that loss of the normal hypointensity in the dentate nucleus on both SWI and FLAIR imaging at 3 T is a highly sensitive and specific biomarker for AOA.
Collapse
|
20
|
Mascalchi M, Vella A. Neuroimaging Applications in Chronic Ataxias. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 143:109-162. [PMID: 30473193 DOI: 10.1016/bs.irn.2018.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT) and positron emission tomography (PET) are the main instruments for neuroimaging investigation of patients with chronic ataxia. MRI has a predominant diagnostic role in the single patient, based on the visual detection of three patterns of atrophy, namely, spinal atrophy, cortical cerebellar atrophy and olivopontocerebellar atrophy, which correlate with the aetiologies of inherited or sporadic ataxia. In fact spinal atrophy is observed in Friedreich ataxia, cortical cerebellar atrophy in Ataxia Telangectasia, gluten ataxia and Sporadic Adult Onset Ataxia and olivopontocerebellar atrophy in Multiple System Atrophy cerebellar type. The 39 types of dominantly inherited spinocerebellar ataxias show either cortical cerebellar atrophy or olivopontocerebellar atrophy. T2 or T2* weighted MR images can contribute to the diagnosis by revealing abnormally increased or decreased signal with a characteristic distribution. These include symmetric T2 hyperintensity of the posterior and lateral columns of the cervical spinal cord in Friedreich ataxia, diffuse and symmetric hyperintensity of the cerebellar cortex in Infantile Neuro-Axonal Dystrophy, symmetric hyperintensity of the peridentate white matter in Cerebrotendineous Xanthomatosis, and symmetric hyperintensity of the middle cerebellar peduncles and peridentate white matter, cerebral white matter and corpus callosum in Fragile X Tremor Ataxia Syndrome. Abnormally decreased T2 or T2* signal can be observed with a multifocal distribution in Ataxia Telangectasia and with a symmetric distribution in the basal ganglia in Multiple System Atrophy. T2 signal hypointensity lining diffusely the outer surfaces of the brainstem, cerebellum and cerebrum enables diagnosis of superficial siderosis of the central nervous system. The diagnostic role of nuclear medicine techniques is smaller. SPECT and PET show decreased uptake of radiotracers investigating the nigrostriatal system in Multiple System Atrophy and in patients with Fragile X Tremor Ataxia Syndrome. Semiquantitative or quantitative MRI, SPECT and PET data describing structural, microstructural and functional changes of the cerebellum, brainstem, and spinal cord have been widely applied to investigate physiopathological changes in patients with chronic ataxias. Moreover they can track diseases progression with a greater sensitivity than clinical scales. So far, a few small-size and single center studies employed neuroimaging techniques as surrogate markers of treatment effects in chronic ataxias.
Collapse
Affiliation(s)
- Mario Mascalchi
- Meyer Children Hospital, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| | | |
Collapse
|
21
|
Slone J, Peng Y, Chamberlin A, Harris B, Kaylor J, McDonald MT, Lemmon M, El-Dairi MA, Tchapyjnikov D, Gonzalez-Krellwitz LA, Sellars EA, McConkie-Rosell A, Reinholdt LG, Huang T. Biallelic mutations in FDXR cause neurodegeneration associated with inflammation. J Hum Genet 2018; 63:1211-1222. [PMID: 30250212 DOI: 10.1038/s10038-018-0515-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/24/2018] [Accepted: 08/26/2018] [Indexed: 01/09/2023]
Abstract
Mitochondrial dysfunction lies behind many neurodegenerative disorders, owing largely to the intense energy requirements of most neurons. Such mitochondrial dysfunction may work through a variety of mechanisms, from direct disruption of the electron transport chain to abnormal mitochondrial biogenesis. Recently, we have identified biallelic mutations in the mitochondrial flavoprotein "ferredoxin reductase" (FDXR) gene as a novel cause of mitochondriopathy, peripheral neuropathy, and optic atrophy. In this report, we expand upon those results by describing two new cases of disease-causing FDXR variants in patients with variable severity of phenotypes, including evidence of an inflammatory response in brain autopsy. To investigate the underlying pathogenesis, we examined neurodegeneration in a mouse model. We found that Fdxr mutant mouse brain tissues share pathological changes similar to those seen in patient autopsy material, including increased astrocytes. Furthermore, we show that these abnormalities are associated with increased levels of markers for both neurodegeneration and gliosis, with the latter implying inflammation as a major factor in the pathology of Fdxr mutations. These data provide further insight into the pathogenic mechanism of FDXR-mediated central neuropathy, and suggest an avenue for mechanistic studies that will ultimately inform treatment.
Collapse
Affiliation(s)
- Jesse Slone
- Division of Human Genetics, Children's Hospital Medical Center, Cincinnati, OH, USA, 45229
| | - Yanyan Peng
- Division of Human Genetics, Children's Hospital Medical Center, Cincinnati, OH, USA, 45229
| | | | | | - Julie Kaylor
- University of Arkansas for Medical Sciences, Section of Genetics and Metabolism, 1 Children's Way, Little Rock, AR, USA, 72202
| | - Marie T McDonald
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA, 27710
| | - Monica Lemmon
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA, 27710
| | | | - Dmitry Tchapyjnikov
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA, 27710
| | | | - Elizabeth A Sellars
- University of Arkansas for Medical Sciences, Section of Genetics and Metabolism, 1 Children's Way, Little Rock, AR, USA, 72202
| | - Allyn McConkie-Rosell
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA, 27710
| | | | - Taosheng Huang
- Division of Human Genetics, Children's Hospital Medical Center, Cincinnati, OH, USA, 45229. .,Human Aging Research Institute, Nanchang University, Nanchang, China.
| |
Collapse
|
22
|
Vavla M, Arrigoni F, Nordio A, De Luca A, Pizzighello S, Petacchi E, Paparella G, D'Angelo MG, Brighina E, Russo E, Fantin M, Colombo P, Martinuzzi A. Functional and Structural Brain Damage in Friedreich's Ataxia. Front Neurol 2018; 9:747. [PMID: 30237783 PMCID: PMC6135889 DOI: 10.3389/fneur.2018.00747] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/17/2018] [Indexed: 11/13/2022] Open
Abstract
Friedreich's ataxia (FRDA) is a rare hereditary neurodegenerative disorder caused by a GAA repeat expansion in the FXN gene. There is still no cure or quantitative biomarkers reliaby correlating with the progression rate and disease severity. Investigation of functional and structural alterations characterizing white (WM) and gray matter (GM) in FRDA are needed prerequisite to monitor progression and response to treatment. Here we report the results of a multimodal cross-sectional MRI study of FRDA including Voxel-Based Morphometry (VBM), diffusion-tensor imaging (DTI), functional MRI (fMRI), and a correlation analysis with clinical severity scores. Twenty-one early-onset FRDA patients and 18 age-matched healthy controls (HCs) were imaged at 3T. All patients underwent a complete cognitive and clinical assessment with ataxia scales. VBM analysis showed GM volume reduction in FRDA compared to HCs bilaterally in lobules V, VI, VIII (L>R), as well as in the crus of cerebellum, posterior lobe of the vermis, in the flocculi and in the left tonsil. Voxel-wise DTI analysis showed a diffuse fractional anisotropy reduction and mean, radial, axial (AD) diffusivity increase in both infratentorial and supratentorial WM. ROI-based analysis confirmed the results showing differences of the same DTI metrics in cortico-spinal-tracts, forceps major, corpus callosum, posterior thalamic radiations, cerebellar penduncles. Additionally, we observed increased AD in superior (SCP) and middle cerebellar peduncles. The WM findings correlated with age at onset (AAO), short-allelle GAA, and disease severity. The intragroup analysis of fMRI data from right-handed 14 FRDA and 15 HCs showed similar findings in both groups, including activation in M1, insula and superior cerebellar hemisphere (lobules V-VIII). Significant differences emerged only during the non-dominant hand movement, with HCs showing a stronger activation in the left superior cerebellar hemisphere compared to FRDA. Significant correlations were found between AAO and the fMRI activation in cerebellar anterior and posterior lobes, insula and temporal lobe. Our multimodal neuroimaging protocol suggests that MRI is a useful tool to document the extension of the neurological impairment in FRDA.
Collapse
Affiliation(s)
- Marinela Vavla
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Filippo Arrigoni
- Neuroimaging Lab, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini, Italy
| | - Andrea Nordio
- Neuroimaging Lab, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini, Italy.,Department of Information Engineering, University of Padova, Padova, Italy
| | - Alberto De Luca
- Neuroimaging Lab, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini, Italy
| | - Silvia Pizzighello
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Elisa Petacchi
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Gabriella Paparella
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Maria Grazia D'Angelo
- NeuroMuscular Unit, Department of NeuroRehabilitation, IRCCS "Eugenio Medea", Bosisio Parini, Italy
| | - Erika Brighina
- NeuroMuscular Unit, Department of NeuroRehabilitation, IRCCS "Eugenio Medea", Bosisio Parini, Italy
| | - Emanuela Russo
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Marianna Fantin
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| | - Paola Colombo
- Neuroimaging Lab, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini, Italy
| | - Andrea Martinuzzi
- Severe Developmental Disabilities Unit, Scientific Institute, IRCCS "Eugenio Medea", Conegliano, Italy
| |
Collapse
|
23
|
Abstract
The dentate nucleus (DN) of the cerebellum is the major output nucleus of the cerebellum and is rich in iron. Quantitative susceptibility mapping (QSM) provides better iron-sensitive MRI contrast to delineate the boundary of the DN than either T2-weighted images or susceptibility-weighted images. Prior DN atlases used T2-weighted or susceptibility-weighted images to create DN atlases. Here, we employ QSM images to develop an improved dentate nucleus atlas for use in imaging studies. The DN was segmented in QSM images from 38 healthy volunteers. The resulting DN masks were transformed to a common space and averaged to generate the DN atlas. The center of mass of the left and right sides of the QSM-based DN atlas in the Montreal Neurological Institute space was -13.8, -55.8, and -36.4 mm, and 13.8, -55.7, and -36.4 mm, respectively. The maximal probability and mean probability of the DN atlas with the individually segmented DNs in this cohort were 100 and 39.3%, respectively, in contrast to the maximum probability of approximately 75% and the mean probability of 23.4 to 33.7% with earlier DN atlases. Using QSM, which provides superior iron-sensitive MRI contrast for delineating iron-rich structures, an improved atlas for the dentate nucleus has been generated. The atlas can be applied to investigate the role of the DN in both normal cortico-cerebellar physiology and the variety of disease states in which it is implicated.
Collapse
|
24
|
Napierala JS, Li Y, Lu Y, Lin K, Hauser LA, Lynch DR, Napierala M. Comprehensive analysis of gene expression patterns in Friedreich's ataxia fibroblasts by RNA sequencing reveals altered levels of protein synthesis factors and solute carriers. Dis Model Mech 2018; 10:1353-1369. [PMID: 29125828 PMCID: PMC5719256 DOI: 10.1242/dmm.030536] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/21/2017] [Indexed: 12/30/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease usually caused by large homozygous expansions of GAA repeat sequences in intron 1 of the frataxin (FXN) gene. FRDA patients homozygous for GAA expansions have low FXN mRNA and protein levels when compared with heterozygous carriers or healthy controls. Frataxin is a mitochondrial protein involved in iron–sulfur cluster synthesis, and many FRDA phenotypes result from deficiencies in cellular metabolism due to lowered expression of FXN. Presently, there is no effective treatment for FRDA, and biomarkers to measure therapeutic trial outcomes and/or to gauge disease progression are lacking. Peripheral tissues, including blood cells, buccal cells and skin fibroblasts, can readily be isolated from FRDA patients and used to define molecular hallmarks of disease pathogenesis. For instance, FXN mRNA and protein levels as well as FXN GAA-repeat tract lengths are routinely determined using all of these cell types. However, because these tissues are not directly involved in disease pathogenesis, their relevance as models of the molecular aspects of the disease is yet to be decided. Herein, we conducted unbiased RNA sequencing to profile the transcriptomes of fibroblast cell lines derived from 18 FRDA patients and 17 unaffected control individuals. Bioinformatic analyses revealed significantly upregulated expression of genes encoding plasma membrane solute carrier proteins in FRDA fibroblasts. Conversely, the expression of genes encoding accessory factors and enzymes involved in cytoplasmic and mitochondrial protein synthesis was consistently decreased in FRDA fibroblasts. Finally, comparison of genes differentially expressed in FRDA fibroblasts to three previously published gene expression signatures defined for FRDA blood cells showed substantial overlap between the independent datasets, including correspondingly deficient expression of antioxidant defense genes. Together, these results indicate that gene expression profiling of cells derived from peripheral tissues can, in fact, consistently reveal novel molecular pathways of the disease. When performed on statistically meaningful sample group sizes, unbiased global profiling analyses utilizing peripheral tissues are critical for the discovery and validation of FRDA disease biomarkers. Summary: Transcriptome profiling of Friedreich's ataxia fibroblasts by RNA sequencing reveals that this peripheral tissue can be used as a disease model for gene expression biomarker discovery.
Collapse
Affiliation(s)
- Jill Sergesketter Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA
| | - Yanjie Li
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA
| | - Yue Lu
- University of Texas MD Anderson Cancer Center, Department of Molecular Carcinogenesis, Center for Cancer Epigenetics, Science Park, Smithville, Texas 78957, USA
| | - Kevin Lin
- University of Texas MD Anderson Cancer Center, Department of Molecular Carcinogenesis, Center for Cancer Epigenetics, Science Park, Smithville, Texas 78957, USA
| | - Lauren A Hauser
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research Center Room 502, Philadelphia, PA 19104, USA
| | - David R Lynch
- Departments of Neurology and Pediatrics, Children's Hospital of Philadelphia, Abramson Research Center Room 502, Philadelphia, PA 19104, USA
| | - Marek Napierala
- University of Alabama at Birmingham, Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, 1825 University Blvd., Birmingham, Alabama 35294, USA .,Department of Molecular Biomedicine, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, 61-704, Poland
| |
Collapse
|
25
|
Keuken MC, Isaacs BR, Trampel R, van der Zwaag W, Forstmann BU. Visualizing the Human Subcortex Using Ultra-high Field Magnetic Resonance Imaging. Brain Topogr 2018; 31:513-545. [PMID: 29497874 PMCID: PMC5999196 DOI: 10.1007/s10548-018-0638-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/28/2018] [Indexed: 12/15/2022]
Abstract
With the recent increased availability of ultra-high field (UHF) magnetic resonance imaging (MRI), substantial progress has been made in visualizing the human brain, which can now be done in extraordinary detail. This review provides an extensive overview of the use of UHF MRI in visualizing the human subcortex for both healthy and patient populations. The high inter-subject variability in size and location of subcortical structures limits the usability of atlases in the midbrain. Fortunately, the combined results of this review indicate that a large number of subcortical areas can be visualized in individual space using UHF MRI. Current limitations and potential solutions of UHF MRI for visualizing the subcortex are also discussed.
Collapse
Affiliation(s)
- M C Keuken
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands.
- Cognitive Psychology Unit, Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands.
| | - B R Isaacs
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - R Trampel
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - B U Forstmann
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Cocozza S, Costabile T, Tedeschi E, Abate F, Russo C, Liguori A, Del Vecchio W, Paciello F, Quarantelli M, Filla A, Brunetti A, Saccà F. Cognitive and functional connectivity alterations in Friedreich's ataxia. Ann Clin Transl Neurol 2018; 5:677-686. [PMID: 29928651 PMCID: PMC5989773 DOI: 10.1002/acn3.555] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 01/26/2023] Open
Abstract
Objective The aim of this study was to perform the first resting-state functional MRI (RS-fMRI) analysis in Friedreich's ataxia (FRDA) patients to assess possible brain functional connectivity (FC) differences in these patients, and test their correlations with neuropsychological performances. Methods In total, 24 FRDA patients (M/F: 15/9, mean age 31.3 ± 15.0) and 24 healthy controls (HC; M/F: 15/9, mean age 30.7 ± 15.5) were enrolled in this cross-sectional study. All patients underwent a thorough neuropsychological battery, investigating different cognitive domains. RS-fMRI data were analyzed using a seed-based approach, probing the FC of cortical areas potentially referable to specific executive and cognitive functions compromised in FRDA. Results Compared to HC, FRDA patients showed overall worse neuropsychological scores in several domains, including global cognitive assessment, spatial memory, visuoperception and visuospatial functions, and executive functions. Analysis of RS-fMRI data showed a higher FC in FRDA patients compared to HC between paracingulate gyri and the medial frontal gryrus, between the superior frontal gyrus and bilateral angular gyri, and between the middle temporal gyrus and the cingulate gyrus, with a reduced FC between the medial frontal gryrus and the cerebellum. Interpretation We found a reduction in FC between frontal areas and the contralateral cerebellar cortex in FRDA, in line with the known alteration in cerebello-cortical pathway in this condition. On the other hand, a higher FC between different cortical areas was shown, possibly reflecting a compensatory phenomenon. These results, in conjunction with clinical findings, may shed new light on the pattern of supratentorial and infratentorial involvement, and on dynamics of brain plasticity in this disease.
Collapse
Affiliation(s)
- Sirio Cocozza
- Department of Advanced Biomedical Sciences University "Federico II" Naples Italy
| | - Teresa Costabile
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| | - Enrico Tedeschi
- Department of Advanced Biomedical Sciences University "Federico II" Naples Italy
| | - Filomena Abate
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| | - Camilla Russo
- Department of Advanced Biomedical Sciences University "Federico II" Naples Italy
| | - Agnese Liguori
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| | - Walter Del Vecchio
- Institute of Biostructure and Bioimaging National Research Council Naples Italy
| | - Francesca Paciello
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| | - Mario Quarantelli
- Institute of Biostructure and Bioimaging National Research Council Naples Italy
| | - Alessandro Filla
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences University "Federico II" Naples Italy
| | - Francesco Saccà
- Department of Neurosciences and Reproductive and Odontostomatological Sciences University "Federico II" Naples Italy
| |
Collapse
|
27
|
Manto M, Hampe CS. Endocrine disorders and the cerebellum: from neurodevelopmental injury to late-onset ataxia. HANDBOOK OF CLINICAL NEUROLOGY 2018; 155:353-368. [PMID: 29891071 DOI: 10.1016/b978-0-444-64189-2.00023-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Hormonal disorders are a source of cerebellar ataxia in both children and adults. Normal development of the cerebellum is critically dependent on thyroid hormone, which crosses both the blood-brain barrier and the blood-cerebrospinal fluid barrier thanks to specific transporters, including monocarboxylate transporter 8 and the organic anion-transporting polypeptide 1C1. In particular, growth and dendritic arborization of Purkinje neurons, synaptogenesis, and myelination are dependent on thyroid hormone. Disturbances of thyroid hormone may also impact on cerebellar ataxias of other origin, decompensating or aggravating the pre-existing ataxia manifesting with motor ataxia, oculomotor ataxia, and/or Schmahmann syndrome. Parathyroid disorders are associated with a genuine cerebellar syndrome, but symptoms may be subtle. The main conditions combining diabetes and cerebellar ataxia are Friedreich ataxia, ataxia associated with anti-GAD antibodies, autoimmune polyglandular syndromes, aceruloplasminemia, and cerebellar ataxia associated with hypogonadism (especially Holmes ataxia/Boucher-Neuhäuser syndrome). The general workup of cerebellar disorders should include the evaluation of hormonal status, including thyroid-stimulating hormone and free thyroxine levels, and hormonal replacement should be considered depending on the laboratory results. Cerebellar deficits may be reversible in some cases.
Collapse
Affiliation(s)
- Mario Manto
- Neurology Service, CHU-Charleroi, Charleroi, Belgium; Neuroscience Service, Université de Mons, Mons, Belgium.
| | - Christiane S Hampe
- Department of Medicine, University of Washington, Seattle, United States
| |
Collapse
|
28
|
Selvadurai LP, Harding IH, Corben LA, Georgiou-Karistianis N. Cerebral abnormalities in Friedreich ataxia: A review. Neurosci Biobehav Rev 2018; 84:394-406. [DOI: 10.1016/j.neubiorev.2017.08.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 06/06/2017] [Accepted: 08/10/2017] [Indexed: 12/31/2022]
|
29
|
Rocca CJ, Goodman SM, Dulin JN, Haquang JH, Gertsman I, Blondelle J, Smith JLM, Heyser CJ, Cherqui S. Transplantation of wild-type mouse hematopoietic stem and progenitor cells ameliorates deficits in a mouse model of Friedreich's ataxia. Sci Transl Med 2017; 9:eaaj2347. [PMID: 29070698 PMCID: PMC5735830 DOI: 10.1126/scitranslmed.aaj2347] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/31/2017] [Accepted: 06/13/2017] [Indexed: 12/14/2022]
Abstract
Friedreich's ataxia (FRDA) is an incurable autosomal recessive neurodegenerative disease caused by reduced expression of the mitochondrial protein frataxin due to an intronic GAA-repeat expansion in the FXN gene. We report the therapeutic efficacy of transplanting wild-type mouse hematopoietic stem and progenitor cells (HSPCs) into the YG8R mouse model of FRDA. In the HSPC-transplanted YG8R mice, development of muscle weakness and locomotor deficits was abrogated as was degeneration of large sensory neurons in the dorsal root ganglia (DRGs) and mitochondrial capacity was improved in brain, skeletal muscle, and heart. Transplanted HSPCs engrafted and then differentiated into microglia in the brain and spinal cord and into macrophages in the DRGs, heart, and muscle of YG8R FRDA mice. We observed the transfer of wild-type frataxin and Cox8 mitochondrial proteins from HSPC-derived microglia/macrophages to FRDA mouse neurons and muscle myocytes in vivo. Our results show the HSPC-mediated phenotypic rescue of FRDA in YG8R mice and suggest that this approach should be investigated further as a strategy for treating FRDA.
Collapse
Affiliation(s)
- Celine J Rocca
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Spencer M Goodman
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jennifer N Dulin
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph H Haquang
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ilya Gertsman
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jordan Blondelle
- Division of Cardiovascular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Janell L M Smith
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Charles J Heyser
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie Cherqui
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
30
|
Deistung A, Schweser F, Reichenbach JR. Overview of quantitative susceptibility mapping. NMR IN BIOMEDICINE 2017; 30:e3569. [PMID: 27434134 DOI: 10.1002/nbm.3569] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 06/06/2023]
Abstract
Magnetic susceptibility describes the magnetizability of a material to an applied magnetic field and represents an important parameter in the field of MRI. With the recently introduced method of quantitative susceptibility mapping (QSM) and its conceptual extension to susceptibility tensor imaging (STI), the non-invasive assessment of this important physical quantity has become possible with MRI. Both methods solve the ill-posed inverse problem to determine the magnetic susceptibility from local magnetic fields. Whilst QSM allows the extraction of the spatial distribution of the bulk magnetic susceptibility from a single measurement, STI enables the quantification of magnetic susceptibility anisotropy, but requires multiple measurements with different orientations of the object relative to the main static magnetic field. In this review, we briefly recapitulate the fundamental theoretical foundation of QSM and STI, as well as computational strategies for the characterization of magnetic susceptibility with MRI phase data. In the second part, we provide an overview of current methodological and clinical applications of QSM with a focus on brain imaging. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Andreas Deistung
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
| | - Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York at Buffalo, NY, USA
- MRI Clinical and Translational Research Center, Jacobs School of Medicine and Biomedical Sciences, The State University of New York at Buffalo, NY, USA
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Jena, Germany
- Michael Stifel Center for Data-driven and Simulation Science Jena, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
31
|
The Pediatric Cerebellum in Inherited Neurodegenerative Disorders: A Pattern-recognition Approach. Neuroimaging Clin N Am 2017; 26:373-416. [PMID: 27423800 DOI: 10.1016/j.nic.2016.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Evaluation of imaging studies of the cerebellum in inherited neurodegenerative disorders is aided by attention to neuroimaging patterns based on anatomic determinants, including biometric analysis, hyperintense signal of structures, including the cerebellar cortex, white matter, dentate nuclei, brainstem tracts, and nuclei, the presence of cysts, brain iron, or calcifications, change over time, the use of diffusion-weighted/diffusion tensor imaging and T2*-weighted sequences, magnetic resonance spectroscopy; and, in rare occurrences, the administration of contrast material.
Collapse
|
32
|
Dusek P, Schneider SA, Aaseth J. Iron chelation in the treatment of neurodegenerative diseases. J Trace Elem Med Biol 2016; 38:81-92. [PMID: 27033472 DOI: 10.1016/j.jtemb.2016.03.010] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 01/14/2023]
Abstract
Disturbance of cerebral iron regulation is almost universal in neurodegenerative disorders. There is a growing body of evidence that increased iron deposits may contribute to degenerative changes. Thus, the effect of iron chelation therapy has been investigated in many neurological disorders including rare genetic syndromes with neurodegeneration with brain iron accumulation as well as common sporadic disorders such as Parkinson's disease, Alzheimer's disease, and multiple sclerosis. This review summarizes recent advances in understanding the role of iron in the etiology of neurodegeneration. Outcomes of studies investigating the effect of iron chelation therapy in neurodegenerative disorders are systematically presented in tables. Iron chelators, particularly the blood brain barrier-crossing compound deferiprone, are capable of decreasing cerebral iron in areas with abnormally high concentrations as documented by MRI. Yet, currently, there is no compelling evidence of the clinical effect of iron removal therapy on any neurological disorder. However, several studies indicate that it may prevent or slow down disease progression of several disorders such as aceruloplasminemia, pantothenate kinase-associated neurodegeneration or Parkinson's disease.
Collapse
Affiliation(s)
- Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital in Prague, Czech Republic; Institute of Neuroradiology, University Göttingen, Göttingen, Germany.
| | | | - Jan Aaseth
- Innlandet Hospital Trust, Kongsvinger, Norway; Hedmark University College, Elverum, Norway
| |
Collapse
|
33
|
Chen K, Ho TSY, Lin G, Tan KL, Rasband MN, Bellen HJ. Loss of Frataxin activates the iron/sphingolipid/PDK1/Mef2 pathway in mammals. eLife 2016; 5:e20732. [PMID: 27901468 PMCID: PMC5130293 DOI: 10.7554/elife.20732] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/17/2016] [Indexed: 01/05/2023] Open
Abstract
Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused by mutations in Frataxin (FXN). Loss of FXN causes impaired mitochondrial function and iron homeostasis. An elevated production of reactive oxygen species (ROS) was previously proposed to contribute to the pathogenesis of FRDA. We recently showed that loss of frataxin homolog (fh), a Drosophila homolog of FXN, causes a ROS independent neurodegeneration in flies (Chen et al., 2016). In fh mutants, iron accumulation in the nervous system enhances the synthesis of sphingolipids, which in turn activates 3-phosphoinositide dependent protein kinase-1 (Pdk1) and myocyte enhancer factor-2 (Mef2) to trigger neurodegeneration of adult photoreceptors. Here, we show that loss of Fxn in the nervous system in mice also activates an iron/sphingolipid/PDK1/Mef2 pathway, indicating that the mechanism is evolutionarily conserved. Furthermore, sphingolipid levels and PDK1 activity are also increased in hearts of FRDA patients, suggesting that a similar pathway is affected in FRDA.
Collapse
Affiliation(s)
- Kuchuan Chen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Tammy Szu-Yu Ho
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Kai Li Tan
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Matthew N Rasband
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute, Houston, United States
| |
Collapse
|
34
|
Harding IH, Raniga P, Delatycki MB, Stagnitti MR, Corben LA, Storey E, Georgiou-Karistianis N, Egan GF. Tissue atrophy and elevated iron concentration in the extrapyramidal motor system in Friedreich ataxia: the IMAGE-FRDA study. J Neurol Neurosurg Psychiatry 2016; 87:1261-1263. [PMID: 27010617 DOI: 10.1136/jnnp-2015-312665] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 03/09/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Ian H Harding
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Parnesh Raniga
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| | - Martin B Delatycki
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Melbourne, Victoria, Australia Bruce Lefroy Centre, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia Department of Clinical Genetics, Austin Health, Melbourne, Victoria, Australia
| | - Monique R Stagnitti
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Louise A Corben
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Melbourne, Victoria, Australia Bruce Lefroy Centre, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia Monash Medical Centre, Monash Health, Melbourne, Victoria, Australia
| | - Elsdon Storey
- Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences & Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Stroh MA, Winter MK, Swerdlow RH, McCarson KE, Zhu H. Loss of NCB5OR in the cerebellum disturbs iron pathways, potentiates behavioral abnormalities, and exacerbates harmaline-induced tremor in mice. Metab Brain Dis 2016; 31:951-64. [PMID: 27188291 PMCID: PMC5929129 DOI: 10.1007/s11011-016-9834-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/01/2016] [Indexed: 12/13/2022]
Abstract
Iron dyshomeostasis has been implicated in many diseases, including a number of neurological conditions. Cytosolic NADH cytochrome b5 oxidoreductase (NCB5OR) is ubiquitously expressed in animal tissues and is capable of reducing ferric iron in vitro. We previously reported that global gene ablation of NCB5OR resulted in early-onset diabetes and altered iron homeostasis in mice. To further investigate the specific effects of NCB5OR deficiency on neural tissue without contributions from known phenotypes, we generated a conditional knockout (CKO) mouse that lacks NCB5OR only in the cerebellum and midbrain. Assessment of molecular markers in the cerebellum of CKO mice revealed changes in pathways associated with cellular and mitochondrial iron homeostasis. (59)Fe pulse-feeding experiments revealed cerebellum-specific increased or decreased uptake of iron by 7 and 16 weeks of age, respectively. Additionally, we characterized behavioral changes associated with loss of NCB5OR in the cerebellum and midbrain in the context of dietary iron deprivation-evoked generalized iron deficiency. Locomotor activity was reduced and complex motor task execution was altered in CKO mice treated with an iron deficient diet. A sucrose preference test revealed that the reward response was intact in CKO mice, but that iron deficient diet consumption altered sucrose preference in all mice. Detailed gait analysis revealed locomotor changes in CKO mice associated with dysfunctional proprioception and locomotor activation independent of dietary iron deficiency. Finally, we demonstrate that loss of NCB5OR in the cerebellum and midbrain exacerbated harmaline-induced tremor activity. Our findings suggest an essential role for NCB5OR in maintaining both iron homeostasis and the proper functioning of various locomotor pathways in the mouse cerebellum and midbrain.
Collapse
Affiliation(s)
- Matthew A Stroh
- Landon Center on Aging, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 1005, Kansas City, KS, 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA
| | - Michelle K Winter
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3051, Kansas City, KS, 66160, USA
| | - Russell H Swerdlow
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA
- Department of Neurology, University of Kansas Medical Center, 3599 Rainbow Blvd., MSN 2012, Kansas City, KS, 66160, USA
| | - Kenneth E McCarson
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3051, Kansas City, KS, 66160, USA
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 1018, Kansas City, KS, 66160, USA
| | - Hao Zhu
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3030, Kansas City, KS, 66160, USA.
- Neuroscience Graduate Program, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 3038, Kansas City, KS, 66160, USA.
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, 3901 Rainbow Blvd., MSN 4048G-Eaton, Kansas City, KS, 66160, USA.
| |
Collapse
|
36
|
Chen K, Lin G, Haelterman NA, Ho TSY, Li T, Li Z, Duraine L, Graham BH, Jaiswal M, Yamamoto S, Rasband MN, Bellen HJ. Loss of Frataxin induces iron toxicity, sphingolipid synthesis, and Pdk1/Mef2 activation, leading to neurodegeneration. eLife 2016; 5:e16043. [PMID: 27343351 PMCID: PMC4956409 DOI: 10.7554/elife.16043] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/24/2016] [Indexed: 12/22/2022] Open
Abstract
Mutations in Frataxin (FXN) cause Friedreich's ataxia (FRDA), a recessive neurodegenerative disorder. Previous studies have proposed that loss of FXN causes mitochondrial dysfunction, which triggers elevated reactive oxygen species (ROS) and leads to the demise of neurons. Here we describe a ROS independent mechanism that contributes to neurodegeneration in fly FXN mutants. We show that loss of frataxin homolog (fh) in Drosophila leads to iron toxicity, which in turn induces sphingolipid synthesis and ectopically activates 3-phosphoinositide dependent protein kinase-1 (Pdk1) and myocyte enhancer factor-2 (Mef2). Dampening iron toxicity, inhibiting sphingolipid synthesis by Myriocin, or reducing Pdk1 or Mef2 levels, all effectively suppress neurodegeneration in fh mutants. Moreover, increasing dihydrosphingosine activates Mef2 activity through PDK1 in mammalian neuronal cell line suggesting that the mechanisms are evolutionarily conserved. Our results indicate that an iron/sphingolipid/Pdk1/Mef2 pathway may play a role in FRDA.
Collapse
Affiliation(s)
- Kuchuan Chen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Nele A Haelterman
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Tammy Szu-Yu Ho
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Tongchao Li
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Zhihong Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Lita Duraine
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Brett H Graham
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Manish Jaiswal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Shinya Yamamoto
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, United States
| | - Matthew N Rasband
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, United States
| |
Collapse
|
37
|
Kemp KC, Cook AJ, Redondo J, Kurian KM, Scolding NJ, Wilkins A. Purkinje cell injury, structural plasticity and fusion in patients with Friedreich's ataxia. Acta Neuropathol Commun 2016; 4:53. [PMID: 27215193 PMCID: PMC4877974 DOI: 10.1186/s40478-016-0326-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/11/2016] [Indexed: 12/05/2022] Open
Abstract
Purkinje cell pathology is a common finding in a range of inherited and acquired cerebellar disorders, with the degree of Purkinje cell injury dependent on the underlying aetiology. Purkinje cells have an unparalleled resistance to insult and display unique regenerative capabilities within the central nervous system. Their response to cell injury is not typical of most neurons and likely represents both degenerative, compensatory and regenerative mechanisms. Here we present a pathological study showing novel and fundamental insights into Purkinje cell injury, remodelling and repair in Friedreich’s ataxia; the most common inherited ataxia. Analysing post-mortem cerebellum tissue from patients who had Friedreich's ataxia, we provide evidence of significant injury to the Purkinje cell axonal compartment with relative preservation of both the perikaryon and its extensive dendritic arborisation. Axonal remodelling of Purkinje cells was clearly elevated in the disease. For the first time in a genetic condition, we have also shown a disease-related increase in the frequency of Purkinje cell fusion and heterokaryon formation in Friedreich's ataxia cases; with evidence that underlying levels of cerebellar inflammation influence heterokaryon formation. Our results together further demonstrate the Purkinje cell’s unique plasticity and regenerative potential. Elucidating the biological mechanisms behind these phenomena could have significant clinical implications for manipulating neuronal repair in response to neurological injury.
Collapse
|
38
|
Paap BK, Roeske S, Durr A, Schöls L, Ashizawa T, Boesch S, Bunn LM, Delatycki MB, Giunti P, Lehéricy S, Mariotti C, Melegh J, Pandolfo M, Tallaksen CM, Timmann D, Tsuji S, Schulz JB, van de Warrenburg BP, Klockgether T. Standardized Assessment of Hereditary Ataxia Patients in Clinical Studies. Mov Disord Clin Pract 2016; 3:230-240. [PMID: 30363623 PMCID: PMC6178745 DOI: 10.1002/mdc3.12315] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/12/2015] [Accepted: 11/02/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hereditary ataxias are a heterogeneous group of degenerative diseases of the cerebellum, brainstem, and spinal cord. They may present with isolated ataxia or with additional symptoms going beyond cerebellar deficits. There are an increasing number of clinical studies with the goal to define the natural history of these disorders, develop biomarkers, and investigate therapeutic interventions. Especially, early and preclinical disease stages are currently of particular interest. METHODS AND RESULTS Evidence-based, we review standards for sampling and storage of biomaterials, clinical and neuropsychological assessment, as well as neurophysiology and neuroimaging and recommendations for standardized assessment of ataxia patients in multicenter studies. CONCLUSIONS DNA, RNA, serum, and, if possible, cerebrospinal fluid samples should be processed following established standards. Clinical assessment in ataxia studies must include use of a validated clinical ataxia scale. There are several validated clinical ataxia scales available. There are no instruments that were specifically designed for assessing neuropsychological and psychiatric symptoms in ataxia disorders. We provide a list of tests that may prove valuable. Quantitative performance tests have the potential to supplement clinical scales. They provide additional objective and quantitative information. Posturography and quantitative movement analysis-despite valid approaches-require standardization before implemented in multicenter studies. Standardization of neurophysiological tools, as required for multicenter interventional trials, is still lacking. Future multicenter neuroimaging studies in ataxias should implement quality assurance measures as defined by the ADNI or other consortia. MRI protocols should allow morphometric analyses.
Collapse
Affiliation(s)
| | - Sandra Roeske
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Alexandra Durr
- APHP Department of GeneticsGroupe Hospitalier Pitié‐SalpêtrièreParisFrance
- Institut du Cerveau et de la MoelleINSERM U1127CNRS UMR7225Sorbonne Universités–UPMC Université Paris VI UMR_S1127ParisFrance
| | - Ludger Schöls
- Department of Neurology and Hertie‐Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Tetsuo Ashizawa
- Department of Neurology and Neuroscience Research ProgramMethodist Hospital Research InstituteHoustonTexasUSA
| | - Sylvia Boesch
- Department of NeurologyMedical University InnsbruckInnsbruckAustria
| | - Lisa M. Bunn
- School of Health ProfessionsPeninsula Allied Health CenterUniversity of PlymouthUnited Kingdom
| | - Martin B. Delatycki
- Murdoch Children's Research InstituteMelbourneAustralia
- Clinical GeneticsAustin HealthHeidelbergVictoriaAustralia
| | - Paola Giunti
- Ataxia Centre Department of MolecularneuroscienceUCL Institute of NeurologyLondonUnited Kingdom
| | - Stéphane Lehéricy
- Institut du Cerveau et de la MoelleINSERM U1127CNRS UMR7225Sorbonne Universités–UPMC Université Paris VI UMR_S1127ParisFrance
- Institut du Cerveau et de la Moelle (ICM) epiniereCentre de NeuroImagerie de Recherche (CENIR)ParisFrance
- Service de NeuroradiologieGroupe Hospitalier Pitie‐SalpetriereParisFrance
| | - Caterina Mariotti
- Unit of Genetics of Neurodegenerative and Metabolic DisordersFondazione IRCCS‐Istituto Neurologico Carlo BestaMilanItaly
| | - Jörg Melegh
- Department of Medical Genetics and Szentagothai Research CenterUniversity of PécsPécsHungary
| | | | - Chantal M.E. Tallaksen
- Department of NeurologyOslo University HospitalOsloNorway
- Faculty of MedicineOslo UniversityOsloNorway
| | - Dagmar Timmann
- Department of NeurologyEssen University HospitalUniversity of Duisburg‐EssenEssenGermany
| | - Shoji Tsuji
- Department of NeurologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Jörg Bela Schulz
- Department of Neurology and JARA BrainUniversity HospitalRWTH AachenAachenGermany
| | - Bart P. van de Warrenburg
- Department of NeurologyRadboud University Medical CenterDonders Institute for Brain, Cognition, and BehaviorNijmegenThe Netherlands
| | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of NeurologyUniversity Hospital of BonnBonnGermany
| |
Collapse
|
39
|
Gramegna LL, Tonon C, Manners DN, Pini A, Rinaldi R, Zanigni S, Bianchini C, Evangelisti S, Fortuna F, Carelli V, Testa C, Lodi R. Combined Cerebellar Proton MR Spectroscopy and DWI Study of Patients with Friedreich’s Ataxia. THE CEREBELLUM 2016; 16:82-88. [DOI: 10.1007/s12311-016-0767-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
40
|
Structural and Functional Magnetic Resonance Imaging of the Cerebellum: Considerations for Assessing Cerebellar Ataxias. THE CEREBELLUM 2015; 15:21-25. [DOI: 10.1007/s12311-015-0738-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
41
|
Liddell JR. Targeting mitochondrial metal dyshomeostasis for the treatment of neurodegeneration. Neurodegener Dis Manag 2015; 5:345-64. [DOI: 10.2217/nmt.15.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial impairment and metal dyshomeostasis are suggested to be associated with many neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and Friedreich's ataxia. Treatments aimed at restoring metal homeostasis are highly effective in models of these diseases, and clinical trials hold promise. However, in general, the effect of these treatments on mitochondrial metal homeostasis is unclear, and the contribution of mitochondrial metal dyshomeostasis to disease pathogenesis requires further investigation. This review describes the role of metals in mitochondria in health, how mitochondrial metals are disrupted in neurodegenerative diseases, and potential therapeutics aimed at restoring mitochondrial metal homeostasis and function.
Collapse
Affiliation(s)
- Jeffrey R Liddell
- Department of Pathology, University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW This article discusses recent advances in the understanding of clinical and genetic aspects of primary ataxias, including congenital, autosomal recessive, autosomal dominant, episodic, X-linked, and mitochondrial ataxias, as well as idiopathic degenerative and secondary ataxias. RECENT FINDINGS Many important observations have been published in recent years in connection with primary ataxias, particularly new loci and genes. The most commonly inherited ataxias may present with typical and atypical phenotypes. In the group of idiopathic degenerative ataxias, genes have been found in patients with multiple system atrophy type C. Secondary ataxias represent an important group of sporadic, cerebellar, and afferent/sensory ataxias. SUMMARY Knowledge of primary ataxias has been growing rapidly in recent years. Here we review different forms of primary ataxia, including inherited forms, which are subdivided into congenital, autosomal recessive cerebellar ataxias, autosomal dominant cerebellar ataxias, episodic ataxias, X-linked ataxias, and mitochondrial ataxias, as well as sporadic ataxias and idiopathic degenerative ataxias. Secondary or acquired ataxias are also reviewed and the most common causes are discussed.
Collapse
Affiliation(s)
- Hélio A.G. Teive
- Department of Internal Medicine, Movement Disorders Unit and Neurology Service, Hospital de Clínicas, Federal University of Paraná, Curitiba, Paraná, Brazil and
| | - Tetsuo Ashizawa
- Department of Neurology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
43
|
Abrahão A, Pedroso JL, Braga-Neto P, Bor-Seng-Shu E, de Carvalho Aguiar P, Barsottini OGP. Milestones in Friedreich ataxia: more than a century and still learning. Neurogenetics 2015; 16:151-60. [PMID: 25662948 DOI: 10.1007/s10048-015-0439-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/20/2015] [Indexed: 10/24/2022]
Abstract
Friedreich ataxia (FRDA) is the most common autosomal recessive ataxia worldwide. This review highlights the main clinical features, pathophysiological mechanisms, and therapeutic approaches for FRDA patients. The disease is characterized by a combination of neurological involvement (ataxia and neuropathy), cardiomyopathy, skeletal abnormalities, and glucose metabolism disturbances. FRDA is caused by expanded guanine-adenine-adenine (GAA) triplet repeats in the first intron of the frataxin gene (FXN), resulting in reduction of messenger RNA and protein levels of frataxin in different tissues. The molecular and metabolic disturbances, including iron accumulation, lead to pathological changes characterized by spinal cord and dorsal root ganglia atrophy, dentate nucleus atrophy without global cerebellar volume reduction, and hypertrophic cardiomyopathy. DNA analysis is the hallmark for the diagnosis of FRDA. There is no specific treatment to stop the disease progression in FRDA patients. However, a number of drugs are under investigation. Therapeutic approaches intend to improve mitochondrial functioning and to increase FXN expression.
Collapse
Affiliation(s)
- Agessandro Abrahão
- Division of General Neurology and Ataxia Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, Rua Pedro de Toledo 650 Vila Clementino, São Paulo, 04039-002, SP, Brazil,
| | | | | | | | | | | |
Collapse
|
44
|
Bareš M, Apps R, Kikinis Z, Timmann D, Oz G, Ashe JJ, Loft M, Koutsikou S, Cerminara N, Bushara KO, Kašpárek T. Proceedings of the workshop on Cerebellum, Basal Ganglia and Cortical Connections Unmasked in Health and Disorder held in Brno, Czech Republic, October 17th, 2013. CEREBELLUM (LONDON, ENGLAND) 2015; 14:142-50. [PMID: 25205331 PMCID: PMC5035040 DOI: 10.1007/s12311-014-0595-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The proceedings of the workshop synthesize the experimental, preclinical, and clinical data suggesting that the cerebellum, basal ganglia (BG), and their connections play an important role in pathophysiology of various movement disorders (like Parkinson's disease and atypical parkinsonian syndromes) or neurodevelopmental disorders (like autism). The contributions from individual distinguished speakers cover the neuroanatomical research of complex networks, neuroimaging data showing that the cerebellum and BG are connected to a wide range of other central nervous system structures involved in movement control. Especially, the cerebellum plays a more complex role in how the brain functions than previously thought.
Collapse
Affiliation(s)
- Martin Bareš
- Central European Institute of Technology, CEITEC MU, Behavioral and Social Neuroscience Research Group, Masaryk University, Brno, Czech Republic,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Stefanescu MR, Dohnalek M, Maderwald S, Thürling M, Minnerop M, Beck A, Schlamann M, Diedrichsen J, Ladd ME, Timmann D. Structural and functional MRI abnormalities of cerebellar cortex and nuclei in SCA3, SCA6 and Friedreich's ataxia. Brain 2015; 138:1182-97. [PMID: 25818870 DOI: 10.1093/brain/awv064] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
Spinocerebellar ataxia type 3, spinocerebellar ataxia type 6 and Friedreich's ataxia are common hereditary ataxias. Different patterns of atrophy of the cerebellar cortex are well known. Data on cerebellar nuclei are sparse. Whereas cerebellar nuclei have long been thought to be preserved in spinocerebellar ataxia type 6, histology shows marked atrophy of the nuclei in Friedreich's ataxia and spinocerebellar ataxia type 3. In the present study susceptibility weighted imaging was used to assess atrophy of the cerebellar nuclei in patients with spinocerebellar ataxia type 6 (n = 12, age range 41-76 years, five female), Friedreich's ataxia (n = 12, age range 21-55 years, seven female), spinocerebellar ataxia type 3 (n = 10, age range 34-67 years, three female), and age- and gender-matched controls (total n = 23, age range 22-75 years, 10 female). T1-weighted magnetic resonance images were used to calculate the volume of the cerebellum. In addition, ultra-high field functional magnetic resonance imaging was performed with optimized normalization methods to assess function of the cerebellar cortex and nuclei during simple hand movements. As expected, the volume of the cerebellum was markedly reduced in spinocerebellar ataxia type 6, preserved in Friedreich's ataxia, and mildy reduced in spinocerebellar ataxia type 3. The volume of the cerebellar nuclei was reduced in the three patient groups compared to matched controls (P-values < 0.05; two-sample t-tests). Atrophy of the cerebellar nuclei was most pronounced in spinocerebellar ataxia type 6. On a functional level, hand-movement-related cerebellar activation was altered in all three disorders. Within the cerebellar cortex, functional magnetic resonance imaging signal was significantly reduced in spinocerebellar ataxia type 6 and Friedreich's ataxia compared to matched controls (P-values < 0.001, bootstrap-corrected cluster-size threshold; two-sample t-tests). The difference missed significance in spinocerebellar ataxia type 3. Within the cerebellar nuclei, reductions were significant when comparing spinocerebellar ataxia type 6 and Friedreich's ataxia to matched controls (P < 0.01, bootstrap-corrected cluster-size threshold; two-sample t-tests). Susceptibility weighted imaging allowed depiction of atrophy of the cerebellar nuclei in patients with Friedreich's ataxia and spinocerebellar ataxia type 3. In spinocerebellar ataxia type 6, pathology was not restricted to the cerebellar cortex but also involved the cerebellar nuclei. Functional magnetic resonance imaging data, on the other hand, revealed that pathology in Friedreich's ataxia and spinocerebellar ataxia type 3 is not restricted to the cerebellar nuclei. There was functional involvement of the cerebellar cortex despite no or little structural changes.
Collapse
Affiliation(s)
- Maria R Stefanescu
- 1 Department of Neurology, University of Duisburg-Essen, Essen, Germany 2 Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany
| | - Moritz Dohnalek
- 1 Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - Stefan Maderwald
- 2 Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany
| | - Markus Thürling
- 1 Department of Neurology, University of Duisburg-Essen, Essen, Germany 2 Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany
| | - Martina Minnerop
- 3 Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany 4 Department of Neurology, University of Bonn, Bonn, Germany
| | - Andreas Beck
- 5 Department of Computer Sciences, University of Düsseldorf, Düsseldorf, Germany
| | - Marc Schlamann
- 6 Department of Diagnostic and Interventional Radiology and Neuroradiology, University of Duisburg-Essen, Essen, Germany
| | - Joern Diedrichsen
- 7 Institute of Cognitive Neuroscience, University College London, London, UK
| | - Mark E Ladd
- 2 Erwin L. Hahn Institute for Magnetic Resonance Imaging, University of Duisburg-Essen, Essen, Germany 6 Department of Diagnostic and Interventional Radiology and Neuroradiology, University of Duisburg-Essen, Essen, Germany 8 Division of Medical Physics in Radiology, University of Heidelberg and German Cancer Research Centre, Heidelberg, Germany
| | - Dagmar Timmann
- 1 Department of Neurology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
46
|
Koeppen AH, Ramirez L, Becker AB, Feustel PJ, Mazurkiewicz JE. Friedreich ataxia: failure of GABA-ergic and glycinergic synaptic transmission in the dentate nucleus. J Neuropathol Exp Neurol 2015; 74:166-76. [PMID: 25575136 PMCID: PMC4294979 DOI: 10.1097/nen.0000000000000160] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Atrophy of large neurons in the dentate nucleus (DN) is an important pathologic correlate of neurologic disability in patients with Friedreich ataxia (FA). Thinning of the DN was quantified in 29 autopsy cases of FA and 2 carriers by measuring the thickness of the gray matter ribbon on stains with anti-glutamic acid decarboxylase, the rate-limiting enzyme in the biosynthesis of γ-amino-butyric acid (GABA). The DN was thinner than normal in all cases of FA, and atrophy correlated inversely with disease duration but not with age at onset or length of the homozygous guanine-adenine-adenine trinucleotide expansions. In 13 of the FA cases, frozen DN tissue was available for assay of frataxin. Dentate nucleus atrophy was more severe when frataxin was very low. Immunohistochemical staining for glutamic acid decarboxylase revealed grumose reaction and preservation of small GABA-ergic neurons in the DN of FA patients. Residual small DN neurons and varicose axons also contained the glycine transporter 2, identifying them as glycinergic. Immunohistochemistry also confirmed severe loss of GABA-A and glycine receptors in the DN with comparable depletion of the receptor-anchoring protein gephyrin. Thus, loss of gephyrin and failure to position GABA-A and glycine receptors correctly may reduce trophic support of large DN neurons and contribute to their atrophy. By contrast, Purkinje cells may escape retrograde atrophy in FA by issuing new axonal sprouts to small surviving DN neurons where they form reparative grumose clusters.
Collapse
Affiliation(s)
- Arnulf H. Koeppen
- Research Service, VA Medical Center, Albany, New York
- Department of Neurology, Albany Medical College, Albany, New York
- Department of Pathology, Albany Medical College, Albany, New York
| | - Liane Ramirez
- Research Service, VA Medical Center, Albany, New York
| | | | - Paul J. Feustel
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York
| | | |
Collapse
|