Friedreich Ataxia

Back

Background

Friedreich ataxia (FA, FRDA, FRIEDREICH ATAXIA 1, OMIM# *229300) is an autosomal recessive ataxia resulting from a mutation of a gene locus on chromosome 9. The entity was first described in 1863 by Nikolaus Friedreich, a professor of medicine in Heidelberg, Germany.

FA was the earliest of the inherited ataxias to be distinguished from other locomotor ataxias and is the most common of the autosomal recessive ataxias. It accounts for at least 50% of cases of hereditary ataxias in most large series. Cardinal features include progressive limb and gait ataxia, dysarthria, loss of joint position and vibration senses, absent tendon reflexes in the legs, and extensor plantar responses.

 

Pathophysiology

The major pathophysiologic finding in Friedreich ataxia is a "dying back phenomena" of axons, beginning in the periphery with ultimate loss of neurons and a secondary gliosis. The primary sites of these changes are the spinal cord and spinal roots. This results in loss of large myelinated axons in peripheral nerves, which increases with age and disease duration. Unmyelinated fibers in sensory roots and peripheral sensory nerves are spared.

The posterior columns and corticospinal, ventral, and lateral spinocerebellar tracts all show demyelination and depletion of large myelinated nerve fibers to differing extents. This is accompanied by a fibrous gliosis that does not replace the bulk of the lost fibers. Overall, the spinal cord becomes thin and the anteroposterior (AP) and transverse diameters of the thoracic cord are reduced. The dorsal spinal ganglia show shrinkage and eventual disappearance of neurons associated with proliferation of capsular cells. The posterior column degeneration accounts for the loss of position and vibration senses and the sensory ataxia. The loss of large neurons in the sensory ganglia causes extinction of tendon reflexes.

Large neurons of the dorsal root ganglia, especially lumbosacral, and nerve cells in the Clarke column are reduced in number. The posterior roots become thin. The dentate nuclei exhibit mild to moderate neuronal loss and the middle and superior cerebellar peduncles are reduced in size. Patchy loss of Purkinje cells in the superior vermis of the cerebellum and of neurons in corresponding portions of the inferior olivary nuclei is typical. Mild degenerative changes occur in the pontine and medullary nuclei and optic tracts. The cerebellar ataxia is explained by loss of the lateral and ventral spinocerebellar tracts and involvement of the Clarke column, dentate nucleus, superior vermis, and dentatorubral pathways.

The corticospinal tracts are relatively spared down to the level of the cervicomedullary junction. Beyond this point, the corticospinal tracts are severely degenerated, which becomes progressively more severe moving down the spinal cord. This explains the common finding of bilateral extensor plantar responses and weakness late in the disease.

Loss of cells in the nuclei of cranial nerves VII, X, and XII results in facial weakness and speech and swallowing difficulties.

Myocardial muscle fibers also show degeneration and are replaced by macrophages and fibroblasts. Essentially, chronic interstitial myocarditis occurs with hypertrophy of cardiac muscle fibers; fibers become hypertrophied and lose their striations. This is followed by swelling and vacuolation and finally interstitial fibrosis. The nuclei appear hyperchromatic and occasionally vacuolated. The cytoplasm appears granular with frequent lipofuscin depositions.

Kyphoscoliosis is likely; it is secondary to spinal muscular imbalance.

Epidemiology

Frequency

Friedreich ataxia is a relatively common disorder. It is the most common autosomal recessive ataxia, accounting for approximately 50% of all cases of hereditary ataxia. Estimates of incidence range anywhere from 1 in 22,000 to 2 in 100,000, with most studies yielding an incidence among Europeans and North Americans of European descent of approximately 1.5 per 100,000 per year; a slightly higher incidence has been reported in Quebec.

Overall, the FA carrier rate has been estimated recently to be 1 in 60 to 1 in 90 with a disease prevalence of 1 per 29,000.

Mortality/Morbidity

Friedreich ataxia is a progressive disorder with significant morbidity. Loss of ambulation typically occurs 15 years after disease onset. More than 95% of patients are wheelchair bound by age 45 years.

In a series by Harding et al, the average age of death was 37.7 years±14.4 years (range, 21-69).[1]

Race

Friedreich ataxia is most prevalent in white populations. Most FA carriers and affected FA patients are believed to originate from a common European ancestor who lived more than 10,000 years ago. Frataxin gene expansions are therefore almost nonexistent among black African and Asian populations.

Age

The onset of Friedreich ataxia is early; it typically presents in children aged 8-15 years and almost always presents before age 20 years.

In a study by Harding et al, the mean onset of symptoms occurred at age 10.52±7.4 years (range, 1.5-27).[1]

History

Onset of Friedreich ataxia (FA) is early, with gait ataxia being the usual presenting symptom. Typically, both lower extremities are affected equally. Some patients may have hemiataxia initially before the symptoms become generalized. In some instances, the ataxia begins abruptly following a febrile illness in which ataxia of one lower extremity precedes that of the other.

Gait ataxia manifests as progressively slow and clumsy walking, which often begins after normal walking has developed. The ataxia may be associated with difficulty standing and running. The gait ataxia is both of a sensory and cerebellar type. This combination has been referred to as a tabetocerebellar gait. Opinions are conflicting as to whether the sensory or cerebellar features predominate. The cerebellar features of gait ataxia in FA include a wide-based gait with constant shifting of position to maintain balance. Sitting and standing are associated with titubation.

The sensory ataxia resulting from a loss of joint position sense contributes to the wide-based stance and gait but a steppage gait also is present, characterized by uneven and irregular striking of the floor by the bottom of the feet. Attempts to correct any imbalance typically result in abrupt and wild movements.

As the disease progresses, ataxia affects the trunk, legs, and arms. As the arms become grossly ataxic, both action and intention tremors may develop. Titubation of the trunk may appear. Facial, buccal, and arm muscles may become tremulous and occasionally display choreiform movements. The patient may experience easy fatigability.

Parkinson et al in their recent study revealed that early-onset cases deteriorate rapidly and are more frequently associated with non-neurological features such as diabetes, cardiomyopathy, scoliosis, and pes cavus. Late-onset cases have smaller GAA expansions and tend to have slower progression.[2]

Patients with advanced FA may have profound distal weakness of the legs and feet, although significant weakness of the arms is rare before the patient becomes bedridden. Eventually, the patient is unable to walk because of the progressive weakness and ataxia, becoming wheelchair bound and ultimately bedridden.

With disease progression, dysarthria and dysphagia appear. Speech becomes slurred, slow, and eventually incomprehensible. Patients may experience mild weakening of the facial muscles with associated weakness of swallowing. Incoordination of breathing, speaking, swallowing, and laughing may result in the patient nearly choking while speaking.

Nieto et al administered comprehensive neuropsychological testing measuring multiple domains in 36 FA patients.[3] The observed pattern of neuropsychological impairment is indicative of executive problems and parietotemporal dysfunction. Thus, cognitive impairment in FA is probably caused by the interruption of cerebrocerebellar circuits that have been proposed as the anatomical substrate of the cerebellar involvement in cognition.

Noval et al (2012) used optical coherence tomography to provide a better understanding of the ophthalmic features of FA.[4] The authors revealed that the visual pathway is affected in FA, but, in most patients, there is no significant visual impairment. In a small majority of patients, visual acuity declines with disease progression.

Family history

FA is characterized by autosomal recessive inheritance. In families with one affected child, the subsequent risk of another affected child is 25%. As in most recessive disorders, the risk of developing FA is highest following birth from a consanguineous union. In North America and Europe, however, most cases are sporadic and occur in nonconsanguineous families.

The overall risk of a patient with FA having a child with the condition is approximately 1 in 200, unless consanguinity is involved. If that same patient has a partner who is found to be a carrier of FA, then the risk becomes 1 in 2. Children descended from the unaffected sibling of a patient with FA and a nonconsanguineous spouse have a 1:1000 risk of developing FA.

Carrier testing is available for relatives of affected patients and their partners. However, the small risk of a point mutation also must be taken into account and incorporated into genetics counseling.

Prenatal diagnosis via direct mutation testing is also available.

Physical

See Staging for a scale used to rate functional disability in patients with Friedreich ataxia.

The cardinal features of FA are as follows:

Physical findings in patients with FA include gradual loss of vibratory and position senses from the onset, initially affecting the feet and hands. With progression, light touch, pain, and temperature sensation also may be diminished. Two-point discrimination may be affected in early cases.

Tendon reflexes are absent in almost all cases but may be weakly present if the patient is examined early in the course of the disease. Patellar and ankle jerks are affected most profoundly. Flexor spasms are common even in the absence of tendon reflexes, indicating that the areflexia is sensory in origin. Plantar reflexes are extensor in 90% of patients and absent in the rest. Abdominal reflexes usually are retained until late in the disease process.

Distal wasting, primarily of the upper extremities, is seen in 50% of patients. Muscle tone is usually normal or decreased. Sphincter control is usually intact but cases of urinary urgency and constipation, which are not usually severe, have occurred.

Kyphoscoliosis is a frequent but nonspecific finding that may be severe. With early onset, severe scoliosis can produce significant cardiopulmonary morbidity and death due to restricted respiratory function. Peripheral cyanosis of the lower limbs is common.

Foot deformities such as high plantar arches, foot inversion, and hammertoes may precede the gait ataxia. Typically, 50% of patients develop either pes cavus or varus deformities of one or both feet.

Visual acuity rarely is affected, but optic atrophy occurs in 25% of patients with FA and has resulted in occasional blindness. Visual evoked potentials (VEP) are abnormal in two thirds of patients, typically displaying reduced amplitude and delayed latency.

Nystagmus of various types may be found. Horizontal nystagmus, present in the primary position and increased on lateral gaze, occurs in 20% of patients with FA. Extraocular movements usually are affected and are characterized by abnormal square-wave jerks and saccadic pursuit, poor fixation, and impaired vestibulo-ocular reflexes. Pupillary reflexes are normal.

Deafness has been noted, in some cases in association with vertigo. Brain stem auditory evoked responses (BAER) are typically consistent with central auditory pathway pathology.

Hypertrophic cardiomyopathy develops in more than 50% of patients. Myocarditis, myocardial fibrosis, cardiac enlargement, progressive cardiac failure, tachycardias, and heart block also may be seen. Cardiac arrhythmia and congestive heart failure contribute to a significant number of deaths in patients with FA.

Approximately 10% of patients with FA develop diabetes mellitus. An even larger percentage demonstrate impaired glucose tolerance, which has been associated with an insulin receptor abnormality.

Intellectual disability, psychosis, and dementia are uncommon, but some degree of cognitive dysfunction may occur. Emotional lability, in the presence of normal cognitive function, is frequently present.

Causes

Classic Friedreich ataxia is the result of a gene mutation at the centromeric region of chromosome 9 (9q13-21.1) at the site of the gene encoding for the 210-amino-acid protein frataxin. This mutation is characterized by an excessive number of repeats of the GAA (guanine adenine adenine) trinucleotide DNA sequence in the first intron of the gene coding for frataxin. It is the only disease known to be the result of a GAA trinucleotide repeat. This expansion alters the expression of the gene, decreasing the synthesis of frataxin protein. The expanded GAA repeat is thought to result in frataxin deficiency by interfering with transcription of the gene by adopting an unstable helical structure. The larger the number of repeats, the more profound is the reduction in frataxin expression. A recent study from Stolle and colleagues have shown that interrupted GAA repeats do not clearly impact the age of onset in FA.[5]

In a normal chromosome, this trinucleotide sequence is repeated up to 50 times. In patients with FA, this sequence is repeated at least 200 times and often more than 1000 times.

Variability in the clinical presentation of FA may be explained by the extent of this trinucleotide repeat expansion. The age of disease onset, its severity, rate of progression, and extent of neurological involvement vary with the number of repetitive GAA sequences.

In a study by Durr et al, the large majority of patients with FA (94%) were homozygous for the GAA trinucleotide (the GAA expansion was present on both alleles of the frataxin gene). The remaining 6% were compound heterozygotes for a GAA expansion and a frataxin point mutation (one allele had a GAA expansion, and the other had a point mutation with no expansion). No patients have been described who were homozygous for a point mutation.[6]

Point mutations not only reduce levels of the frataxin protein but are also responsible for the creation of abnormal protein. They also represent another source of variability in the clinical presentation of FA. Seventeen different point mutations have so far been described in FA.

Cells and tissues of the body are differentially sensitive to frataxin deficiency. Cells normally requiring and producing greater amounts of frataxin tend to be most affected by FA. For example, sensory neurons in the dorsal root ganglion responsible for position sense highly express the frataxin gene and are affected greatly in FA. Myocardial muscle fiber also requires comparably larger amounts of frataxin than other tissues and is affected markedly in FA.

A number of experiments have confirmed the mitochondrial subcellular localization of frataxin in mammals. Frataxin has been shown to be essential for normal mitochondrial function, both for oxidative phosphorylation and iron homeostasis. Strong evidence exists that frataxin deficiency results in iron accumulation within mitochondria of affected cells in cell culture lines. Apparently, the rate of mitochondrial export is reduced. Hearts of patients with FA have revealed mitochondrial ironlike deposits that are not present in healthy hearts. Frataxin-deficient cells not only generate more free radicals, but also show a reduced capacity to mobilize antioxidant defenses. The search for experimental drugs increasing the amount of frataxin is a very active and timely area of investigation.[7]

The excessive mitochondrial accumulation of iron affects cytosolic iron levels. Excess intracellular iron stimulates the increased generation of free radicals and mitochondrial damage. Iron excess inactivates mitochondrial enzymes essential for the production of adenosine triphosphate (ATP). Cell death, particularly of neurons of the spinal cord and peripheral nervous system, ensues. A mouse model of FA is being developed to confirm evidence of this process in living animal models.

Laboratory Studies

Genetic counseling is available for prenatal diagnosis of Friedreich ataxia for parents with one affected child. Population screening for carriers of the defective gene is not practical. A specific trinucleotide repeat expansion assay is available commercially in the United States and should be performed in all suspected cases of FA.

Total levels of Fe in bulk extracts were not significantly higher than normal, and the concentrations of Zn also remained in the normal range. Cu levels, however, were significantly lower in FA.

No evidence of CSF abnormality exists in patients with FA.

Imaging Studies

Magnetic resonance imaging (MRI) is the study of choice in the evaluation of the atrophic changes seen in Friedreich ataxia. MRI of the brain and spinal cord in patients with FA consistently shows atrophy of the cervical spinal cord with minimal evidence of cerebellar atrophy.

Transcranial sonography provides a quick-to-apply and inexpensive in vivo assessment of both cerebellar and noncerebellar abnormalities in FA, in particular highlighting dentate hyperechogenicity as a core feature.[8]

Other Tests

Echocardiography reveals symmetric, concentric ventricular hypertrophy, although some have asymmetric septal hypertrophy.

Approximately 65% of patients with FA have abnormal ECG findings. The most common findings are T-wave inversion, particularly in the inferior standard and lateral chest leads, and ventricular hypertrophy.

Nerve conduction velocity (NCV) study findings in FA usually are normal or display only mildly reduced velocities. Sensory nerve action potentials (SNAP) are absent in greater than 90% of patients with FA. The remaining 10% display reduced-amplitude SNAPs.

Brainstem auditory evoked responses are typically abnormal in FA, displaying absent waves III and IV with preservation of wave I. This is suggestive of involvement of central auditory pathways.

Visual evoked potentials are abnormal in two thirds of patients with FA. Absent or delayed latency and reduced amplitude of the p100 wave are seen.

Somatosensory evoked potentials (SSEP) reveal delayed central conduction time (N13a/N20, N13b/N20), dispersed potentials at the sensory cortex, as well as abnormal central motor conduction.

Borchers et al compared clinical tests for joint position sense (JPS) and vibration sense (VS) to a test of spatial position sense (SPS).[9] The SPS test was more sensitive than JPS and VS and revealed deficits potentially earlier than clinical screening tests. Only the SPS showed a positive correlation with ataxia severity. Their results indicate that proprioceptive deficits in Friedreich ataxia start earlier and are more severe than indicated by routine standard clinical testing.

Histologic Findings

A cross-section through the lower cervical cord clearly shows loss of myelinated fibers of the dorsal columns and the corticospinal tracts (Weil stain). Milder involvement of spinocerebellar tracts is also present. The affected tracts show compact fibrillary gliosis on hematoxylin and eosin (H&E) stain but no breakdown products or macrophages, reflecting the very slow rate of degeneration and death of fibers. The dorsal spinal ganglia show shrinkage and eventual disappearance of neurons associated with proliferation of capsular cells (H&E). The posterior roots are nearly devoid of large myelinated fibers. Within the thoracic spinal cord, degeneration and loss of cells of the Clarke column are apparent.

Staging

Currently, there are 3 different scales that are most frequently applied: The International Cooperative Ataxia Rating Scale (ICARS), the Friedreich Ataxia Rating Scale (FARS), and the Scale for the Assessment and Rating of Ataxia (SARA). All scales have been validated and compared with regard to their testing properties.[10] )

International Cooperative Ataxia Rating Scale (World Federation of Neurology)

Posture and gait disturbances

Kinetic functions

Speech disorders

Oculomotor disorders

Medical Care

The results of treating ataxia in Friedreich ataxia (FA) have generally been disappointing. No therapeutic measures are known to alter the natural history of the neurological disease. Standard treatment is administered for heart failure, arrhythmias, and diabetes mellitus.

High-dose propranolol has been described in a case report by Kosutic with reduction in thickness of the septal and posterior left ventricular walls and with complete normalization of diffuse electrocardiographic repolarization abnormalities.[11]

Li and colleagues have shown that therapeutic efforts should focus on an approach that combines iron removal from mitochondria with a treatment that increases cytosolic iron levels to maximize residual frataxin expression in patients with FA.[12]

The other therapies that have been used are as follows:

5-Hydroxytryptophan

5-Hydroxytryptophan is a serotonin precursor that has been used for a decade or more by Trouillas et al to treat various forms of ataxia with mixed results.[13] This drug was known to suppress posthypoxic action myoclonus. The rationale for use of the drug in FA was that FA may in part be due to a cerebellar deficiency of serotonin.

The results of a double-blind, cross-over study by Trouillas et al demonstrated that the levorotatory form of 5-hydroxytryptophan was able to significantly modify the cerebellar symptoms in patients with FA; however, the effect was only partial and not clinically major.[13]

A study by Wessel demonstrated stabilization of posture in patients receiving long-term treatment with 5-hydroxytryptophan and a clear deterioration in patients who did not receive the treatment. This form of treatment requires further study.

Coenzyme Q

Coenzyme Q is an antioxidant that can buffer free radical formation that is induced by excess mitochondrial iron. A combined coenzyme Q (400 mg/d) and vitamin E (2100 IU/d) therapy has been used in a study of 10 patients with slowing of the progression of certain clinical features and a significant improvement in cardiac function.

Experimental studies are underway to evaluate the use of coenzyme Q derivatives in limiting the toxicity of iron to mitochondrial structures.

Idebenone

Idebenone has been used as therapy for Friedreich ataxia for more than a decade. At present, several studies have assessed the influence of therapy on neurologic or cardiac function.

The effect of intermediate-dose idebenone (20 mg/kg/d) on quality of life and neurologic function was assessed in a recent study by Brandsema et al.[14] The Pediatric Quality of Life Inventory, the International Cooperative Ataxia Rating Scale, and an Activities of Daily Living Scale before initiation of idebenone therapy and after 1 year of therapy were assessed.

Tomassini et al (2012) have revealed that in vivo treatment with interferon-gamma increases frataxin expression in dorsal root ganglia neurons, prevents their pathological changes, and ameliorates the sensorimotor performance in FA mice. These results disclose new roles for interferon-gamma in cellular metabolism and have direct implications for the treatment of FA.[17]

Pandolfo et al have recently reviewed the utility of deferiprone with specific regard to its iron chelating properties and clinical benefits.[18]

In a study done by Elincx-Benizri et al, the authors presented their experience of 5 FA patients treated with deferiprone (20 mg/kg/day), in addition to idebenone treatment, followed over a period of 10-24 months, under off-label authorization. The authors concluded that combination therapy of a low dose of deferiprone with idebenone is relatively safe and might improve neurological function and heart hypertrophy.[54] However, future studies are needed.

Surgical Care

Apart from surgery for scoliosis and foot deformities that may be helpful in selected cases, no significant surgical treatment is available for Friedreich ataxia. A study from Milbrandt and colleagues emphasized that in scoliosis braces were seldom effective and that segmental constructs are effective in creating substantial intraoperative correction and maintaining correction postoperatively.[19]

In addition, heart transplantation for FA dilated cardiomyopathy has been performed.

Consultations

Goulipian and colleagues have mentioned the role of orthopedic shoes combined with physical therapy. Their results demonstrated that orthopedic shoes improved gait disorders in a patient with Friedreich ataxia.[20]

Diet

No data exist to suggest that any alteration of diet would affect the onset, progression, or outcome of this disease.

Activity

No data exist to suggest that any alteration of activity level would affect the onset, progression, or outcome of this disease.

In regards to the delivery and utilization of oxygen in response to exercise, near infrared muscle spectroscopy may be an effective tool for monitoring the biochemical and functional features of FA.

A study by Milne, et al. concluded that management of spasticity and reduced muscle length should be considered in people with FA at disease onset to optimize function.[55]

Prognosis

See the list below:

Author

Jasvinder Chawla, MD, MBA, Chief of Neurology, Hines Veterans Affairs Hospital; Professor of Neurology, Loyola University Medical Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Florian P Thomas, MD, PhD, MA, MS, Chair, Neuroscience Institute and Department of Neurology, Director, National MS Society Multiple Sclerosis Center and Hereditary Neuropathy Foundation Center of Excellence, Hackensack University Medical Center; Founding Chair and Professor, Department of Neurology, Hackensack Meridian School of Medicine at Seton Hall University; Professor Emeritus, Department of Neurology, St Louis University School of Medicine; Editor-in-Chief, Journal of Spinal Cord Medicine

Disclosure: Nothing to disclose.

Chief Editor

Selim R Benbadis, MD, Professor, Director of Comprehensive Epilepsy Program, Departments of Neurology and Neurosurgery, Tampa General Hospital, University of South Florida Morsani College of Medicine

Disclosure: Serve(d) as a director, officer, partner, employee, advisor, consultant or trustee for: Ceribell, Eisai, Greenwich, Growhealthy, LivaNova, Neuropace, SK biopharmaceuticals, Sunovion<br/>Serve(d) as a speaker or a member of a speakers bureau for: Eisai, Greenwich, LivaNova, Sunovion<br/>Received research grant from: Cavion, LivaNova, Greenwich, Sunovion, SK biopharmaceuticals, Takeda, UCB.

Additional Contributors

Dianna Quan, MD, Professor of Neurology, Director of Electromyography Laboratory, University of Colorado School of Medicine

Disclosure: Received research grant from: Alnylam; Pfizer; Cytokinetics; Momenta.

References

  1. Harding AE, Zilkha KJ. Pseudo-dominant' inheritance in Friedreich's ataxia. J Med Genet. 1981 Aug. 18(4):285-7. [View Abstract]
  2. Parkinson MH, Boesch S, Nachbauer W, Mariotti C, Giunti P. Clinical features of Friedreich's ataxia: classical and atypical phenotypes. J Neurochem. 2013 Aug. 126 Suppl 1:103-17. [View Abstract]
  3. Nieto A, Correia R, de Nóbrega E, Montón F, Hess S, Barroso J. Cognition in Friedreich Ataxia. Cerebellum. 2012 Feb 16. [View Abstract]
  4. Noval S, Contreras I, Sanz-Gallego I, Manrique RK, Arpa J. Ophthalmic features of Friedreich ataxia. Eye (Lond). 2012 Feb. 26(2):315-20. [View Abstract]
  5. Stolle CA, Frackelton EC, McCallum J, Farmer JM, Tsou A, Wilson RB, et al. Novel, complex interruptions of the GAA repeat in small, expanded alleles of two affected siblings with late-onset Friedreich ataxia. Mov Disord. 2008 Jul 15. 23(9):1303-6. [View Abstract]
  6. Dürr A, Cossee M, Agid Y, Campuzano V, Mignard C, Penet C, et al. Clinical and genetic abnormalities in patients with Friedreich's ataxia. N Engl J Med. 1996 Oct 17. 335(16):1169-75. [View Abstract]
  7. Marmolino D. Friedreich's ataxia: past, present and future. Brain Res Rev. 2011 Jun 24. 67(1-2):311-30. [View Abstract]
  8. Synofzik M, Godau J, Lindig T, Schöls L, Berg D. Transcranial sonography reveals cerebellar, nigral, and forebrain abnormalities in Friedreich's ataxia. Neurodegener Dis. 2011. 8(6):470-5. [View Abstract]
  9. Borchers S, Synofzik M, Kiely E, Himmelbach M. Routine clinical testing underestimates proprioceptive deficits in Friedreich's ataxia. Cerebellum. 2013 Dec. 12(6):916-22. [View Abstract]
  10. Bürk K, Schulz SR, Schulz JB. Monitoring progression in Friedreich ataxia (FRDA): the use of clinical scales. J Neurochem. 2013 Aug. 126 Suppl 1:118-24. [View Abstract]
  11. Kosutic J, Zamurovic D. High-dose beta-blocker hypertrophic cardiomyopathy therapy in a patient with Friedreich ataxia. Pediatr Cardiol. 2005 Sep-Oct. 26(5):727-30. [View Abstract]
  12. Li K, Besse EK, Ha D, Kovtunovych G, Rouault TA. Iron-dependent regulation of frataxin expression: implications for treatment of Friedreich ataxia. Hum Mol Genet. 2008 Aug 1. 17(15):2265-73. [View Abstract]
  13. Trouillas P, Serratrice G, Laplane D, Rascol A, Augustin P, Barroche G, et al. Levorotatory form of 5-hydroxytryptophan in Friedreich's ataxia. Results of a double-blind drug-placebo cooperative study. Arch Neurol. 1995 May. 52(5):456-60. [View Abstract]
  14. Brandsema JF, Stephens D, Hartley J, Yoon G. Intermediate-dose idebenone and quality of life in Friedreich ataxia. Pediatr Neurol. 2010 May. 42(5):338-42. [View Abstract]
  15. Parkinson MH, Schulz JB, Giunti P. Co-enzyme Q10 and idebenone use in Friedreich's ataxia. J Neurochem. 2013 Aug. 126 Suppl 1:125-41. [View Abstract]
  16. Lynch DR, Perlman SL, Meier T. A Phase 3, Double-blind, Placebo-Controlled Trial of Idebenone in Friedreich Ataxia. Arch Neurol. 2010 Aug. 67(8):941-7. [View Abstract]
  17. Tomassini B, Arcuri G, Fortuni S, Sandi C, Ezzatizadeh V, Casali C, et al. Interferon gamma upregulates frataxin and corrects the functional deficits in a Friedreich ataxia model. Hum Mol Genet. 2012 Mar 23. [View Abstract]
  18. Pandolfo M, Hausmann L. Deferiprone for the treatment of Friedreich's ataxia. J Neurochem. 2013 Aug. 126 Suppl 1:142-6. [View Abstract]
  19. Milbrandt TA, Kunes JR, Karol LA. Friedreich's ataxia and scoliosis: the experience at two institutions. J Pediatr Orthop. 2008 Mar. 28(2):234-8. [View Abstract]
  20. Goulipian C, Bensoussan L, Viton JM, Milhe-De Bovis V, Ramon J, Delarque A. Orthopedic shoes improve gait in Friedreich's ataxia: a clinical and quantified case study. Eur J Phys Rehabil Med. 2008 Mar. 44(1):93-8. [View Abstract]
  21. Tsou AY, Paulsen EK, Lagedrost SJ, Perlman SL, Mathews KD, Wilmot GR, et al. Mortality in Friedreich ataxia. J Neurol Sci. 2011 Aug 15. 307(1-2):46-9. [View Abstract]
  22. Weidemann F, Rummey C, Bijnens B, Störk S, Jasaityte R, Dhooge J, et al. The Heart in Friedreich Ataxia: Definition of Cardiomyopathy, Disease Severity, and Correlation with Neurological Symptoms. Circulation. 2012 Feb 29. [View Abstract]
  23. Wilson CL, Fahey MC, Corben LA, Collins VR, Churchyard AJ, Lamont PJ, et al. Quality of life in Friedreich ataxia: what clinical, social and demographic factors are important?. Eur J Neurol. 2007 Sep. 14(9):1040-7. [View Abstract]
  24. Paulsen EK, Friedman LS, Myers LM, Lynch DR. Health-related quality of life in children with Friedreich ataxia. Pediatr Neurol. 2010 May. 42(5):335-7. [View Abstract]
  25. Beltinger A, Riffel B, Stöhr M. Somatosensory evoked potentials following median and tibial nerve stimulation in patients with Friedreich's ataxia. Eur Arch Psychiatry Neurol Sci. 1987. 236(6):358-63. [View Abstract]
  26. Chamberlain S, Koenig M, Richter A, Palau F, Pandolfo M. Molecular analysis of the Friedreich's ataxia locus. Adv Neurol. 1993. 61:193-204. [View Abstract]
  27. Collins A. Clinical neurogenetics: friedreich ataxia. Neurol Clin. 2013 Nov. 31(4):1095-120. [View Abstract]
  28. Currier RD. A treatment for ataxia [editorial]. Arch Neurol. 1995 May. 52(5):449. [View Abstract]
  29. Delatycki MB, Williamson R, Forrest SM. Friedreich ataxia: an overview. J Med Genet. 2000 Jan. 37(1):1-8. [View Abstract]
  30. Di Prospero NA, Baker A, Jeffries N, Fischbeck KH. Neurological effects of high-dose idebenone in patients with Friedreich's ataxia: a randomised, placebo-controlled trial. Lancet Neurol. 2007 Oct. 6(10):878-86. [View Abstract]
  31. Epstein E, Farmer JM, Tsou A, Perlman S, Subramony SH, Gomez CM, et al. Health related quality of life measures in Friedreich Ataxia. J Neurol Sci. 2008 Sep 15. 272(1-2):123-8. [View Abstract]
  32. Forrest SM, Knight M, Delatycki MB, et al. The correlation of clinical phenotype in Friedreich ataxia with the site of point mutations in the FRDA gene. Neurogenetics. 1998 Aug. 1(4):253-7. [View Abstract]
  33. Friedman JH. Machado-Joseph disease/spinocerebellar ataxia 3 responsive to buspirone. Mov Disord. 1997 Jul. 12(4):613-4. [View Abstract]
  34. Friedman LS, Farmer JM, Perlman S, Wilmot G, Gomez CM, Bushara KO, et al. Measuring the rate of progression in Friedreich ataxia: implications for clinical trial design. Mov Disord. 2010 Mar 15. 25(4):426-32. [View Abstract]
  35. Hart PE, Lodi R, Rajagopalan B. Antioxidant treatment of patients with Friedreich ataxia: four-year follow-up. Arch Neurol. 2005 Apr. 62(4):621-6. [View Abstract]
  36. Jitpimolmard S, Small J, King RH, Geddes J, Misra P, McLaughlin J, et al. The sensory neuropathy of Friedreich's ataxia: an autopsy study of a case with prolonged survival. Acta Neuropathol. 1993. 86(1):29-35. [View Abstract]
  37. Johnson WG. Friedreich ataxia. Clin Neurosci. 1995. 3(1):33-8. [View Abstract]
  38. Koeppen AH. The hereditary ataxias. J Neuropathol Exp Neurol. 1998 Jun. 57(6):531-43. [View Abstract]
  39. Lane DJ, Richardson DR. Frataxin, a molecule of mystery: trading stability for function in its iron-binding site. Biochem J. 2010 Feb 9. 426(2):e1-3. [View Abstract]
  40. Lynch DR, Lech G, Farmer JM, Balcer LJ, Bank W, Chance B, et al. Near infrared muscle spectroscopy in patients with Friedreich's ataxia. Muscle Nerve. 2002 May. 25(5):664-73. [View Abstract]
  41. Mello KA, Abbott BP. Effect of sulfamethoxazole and trimethoprim on neurologic dysfunction in a patient with Joseph's disease. Arch Neurol. 1988 Feb. 45(2):210-3. [View Abstract]
  42. Pandolfo M. Friedreich ataxia. Handb Clin Neurol. 2012. 103:275-94. [View Abstract]
  43. Pandolfo M. Friedreich's ataxia: new development and perspectives. Generations: The Official Publication of the National Ataxia Foundation. 1999. 27(1):6-9.
  44. Schipper HM. Neurodegeneration with brain iron accumulation - clinical syndromes and neuroimaging. Biochim Biophys Acta. 2012 Mar. 1822(3):350-60. [View Abstract]
  45. Schmucker S, Puccio H. Understanding the molecular mechanisms of Friedreich Ataxia to develop therapeutic approaches. Hum Mol Genet. 2010 Apr 22. [View Abstract]
  46. Schulz JB, Pandolfo M. 150 years of Friedreich ataxia: from its discovery to therapy. J Neurochem. 2013 Aug. 126 Suppl 1:1-3. [View Abstract]
  47. Sedlak TL, Chandavimol M, Straatman L. Cardiac transplantation: a temporary solution for Friedreich's ataxia-induced dilated cardiomyopathy. J Heart Lung Transplant. 2004 Nov. 23(11):1304-6. [View Abstract]
  48. Stockner H, Sojer M, Hering S, Nachbauer W, Seppi K, Schmidauer C, et al. Substantia nigra hypoechogenicity in Friedreich ataxia. Mov Disord. 2012 Feb. 27(2):332-3. [View Abstract]
  49. Subramony SH, McDaniel O, Vedanarayanan VV. Very late-onset Friedreich's ataxia. Mov Disord. 1999. 14(5):904.
  50. Trouillas P, Takayanagi T, Hallett M. International Cooperative Ataxia Rating Scale for pharmacological assessment of the cerebellar syndrome. The Ataxia Neuropharmacology Committee of the World Federation of Neurology. J Neurol Sci. 1997 Feb 12. 145(2):205-11. [View Abstract]
  51. Voncken M, Ioannou P, Delatycki MB. Friedreich ataxia-update on pathogenesis and possible therapies. Neurogenetics. 2004 Feb. 5(1):1-8. [View Abstract]
  52. Wills AJ, Marsden CD. Fifty Neurologic Cases from the National Hospital. 1999.
  53. Zouari M, Feki M, Ben Hamida C, Larnaout A, Turki I, Belal S, et al. Electrophysiology and nerve biopsy: comparative study in Friedreich's ataxia and Friedreich's ataxia phenotype with vitamin E deficiency. Neuromuscul Disord. 1998 Aug. 8(6):416-25. [View Abstract]
  54. Elincx-Benizri S, Glik A, Merkel D, Arad M, Freimark D, Kozlova E, et al. Clinical Experience With Deferiprone Treatment for Friedreich Ataxia. J Child Neurol. 2016 Mar 29. [View Abstract]
  55. Milne SC, Corben LA, Yiu E, Delatycki MB, Georgiou-Karistianis N. Gastrocnemius and soleus spasticity and muscle length in Friedreich's ataxia. J Clin Neurosci. 2016 Mar 25. [View Abstract]
  56. Aranca TV, Jones TM, Shaw JD, Staffetti JS, Ashizawa T, Kuo SH, et al. Emerging therapies in Friedreich's ataxia. Neurodegener Dis Manag. 2016 Feb. 6 (1):49-65. [View Abstract]
  57. Rezende TJ, Silva CB, Yassuda CL, Campos BM, D'Abreu A, Cendes F, et al. Longitudinal magnetic resonance imaging study shows progressive pyramidal and callosal damage in Friedreich's ataxia. Mov Disord. 2016 Jan. 31 (1):70-8. [View Abstract]