Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
EBioMedicine ; 102: 105076, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38507876

ABSTRACT

BACKGROUND: GAA-FGF14 disease/spinocerebellar ataxia 27B is a recently described neurodegenerative disease caused by (GAA)≥250 expansions in the fibroblast growth factor 14 (FGF14) gene, but its phenotypic spectrum, pathogenic threshold, and evidence-based treatability remain to be established. We report on the frequency of FGF14 (GAA)≥250 and (GAA)200-249 expansions in a large cohort of patients with idiopathic downbeat nystagmus (DBN) and their response to 4-aminopyridine. METHODS: Retrospective cohort study of 170 patients with idiopathic DBN, comprising in-depth phenotyping and assessment of 4-aminopyridine treatment response, including re-analysis of placebo-controlled video-oculography treatment response data from a previous randomised double-blind 4-aminopyridine trial. FINDINGS: Frequency of FGF14 (GAA)≥250 expansions was 48% (82/170) in patients with idiopathic DBN. Additional cerebellar ocular motor signs were observed in 100% (82/82) and cerebellar ataxia in 43% (35/82) of patients carrying an FGF14 (GAA)≥250 expansion. FGF14 (GAA)200-249 alleles were enriched in patients with DBN (12%; 20/170) compared to controls (0.87%; 19/2191; OR, 15.20; 95% CI, 7.52-30.80; p < 0.0001). The phenotype of patients carrying a (GAA)200-249 allele closely mirrored that of patients carrying a (GAA)≥250 allele. Patients carrying a (GAA)≥250 or a (GAA)200-249 allele had a significantly greater clinician-reported (80%, 33/41 vs 31%, 5/16; RR, 2.58; 95% CI, 1.23-5.41; Fisher's exact test, p = 0.0011) and self-reported (59%, 32/54 vs 11%, 2/19; RR, 5.63; 95% CI, 1.49-21.27; Fisher's exact test, p = 0.00033) response to 4-aminopyridine treatment compared to patients carrying a (GAA)<200 allele. Placebo-controlled video-oculography data, available for four patients carrying an FGF14 (GAA)≥250 expansion, showed a significant decrease in slow phase velocity of DBN with 4-aminopyridine, but not placebo. INTERPRETATION: This study confirms that FGF14 GAA expansions are a frequent cause of DBN syndromes. It provides preliminary evidence that (GAA)200-249 alleles might be pathogenic. Finally, it provides large real-world and preliminary piloting placebo-controlled evidence for the efficacy of 4-aminopyridine in GAA-FGF14 disease. FUNDING: This work was supported by the Clinician Scientist program "PRECISE.net" funded by the Else Kröner-Fresenius-Stiftung (to CW, AT, and MSy), the grant 779257 "Solve-RD" from the European's Union Horizon 2020 research and innovation program (to MSy), and the grant 01EO 1401 by the German Federal Ministry of Education and Research (BMBF) (to MSt). This work was also supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) N° 441409627, as part of the PROSPAX consortium under the frame of EJP RD, the European Joint Programme on Rare Diseases, under the EJP RD COFUND-EJP N° 825575 (to MSy, BB and-as associated partner-SZ), the NIH National Institute of Neurological Disorders and Stroke (grant 2R01NS072248-11A1 to SZ), the Fondation Groupe Monaco (to BB), and the Montreal General Hospital Foundation (grant PT79418 to BB). The Care4Rare Canada Consortium is funded in part by Genome Canada and the Ontario Genomics Institute (OGI-147 to KMB), the Canadian Institutes of Health Research (CIHR GP1-155867 to KMB), Ontario Research Foundation, Genome Quebec, and the Children's Hospital of Eastern Ontario Foundation. The funders had no role in the conduct of this study.


Subject(s)
Fibroblast Growth Factors , Neurodegenerative Diseases , Nystagmus, Pathologic , Child , Humans , 4-Aminopyridine/therapeutic use , Neurodegenerative Diseases/drug therapy , Nystagmus, Pathologic/chemically induced , Nystagmus, Pathologic/drug therapy , Ontario , Retrospective Studies
3.
J Neurol ; 271(5): 2886-2892, 2024 May.
Article in English | MEDLINE | ID: mdl-38381176

ABSTRACT

OBJECTIVES: The cause of downbeat nystagmus (DBN) remains unknown in a substantial number of patients ("idiopathic"), although intronic GAA expansions in FGF14 have recently been shown to account for almost 50% of yet idiopathic cases. Here, we hypothesized that biallelic RFC1 expansions may also represent a recurrent cause of DBN syndrome. METHODS: We genotyped the RFC1 repeat and performed in-depth phenotyping in 203 patients with DBN, including 65 patients with idiopathic DBN, 102 patients carrying an FGF14 GAA expansion, and 36 patients with presumed secondary DBN. RESULTS: Biallelic RFC1 AAGGG expansions were identified in 15/65 patients with idiopathic DBN (23%). None of the 102 GAA-FGF14-positive patients, but 2/36 (6%) of patients with presumed secondary DBN carried biallelic RFC1 expansions. The DBN syndrome in RFC1-positive patients was characterized by additional cerebellar impairment in 100% (15/15), bilateral vestibulopathy (BVP) in 100% (15/15), and polyneuropathy in 80% (12/15) of cases. Compared to GAA-FGF14-positive and genetically unexplained patients, RFC1-positive patients had significantly more frequent neuropathic features on examination and BVP. Furthermore, vestibular function, as measured by the video head impulse test, was significantly more impaired in RFC1-positive patients. DISCUSSION: Biallelic RFC1 expansions are a common monogenic cause of DBN syndrome.


Subject(s)
Nystagmus, Pathologic , Phenotype , Replication Protein C , Humans , Replication Protein C/genetics , Male , Female , Middle Aged , Adult , Nystagmus, Pathologic/genetics , Aged , DNA Repeat Expansion/genetics , Fibroblast Growth Factors/genetics , Young Adult , Bilateral Vestibulopathy/genetics , Bilateral Vestibulopathy/physiopathology
4.
Laryngorhinootologie ; 103(6): 413-421, 2024 Jun.
Article in German | MEDLINE | ID: mdl-38195848

ABSTRACT

The diagnosis of ocular motor disorders and the different forms of a nystagmus is based on a systematic clinical examination of all types of eye movements: eye position, spontaneous nystagmus, range of eye movements, smooth pursuit, saccades, gaze-holding function, vergence, optokinetic nystagmus, as well as testing of the function of the vestibulo-ocular reflex (VOR) and visual fixation suppression of the VOR. Relevant anatomical structures are the midbrain, pons, medulla, cerebellum, and cortex. There is a simple clinical rule: vertical and torsional eye movements are generated in the midbrain, horizontal in the pons. The cerebellum is relevant for almost all types of eye movements; typical pathological findings are saccadic smooth pursuit, gaze-evoked nystagmus or dysmetric saccades.Nystagmus is defined as a rhythmic, most often involuntary eye movement. It normally consists of a slow (pathological) drift of the eyes and a fast central compensatory movement of the eyes back to the primary position (re-fixation saccade). There are three major categories: first, spontaneous nystagmus, i. e. nystagmus which occurs in the gaze straight ahead position as upbeat or downbeat nystagmus; second, nystagmus that becomes visible at eccentric gaze only and third, nystagmus which can be elicited by certain maneuvers, e. g. head-shaking, head positioning, air pressure or hyperventilation, most of which are of peripheral vestibular origin. The most frequent central types of spontaneous nystagmus are downbeat and upbeat, infantile, pure torsional, pendular fixation, periodic alternating, and seesaw nystagmus. Many types of central nystagmus allow a precise neuroanatomical localization: for instance, downbeat nystagmus, which is most often caused by a bilateral floccular lesion or dysfunction, or upbeat nystagmus, which is caused by a lesion in the mesencephalon or medulla oblongata. Examples of pharmacotherapy are the use of 4-aminopyridine for downbeat and upbeat nystagmus, memantine or gabapentin for fixation pendular nystagmus or baclofen for periodic alternating nystagmus.


Subject(s)
Nystagmus, Pathologic , Reflex, Vestibulo-Ocular , Humans , Nystagmus, Pathologic/diagnosis , Nystagmus, Pathologic/physiopathology , Reflex, Vestibulo-Ocular/physiology , Ocular Motility Disorders/physiopathology , Ocular Motility Disorders/diagnosis , Ocular Motility Disorders/therapy , Saccades/physiology
5.
N Engl J Med ; 390(5): 421-431, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38294974

ABSTRACT

BACKGROUND: Niemann-Pick disease type C is a rare lysosomal storage disorder. We evaluated the safety and efficacy of N-acetyl-l-leucine (NALL), an agent that potentially ameliorates lysosomal and metabolic dysfunction, for the treatment of Niemann-Pick disease type C. METHODS: In this double-blind, placebo-controlled, crossover trial, we randomly assigned patients 4 years of age or older with genetically confirmed Niemann-Pick disease type C in a 1:1 ratio to receive NALL for 12 weeks, followed by placebo for 12 weeks, or to receive placebo for 12 weeks, followed by NALL for 12 weeks. NALL or matching placebo was administered orally two to three times per day, with patients 4 to 12 years of age receiving weight-based doses (2 to 4 g per day) and those 13 years of age or older receiving a dose of 4 g per day. The primary end point was the total score on the Scale for the Assessment and Rating of Ataxia (SARA; range, 0 to 40, with lower scores indicating better neurologic status). Secondary end points included scores on the Clinical Global Impression of Improvement, the Spinocerebellar Ataxia Functional Index, and the Modified Disability Rating Scale. Crossover data from the two 12-week periods in each group were included in the comparisons of NALL with placebo. RESULTS: A total of 60 patients 5 to 67 years of age were enrolled. The mean baseline SARA total scores used in the primary analysis were 15.88 before receipt of the first dose of NALL (60 patients) and 15.68 before receipt of the first dose of placebo (59 patients; 1 patient never received placebo). The mean (±SD) change from baseline in the SARA total score was -1.97±2.43 points after 12 weeks of receiving NALL and -0.60±2.39 points after 12 weeks of receiving placebo (least-squares mean difference, -1.28 points; 95% confidence interval, -1.91 to -0.65; P<0.001). The results for the secondary end points were generally supportive of the findings in the primary analysis, but these were not adjusted for multiple comparisons. The incidence of adverse events was similar with NALL and placebo, and no treatment-related serious adverse events occurred. CONCLUSIONS: Among patients with Niemann-Pick disease type C, treatment with NALL for 12 weeks led to better neurologic status than placebo. A longer period is needed to determine the long-term effects of this agent in patients with Niemann-Pick disease type C. (Funded by IntraBio; ClinicalTrials.gov number, NCT05163288; EudraCT number, 2021-005356-10.).


Subject(s)
Central Nervous System Agents , Niemann-Pick Disease, Type C , Humans , Data Collection , Double-Blind Method , Leucine/analogs & derivatives , Leucine/therapeutic use , Niemann-Pick Disease, Type C/complications , Niemann-Pick Disease, Type C/diagnosis , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Treatment Outcome , Cross-Over Studies , Child, Preschool , Child , Adolescent , Young Adult , Adult , Middle Aged , Aged , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/therapeutic use
6.
Laryngorhinootologie ; 103(3): 196-206, 2024 03.
Article in German | MEDLINE | ID: mdl-38134907

ABSTRACT

Three forms of peripheral vestibular disorders, each with its typical symptoms and clinical signs, can be differentiated functionally, anatomically and pathophysiologically: 1. inadequate unilateral paroxysmal stimulation or rarely inhibition of the peripheral vestibular system, e. g., BPPV, Menière's disease, vestibular paroxysmia or syndrome of the third mobile windows; 2. acute unilateral vestibulopathy leading to an acute vestibular tone imbalance manifesting as an acute peripheral vestibular syndrome; and 3. loss or impairment of function of the vestibular nerve and/or labyrinth: bilateral vestibulopathy. For all of these diseases, current diagnostic criteria by the Bárány-Society are available with a high clinical and scientific impact, also for clinical trials. The treatment depends on the underlying disease. It basically consists of 5 principles: 1. Explaining the symptoms and signs, pathophysiology, aetiology and treatment options to the patient; this is important for compliance, adherence and persistence. 2. Physical therapy: A) For BPPV specific liberatory maneuvers, depending on canal involved. Posterior canal: The new SémontPLUS maneuver is superior to the regular Sémont and Epley maneuvers; horizontal canal: the modified roll-maneuver; anterior canal the modified Yacovino-maneuver; 3. Symptomatic or causative drug therapy. There is still a deficit of placebo-controlled clinical trials so that the level of evidence for pharmacotherapy is most often low. 4. Surgery, mainly for the syndrome of the third mobile windows. 5. Psychotherapeutic measures for secondary functional dizziness.


Subject(s)
Bilateral Vestibulopathy , Meniere Disease , Vestibular Diseases , Vestibule, Labyrinth , Humans , Vestibular Diseases/diagnosis , Vestibular Diseases/therapy , Vertigo/diagnosis , Vertigo/etiology , Vertigo/therapy , Meniere Disease/diagnosis , Meniere Disease/therapy , Acute Disease
7.
Acta otorrinolaringol. esp ; 73(1): 42-50, feb 2022. tab
Article in Spanish | IBECS | ID: ibc-203220

ABSTRACT

Este artículo describe los criterios diagnósticos de presbivestíbulopatía (PVP) del Comité de Clasificación de la Sociedad Bárány. Se define la PVP como un síndrome vestibular crónico, caracterizado por inestabilidad, alteración de la marcha, y/o caídas recurrentes en presencia de déficits vestibulares bilaterales leves, con hallazgos en las pruebas de función vestibular situados entre la normalidad y los límites establecidos para la vestibulopatía bilateral. El diagnóstico de PVP se basa en la anamnesis, la exploración clínica y la evaluación mediante pruebas de función vestibular. El diagnóstico de PVP requiere una reducción bilateral de la función del reflejo vestíbulo-ocular (RVO). Esta puede ser diagnosticada mediante el video-HIT (vHIT) para el rango de frecuencias altas del RVO; para el rango de frecuencias medias con la prueba de la silla rotatoria; y para el rango de bajas frecuencias mediante la prueba calórica. Para el diagnóstico de PVP, la ganancia de RVO angular horizontal debe ser en ambos lados < 0,8 y > 0,6, y/o la suma de las velocidades pico máximas de fase lenta del nistagmo calórico, tras estimulación con agua caliente y fría en cada lado, que debe estar entre < 25°/s y > 6°/s y/o la ganancia del RVO angular horizontal debe estar entre > 0,1 y < 0,3 con la estimulación sinusoidal en silla rotatoria. La PVP habitualmente ocurre junto con otros déficits relacionados con la edad que afectan a la visión, propiocepción, y/o de las funciones corticales, cerebelosas y extrapiramidales que también contribuyen y que podrían incluso ser necesarias para la manifestación de los síntomas de inestabilidad, alteración de la marcha y caídas. Estos criterios consideran simplemente la presencia de estos síntomas en adultos mayores, junto con el deterioro documentado de la función vestibular. (AU)


This paper describes the diagnostic criteria for presbyvestibulopathy (PVP) of the Classification Committee of the Bárány Society. PVP is defined as a chronic vestibular syndrome characterized by unsteadiness, gait disturbance, and/or recurrent falls in the presence of mild bilateral vestibular deficits, with findings on laboratory tests that are between normal values and the thresholds established for bilateral vestibulopathy. The diagnosis of PVP is based on patient history, bedside examination, and laboratory evaluation. The diagnosis of PVP requires bilaterally reduced function of the vestibulo-ocular reflex (VOR). This can be diagnosed for the high frequency range of the VOR with video-HIT (vHIT), for the middle frequency range with rotary chair testing, and for the low frequency range with caloric testing. For the diagnosis of PVP, the horizontal angular VOR gain on both sides should be < .8 and > .6, and/or the sum of the maximal peak velocities of the slow phase caloric-induced nystagmus for stimulation with warm and cold water on each side should be < 25°/s and > 6°/s, and/or the horizontal angular VOR gain should be > .1 and < .3 upon sinusoidal stimulation on a rotatory chair. PVP typically occurs along with other age-related deficits of vision, proprioception, and/or cortical, cerebellar, and extrapyramidal function which also contribute to and might even be required for symptoms of unsteadiness, gait disturbance, and falls to manifest. These criteria simply consider the presence of these symptoms, along with documented impairment of vestibular function, in older adults. (AU)


Subject(s)
Humans , Young Adult , Adult , Middle Aged , Aged , Aged, 80 and over , Health Sciences , Diagnostic Techniques and Procedures , Bilateral Vestibulopathy , Aging
8.
Acta otorrinolaringol. esp ; 67(1): 1-7, ene.-feb. 2016. tab
Article in Spanish | IBECS | ID: ibc-148951

ABSTRACT

Este trabajo presenta los criterios diagnósticos de enfermedad de Menière elaborados de forma conjunta por el Comité de Clasificación de los Trastornos Vestibulares de la Bárány Society, la Japan Society for Equilibrium Research, la European Academy of Otology and Neurotology (EAONO), el Comité de Equilibrio de American Academy of Otolaryngology-Head and Neck Surgery (AAO-HNS) y la Korean Balance Society. La clasificación establece 2 categorías: enfermedad de Menière definida y enfermedad de Menière probable. El diagnóstico de enfermedad de Menière definida se basa en criterios clínicos y requiere la observación de un síndrome vestibular episódico asociado con hipoacusia neurosensorial de frecuencias bajas y medias y síntomas auditivos fluctuantes (hipoacusia, acúfenos o plenitud ótica) en el oído afectado. La duración de los episodios de vértigo se limita a un período entre 20 min y 12 h. La enfermedad de Menière probable es un concepto más amplio definido por síntomas vestibulares episódicos (vértigo o mareo) asociados a síntomas auditivos fluctuantes que ocurren en un periodo entre 20 min y 24 h (AU)


This paper presents diagnostic criteria for Menière's disease jointly formulated by the Classification Committee of the Bárány Society, The Japan Society for Equilibrium Research, the European Academy of Otology and Neurotology (EAONO), the Equilibrium Committee of the American Academy of Otolaryngology-Head and Neck Surgery (AAO-HNS) and the Korean Balance Society. The classification includes 2 categories: definite Menière's disease and probable Menière's disease. The diagnosis of definite Menière's disease is based on clinical criteria and requires the observation of an episodic vertigo syndrome associated with low-to medium-frequency sensorineural hearing loss and fluctuating aural symptoms (hearing, tinnitus and/or fullness) in the affected ear. Duration of vertigo episodes is limited to a period between 20 min and 12 h. Probable Menière's disease is a broader concept defined by episodic vestibular symptoms (vertigo or dizziness) associated with fluctuating aural symptoms occurring in a period from 20 min to 24 h (AU)


Subject(s)
Humans , Male , Female , Meniere Disease/diagnosis , Meniere Disease/genetics , Meniere Disease/epidemiology , Vertigo/diagnosis , Hearing Loss, Unilateral/diagnosis , Hearing Loss, Sensorineural/diagnosis , Migraine Disorders/diagnosis , Vestibular Diseases/diagnosis , Endolymphatic Hydrops/diagnosis , Diagnosis, Differential , Consensus Development Conferences as Topic , Societies, Medical
SELECTION OF CITATIONS
SEARCH DETAIL