Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Sci Rep ; 13(1): 18600, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37903840

ABSTRACT

Familial dysautonomia (FD) is a rare neurodevelopmental and neurodegenerative disease caused by a splicing mutation in the Elongator Acetyltransferase Complex Subunit 1 (ELP1) gene. The reduction in ELP1 mRNA and protein leads to the death of retinal ganglion cells (RGCs) and visual impairment in all FD patients. Currently patient symptoms are managed, but there is no treatment for the disease. We sought to test the hypothesis that restoring levels of Elp1 would thwart the death of RGCs in FD. To this end, we tested the effectiveness of two therapeutic strategies for rescuing RGCs. Here we provide proof-of-concept data that gene replacement therapy and small molecule splicing modifiers effectively reduce the death of RGCs in mouse models for FD and provide pre-clinical foundational data for translation to FD patients.


Subject(s)
Dysautonomia, Familial , Neurodegenerative Diseases , Mice , Animals , Humans , Retinal Ganglion Cells/metabolism , Dysautonomia, Familial/genetics , Dysautonomia, Familial/therapy , Dysautonomia, Familial/metabolism , Neurodegenerative Diseases/metabolism , RNA Splicing , Genetic Therapy , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
2.
Am J Hum Genet ; 104(4): 638-650, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905397

ABSTRACT

Familial dysautonomia (FD) is a recessive neurodegenerative disease caused by a splice mutation in Elongator complex protein 1 (ELP1, also known as IKBKAP); this mutation leads to variable skipping of exon 20 and to a drastic reduction of ELP1 in the nervous system. Clinically, many of the debilitating aspects of the disease are related to a progressive loss of proprioception; this loss leads to severe gait ataxia, spinal deformities, and respiratory insufficiency due to neuromuscular incoordination. There is currently no effective treatment for FD, and the disease is ultimately fatal. The development of a drug that targets the underlying molecular defect provides hope that the drastic peripheral neurodegeneration characteristic of FD can be halted. We demonstrate herein that the FD mouse TgFD9;IkbkapΔ20/flox recapitulates the proprioceptive impairment observed in individuals with FD, and we provide the in vivo evidence that postnatal correction, promoted by the small molecule kinetin, of the mutant ELP1 splicing can rescue neurological phenotypes in FD. Daily administration of kinetin starting at birth improves sensory-motor coordination and prevents the onset of spinal abnormalities by stopping the loss of proprioceptive neurons. These phenotypic improvements correlate with increased amounts of full-length ELP1 mRNA and protein in multiple tissues, including in the peripheral nervous system (PNS). Our results show that postnatal correction of the underlying ELP1 splicing defect can rescue devastating disease phenotypes and is therefore a viable therapeutic approach for persons with FD.


Subject(s)
Dysautonomia, Familial/therapy , Kinetin/therapeutic use , Proprioception , RNA Splicing , Transcriptional Elongation Factors/genetics , Alleles , Animals , Behavior, Animal , Cell Line , Crosses, Genetic , Disease Models, Animal , Dysautonomia, Familial/genetics , Exons , Fibroblasts , Genotype , Humans , Introns , Kinetin/genetics , Male , Mice , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Phenotype
4.
Nucleic Acids Res ; 46(10): 4833-4844, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29672717

ABSTRACT

Familial dysautonomia (FD) is a rare inherited neurodegenerative disorder caused by a point mutation in the IKBKAP gene that results in defective splicing of its pre-mRNA. The mutation weakens the 5' splice site of exon 20, causing this exon to be skipped, thereby introducing a premature termination codon. Though detailed FD pathogenesis mechanisms are not yet clear, correcting the splicing defect in the relevant tissue(s), thus restoring normal expression levels of the full-length IKAP protein, could be therapeutic. Splice-switching antisense oligonucleotides (ASOs) can be effective targeted therapeutics for neurodegenerative diseases, such as nusinersen (Spinraza), an approved drug for spinal muscular atrophy. Using a two-step screen with ASOs targeting IKBKAP exon 20 or the adjoining intronic regions, we identified a lead ASO that fully restored exon 20 splicing in FD patient fibroblasts. We also characterized the corresponding cis-acting regulatory sequences that control exon 20 splicing. When administered into a transgenic FD mouse model, the lead ASO promoted expression of full-length human IKBKAP mRNA and IKAP protein levels in several tissues tested, including the central nervous system. These findings provide insights into the mechanisms of IKBKAP exon 20 recognition, and pre-clinical proof of concept for an ASO-based targeted therapy for FD.


Subject(s)
Carrier Proteins/genetics , Dysautonomia, Familial/genetics , Dysautonomia, Familial/therapy , Oligonucleotides, Antisense/pharmacology , Animals , Carrier Proteins/metabolism , Cells, Cultured , Dysautonomia, Familial/pathology , Enhancer Elements, Genetic , Exons , Fibroblasts , Humans , Mice, Transgenic , Oligonucleotides, Antisense/chemistry , RNA Splice Sites , RNA Splicing , Transcriptional Elongation Factors
5.
Hum Mol Genet ; 27(14): 2466-2476, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29701768

ABSTRACT

Familial dysautonomia (FD) is a rare genetic disease with no treatment, caused by an intronic point mutation (c.2204+6T>C) that negatively affects the definition of exon 20 in the elongator complex protein 1 gene (ELP1 also known as IKBKAP). This substitution modifies the 5' splice site and, in combination with regulatory splicing factors, induces different levels of exon 20 skipping, in various tissues. Here, we evaluated the therapeutic potential of a novel class of U1 snRNA molecules, exon-specific U1s (ExSpeU1s), in correcting ELP1 exon 20 recognition. Lentivirus-mediated expression of ELP1-ExSpeU1 in FD fibroblasts improved ELP1 splicing and protein levels. We next focused on a transgenic mouse model that recapitulates the same tissue-specific mis-splicing seen in FD patients. Intraperitoneal delivery of ELP1-ExSpeU1s-adeno-associated virus particles successfully increased the production of full-length human ELP1 transcript and protein. This splice-switching class of molecules is the first to specifically correct the ELP1 exon 20 splicing defect. Our data provide proof of principle of ExSpeU1s-adeno-associated virus particles as a novel therapeutic strategy for FD.


Subject(s)
Carrier Proteins/genetics , Dysautonomia, Familial/therapy , Genetic Therapy , RNA, Small Nuclear/genetics , Alternative Splicing/genetics , Animals , Carrier Proteins/therapeutic use , Dependovirus/genetics , Disease Models, Animal , Dysautonomia, Familial/genetics , Dysautonomia, Familial/physiopathology , Exons/genetics , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins , Introns/genetics , Mice , Mice, Transgenic , RNA Splicing/genetics , RNA, Small Nuclear/therapeutic use , Transcriptional Elongation Factors
6.
Clin Auton Res ; 27(4): 235-243, 2017 08.
Article in English | MEDLINE | ID: mdl-28667575

ABSTRACT

Since Riley and Day first described the clinical phenotype of patients with familial dysautonomia (FD) over 60 years ago, the field has made considerable progress clinically, scientifically, and translationally in treating and understanding the etiology of FD. FD is classified as a hereditary sensory and autonomic neuropathy (HSAN type III) and is both a developmental and a progressive neurodegenerative condition that results from an autosomal recessive mutation in the gene IKBKAP, also known as ELP1. FD primarily impacts the peripheral nervous system but also manifests in central nervous system disruption, especially in the retina and optic nerve. While the disease is rare, the rapid progress being made in elucidating the molecular and cellular mechanisms mediating the demise of neurons in FD should provide insight into degenerative pathways common to many neurological disorders. Interestingly, the protein encoded by IKBKAP/ELP1, IKAP or ELP1, is a key scaffolding subunit of the six-subunit Elongator complex, and variants in other Elongator genes are associated with amyotrophic lateral sclerosis (ALS), intellectual disability, and Rolandic epilepsy. Here we review the recent model systems that are revealing the molecular and cellular pathophysiological mechanisms mediating FD. These powerful model systems can now be used to test targeted therapeutics for mitigating neuronal loss in FD and potentially other disorders.


Subject(s)
Disease Models, Animal , Dysautonomia, Familial/pathology , Stem Cells/physiology , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/therapy , Humans , Mice
7.
Odontol. pediatr. (Lima) ; 14(1): 58-61, ene.-jul.2015. ilus
Article in Spanish | LILACS, LIPECS | ID: lil-790456

ABSTRACT

Riley Day es una rara enfermedad o transtorno hereditario que afecta el desarrollo y funcionamiento de los nervios. Ocasiona Insensibilidad al dolor, no tiene cura ni tratamiento paliativo. Se transmite de padres a hijos, la persona debe heredar una copia del gen defectuoso de cada uno de los padres para desarrollar la afecci6n. El odontopediatra debe de estar capacitado para reconocer esta enfermedad, siendo un transtorno hereditario las primeras manifestaciones clínicas y orales se van a dar des de la infancia y van a ir aumentando con la edad; por eso el pro- pósito del presente reporte es describir el caso de un niño de 2 años 4 meses con Riley day, sus manifestaciones clínicas, datos epidemio16gicos y sugerencias de manejo odontologico de los pacientes con esta enfermedad...


Riley Day is a rare hereditary disease or disorder that affects the development and functioning of nerves. It causes insensitivity to pain, has no cure or treatment paliativo. It is transmitted from parent to child, the person must inherit a defective copy of the gene from each parent to develop afeccion. The dentist should be able to recognize this disease, an inherited disorder being the first clinical and oral manifestations are to be given since childhood and will gradually increase with age; therefore the purpose of this report is to describe the case of a 2 years 4 months Riley Day, its clinical manifestations, oral manifestations, epidemiological data and suggestions for dental management of patients with this disease....


Subject(s)
Humans , Male , Child, Preschool , Dysautonomia, Familial , Dysautonomia, Familial/diagnosis , Dysautonomia, Familial/therapy , Pediatric Dentistry
8.
J Clin Gastroenterol ; 47(2): 136-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22739220

ABSTRACT

GOALS: To analyze the neurochemical profile during the recurrent attacks of nausea and vomiting in patients with Riley-day syndrome. BACKGROUND: One of the most disabling features of patients with Riley-day syndrome are recurrent attacks of severe nausea/retching/vomiting accompanied by hypertension, tachycardia, and skin flushing, usually triggered by emotional or other stresses. STUDY: We monitored blood pressure and heart rate and measured plasma catecholamines during typical dysautonomic crises triggered by emotionally charged situations. For comparison, measurements were repeated at follow-up after the symptoms had resolved and the patients were feeling calm and well. RESULTS: During a typical attack, patients were hypertensive and tachycardic. In all patients, circulating levels of norepinephrine (P < 0.002) and dopamine (P < 0.007) increased significantly. CONCLUSIONS: Activation of dopamine receptors in the chemoreceptor trigger zone may explain the cyclic nausea/retching/vomiting of patients with Riley-day syndrome.


Subject(s)
Dopamine/blood , Dysautonomia, Familial/complications , Sympathetic Nervous System/metabolism , Vomiting/etiology , Adolescent , Adult , Blood Pressure , Child, Preschool , Dysautonomia, Familial/blood , Dysautonomia, Familial/physiopathology , Dysautonomia, Familial/psychology , Dysautonomia, Familial/therapy , Emotions , Female , Heart Rate , Humans , Hypertension/blood , Hypertension/etiology , Hypertension/physiopathology , Male , Norepinephrine/blood , Recurrence , Sympathetic Nervous System/physiopathology , Tachycardia/blood , Tachycardia/etiology , Tachycardia/physiopathology , Time Factors , Up-Regulation , Vomiting/blood , Vomiting/physiopathology , Vomiting/psychology , Vomiting/therapy , Young Adult
9.
Arq. bras. ciênc. saúde ; 35(1)jan.-abr. 2010.
Article in Portuguese | LILACS | ID: lil-549830

ABSTRACT

Os autores apresentam dois irmãos com diagnóstico de analgesia congênita, com suas características clínicas e evolução. Essa doença é rara, apresenta alta morbidade e gera complicações osteoarticulares de difícil solução. O objetivo dos autores foi ressaltar a importância do diagnóstico tanto para o tratamento de suas afecções secundárias, quanto para seu aspecto jurídico.


The authors present two brothers with congenital pain insensitivity, with their clinical characteristics and evolution. This disease is rare, has high morbidity and originates complex osteoarticular complications. The aim of the authors was to emphasize the value of the diagnosis for a better treatment and to avoid legal problems to the parents.


Subject(s)
Humans , Male , Child, Preschool , Child , Dysautonomia, Familial/diagnosis , Dysautonomia, Familial/therapy , Pain Insensitivity, Congenital/diagnosis , Pain Insensitivity, Congenital/therapy , Orthopedics
10.
J Anesth ; 23(4): 579-82, 2009.
Article in English | MEDLINE | ID: mdl-19921370

ABSTRACT

Familial dysautonomia is an inherited disorder characterized by autonomic and sensory nervous system neuropathy resulting in extremely labile blood pressure (severe hypertension followed by hypotension). As more patients with familial dysautonomia reach adulthood due to improved medical treatment, perioperative encounters of patients with familial dysautonomia will increase. This report is the first adult case to describe an anesthetic management of kidney transplantation for an adult familial dysautonomia patient. The clinical manifestations of this disease and rationale of our anesthetic management are discussed.


Subject(s)
Anesthesia , Dysautonomia, Familial/complications , Kidney Transplantation , Adult , Blood Pressure/physiology , Dysautonomia, Familial/therapy , Humans , Male , Monitoring, Intraoperative , Perioperative Care , Postoperative Care
11.
Nature ; 461(7262): 402-6, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19693009

ABSTRACT

The isolation of human induced pluripotent stem cells (iPSCs) offers a new strategy for modelling human disease. Recent studies have reported the derivation and differentiation of disease-specific human iPSCs. However, a key challenge in the field is the demonstration of disease-related phenotypes and the ability to model pathogenesis and treatment of disease in iPSCs. Familial dysautonomia (FD) is a rare but fatal peripheral neuropathy, caused by a point mutation in the IKBKAP gene involved in transcriptional elongation. The disease is characterized by the depletion of autonomic and sensory neurons. The specificity to the peripheral nervous system and the mechanism of neuron loss in FD are poorly understood owing to the lack of an appropriate model system. Here we report the derivation of patient-specific FD-iPSCs and the directed differentiation into cells of all three germ layers including peripheral neurons. Gene expression analysis in purified FD-iPSC-derived lineages demonstrates tissue-specific mis-splicing of IKBKAP in vitro. Patient-specific neural crest precursors express particularly low levels of normal IKBKAP transcript, suggesting a mechanism for disease specificity. FD pathogenesis is further characterized by transcriptome analysis and cell-based assays revealing marked defects in neurogenic differentiation and migration behaviour. Furthermore, we use FD-iPSCs for validating the potency of candidate drugs in reversing aberrant splicing and ameliorating neuronal differentiation and migration. Our study illustrates the promise of iPSC technology for gaining new insights into human disease pathogenesis and treatment.


Subject(s)
Dysautonomia, Familial/pathology , Dysautonomia, Familial/therapy , Models, Biological , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Adolescent , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Carrier Proteins/genetics , Cell Dedifferentiation , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Child , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Kinetin/pharmacology , Kinetin/therapeutic use , Male , Mice , Neural Crest/cytology , Neural Crest/drug effects , Organ Specificity , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Transcriptional Elongation Factors
12.
Neuromolecular Med ; 10(3): 148-56, 2008.
Article in English | MEDLINE | ID: mdl-17985250

ABSTRACT

Familial dysautonomia (FD) is a sensory and autonomic neuropathy that affects the development and survival of sensory, sympathetic, and some parasympathetic neurons. It is autosomally inherited and occurs almost exclusively among individuals of Ashkenazi Jewish descent. The pathological and clinical manifestations of FD have been extensively studied and therapeutic modalities have, until recently, focused primarily on addressing the symptoms experienced by those with this fatal disorder. The primary FD-causing mutation is an intronic nucleotide substitution that alters the splicing of the IKBKAP-derived transcript. Recent efforts have resulted in the development of new therapeutic modalities that facilitate the increased production of the correctly spliced transcript and mitigate the symptoms of those with FD. Furthermore, the recent demonstration of the reduced presence of monoamine oxidase A in cells and tissues of individuals with FD has provided new insight into the cause of hypertensive crises experienced by these patients.


Subject(s)
Dysautonomia, Familial/genetics , Amino Acid Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Catecholamines/metabolism , Dysautonomia, Familial/physiopathology , Dysautonomia, Familial/therapy , Humans , Molecular Sequence Data , Monoamine Oxidase/deficiency , Mutation , RNA Splicing , Transcriptional Elongation Factors
14.
Acta Paediatr ; 95(4): 457-62, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16720494

ABSTRACT

AIM: To evaluate the quality of life (QoL) of children, adolescents, and adults treated for familial dysautonomia (FD), a pervasive neurological disorder. METHODS: The Child Health Questionnaire was completed by parents of 71 patients, while an additional 74 patients completed the Short Form--36. RESULTS: FD imposed a greater physical than psychosocial burden on the child, while the young adults reported both mental and physical quality of life within the average range. Self-esteem was problematic and improved with age, while both groups reported lowering physical quality of life as they grew older, with worsening general health that limited their role at school or work. CONCLUSION: Younger FD patients should be closely monitored for lowered self-esteem and referred for counseling when appropriate, while physical and occupational therapy should be provided in advance of expected lowered physical QoL and role fulfillment with increasing age. This becomes important as the need for additional surgical interventions, such as fundoplication with gastrostomy or spinal fusion, contribute to lower physical functioning. Given the high degree of parental involvement required for the varied manifestations of this multisystem disorder, the need for continued parental assessment and psycho-education about this chronic medical illness is warranted.


Subject(s)
Dysautonomia, Familial/physiopathology , Dysautonomia, Familial/psychology , Quality of Life/psychology , Adolescent , Adult , Age Factors , Child , Child, Preschool , Dysautonomia, Familial/therapy , Female , Health Status , Humans , Male , Mental Health , Middle Aged , Parent-Child Relations , Personal Satisfaction , Self Concept , Sex Factors
16.
Clin Auton Res ; 15(1): 15-20, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15768197

ABSTRACT

Familial dysautonomia (FD) is a genetic disease associated with a high incidence of sudden death. If fatal bradyarrhythmia is an etiological factor then the incidence of sudden death should decrease after pacemaker placement. Retrospective review of 596 registered FD patients revealed that 22 FD patients (3.7%) had pacemakers placed between December 1984 and June 2003. Clinical and electrocardiographic indications for placement and demographic data were assessed for 20 of the 22 patients (10 males, 10 females, ages 4 to 48 years). Two patients were excluded because of insufficient data. Prior to pacemaker placement, presenting symptoms were syncope and cardiac arrest, 16/20 (80%) and 6/20 (30 %), respectively. Asystole was the most frequent electrocardiographic finding and was documented in 17/20 patients (85 %). Other electrocardiographic abnormalities included bradycardia, AV block, prolonged QTc and prolonged JTc. The average duration of pacemaker utilization was 5.7 years (range 5 months to 14.5 years). Complications included infection (1 patient) and wire migration (2 patients). In the one patient with infection, the pacemaker was permanently removed. This patient then experienced multiple syncopal episodes and death. There were 7 other deaths. Three deaths occurred suddenly without preceding events, and 4 patients had non-cardiac causes of death. None of these 7 deceased patients had recurrence of syncope after pacemaker placement. In the 12 surviving patients, 6 had recurrence of syncope but none had cardiac arrest. Pacemaker placement may protect FD patients from fatal bradyarrhythmia and may decrease the incidence of syncope. However, data are limited and prospective analysis is needed.


Subject(s)
Arrhythmias, Cardiac/therapy , Death, Sudden, Cardiac/prevention & control , Dysautonomia, Familial/therapy , Pacemaker, Artificial , Arrhythmias, Cardiac/mortality , Dysautonomia, Familial/mortality , Humans
17.
Epilepsia ; 45(11): 1461-2, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509249

ABSTRACT

PURPOSE: Video-EEG in a family of three patients with slow development and familial dysautonomia demonstrated absence seizures associated with 3-Hz generalized spike-and-wave discharges. The seizures were refractory to antiepileptic drugs (AEDs). METHODS: Treatment was given with rice-based cereal electrolyte oral solution. RESULTS: Treatment induced seizure freedom and normalization of EEG in all three patients. Repeated video-EEG monitoring with discontinuation of AEDs and maintenance of the oral hydration therapy was associated with recurrence of epileptic activity. All three patients have remained seizure free (approximately 1 year) with a combination of topiramate and electrolytic therapy. CONCLUSIONS: Rice-based oral electrolyte hydration therapy may play a role in prevention and control of seizures in patients with familial dysautonomia.


Subject(s)
Dysautonomia, Familial/therapy , Electrolytes/therapeutic use , Epilepsy/therapy , Rehydration Solutions/administration & dosage , Anticonvulsants/therapeutic use , Carbohydrates/administration & dosage , Citrates/administration & dosage , Comorbidity , Dysautonomia, Familial/epidemiology , Electrolytes/administration & dosage , Epilepsy/drug therapy , Epilepsy/epidemiology , Humans , Oryza , Potassium/administration & dosage , Sodium/administration & dosage , Treatment Outcome
18.
J Neurol Neurosurg Psychiatry ; 73(3): 299-302, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185162

ABSTRACT

OBJECTIVES: To use the technique of dermal microdialysis to examine sensitivity of skin vessels to noradrenaline (NA) in patients with familial dysautonomia (FD) and in healthy controls. METHODS: In 14 patients with FD and 12 healthy controls, plasma extravasation, local laser Doppler blood flow, and skin blanching were observed before, during, and after application of 10(-6) M NA through a microdialysis membrane, located intradermally in the skin of the lower leg. RESULTS: Maximum local vasoconstriction measured by laser Doppler blood flow did not differ between patients with FD and controls. In contrast, patients with FD had an earlier onset of vasoconstriction (p = 0.02). Moreover, reaction to NA was more prominent and prolonged in FD, shown by a larger zone of skin blanching around the microdialysis membrane (p < 0.001) and delayed reduction of the protein content in the dialysate after termination of NA application (p = 0.03). CONCLUSION: These data support the hypothesis that peripheral blood vessels of patients with FD show a denervation hypersensitivity to catecholamines. This may be one mechanism contributing to the major hypertension that frequently occurs during "dysautonomic crises" in FD.


Subject(s)
Drug Hypersensitivity/etiology , Dysautonomia, Familial/therapy , Membranes, Artificial , Microdialysis/instrumentation , Norepinephrine/adverse effects , Administration, Cutaneous , Adolescent , Adult , Blood Flow Velocity/physiology , Drug Hypersensitivity/diagnosis , Equipment Design , Female , Humans , Laser-Doppler Flowmetry , Leg/blood supply , Male , Norepinephrine/administration & dosage , Proteins/metabolism , Skin/blood supply , Skin/metabolism , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL
...