Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 79
1.
PLoS One ; 19(6): e0298965, 2024.
Article En | MEDLINE | ID: mdl-38829854

Familial Dysautonomia (FD) is a rare disease caused by ELP1 exon 20 skipping. Here we clarify the role of RNA Polymerase II (RNAPII) and chromatin on this splicing event. A slow RNAPII mutant and chromatin-modifying chemicals that reduce the rate of RNAPII elongation induce exon skipping whereas chemicals that create a more relaxed chromatin exon inclusion. In the brain of a mouse transgenic for the human FD-ELP1 we observed on this gene an age-dependent decrease in the RNAPII density profile that was most pronounced on the alternative exon, a robust increase in the repressive marks H3K27me3 and H3K9me3 and a decrease of H3K27Ac, together with a progressive reduction in ELP1 exon 20 inclusion level. In HEK 293T cells, selective drug-induced demethylation of H3K27 increased RNAPII elongation on ELP1 and SMN2, promoted the inclusion of the corresponding alternative exons, and, by RNA-sequencing analysis, induced changes in several alternative splicing events. These data suggest a co-transcriptional model of splicing regulation in which age-dependent changes in H3K27me3/Ac modify the rate of RNAPII elongation and affect processing of ELP1 alternative exon 20.


Alternative Splicing , Chromatin , Dysautonomia, Familial , Exons , RNA Polymerase II , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Humans , Exons/genetics , Animals , Chromatin/metabolism , Chromatin/genetics , Mice , HEK293 Cells , Histones/metabolism , Mice, Transgenic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , Kinetics , RNA Splicing , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
2.
Sci Rep ; 14(1): 570, 2024 01 04.
Article En | MEDLINE | ID: mdl-38177237

Familial dysautonomia (FD) is a rare recessive neurodevelopmental disease caused by a splice mutation in the Elongator acetyltransferase complex subunit 1 (ELP1) gene. This mutation results in a tissue-specific reduction of ELP1 protein, with the lowest levels in the central and peripheral nervous systems (CNS and PNS, respectively). FD patients exhibit complex neurological phenotypes due to the loss of sensory and autonomic neurons. Disease symptoms include decreased pain and temperature perception, impaired or absent myotatic reflexes, proprioceptive ataxia, and progressive retinal degeneration. While the involvement of the PNS in FD pathogenesis has been clearly recognized, the underlying mechanisms responsible for the preferential neuronal loss remain unknown. In this study, we aimed to elucidate the molecular mechanisms underlying FD by conducting a comprehensive transcriptome analysis of neuronal tissues from the phenotypic mouse model TgFD9; Elp1Δ20/flox. This mouse recapitulates the same tissue-specific ELP1 mis-splicing observed in patients while modeling many of the disease manifestations. Comparison of FD and control transcriptomes from dorsal root ganglion (DRG), trigeminal ganglion (TG), medulla (MED), cortex, and spinal cord (SC) showed significantly more differentially expressed genes (DEGs) in the PNS than the CNS. We then identified genes that were tightly co-expressed and functionally dependent on the level of full-length ELP1 transcript. These genes, defined as ELP1 dose-responsive genes, were combined with the DEGs to generate tissue-specific dysregulated FD signature genes and networks. Within the PNS networks, we observed direct connections between Elp1 and genes involved in tRNA synthesis and genes related to amine metabolism and synaptic signaling. Importantly, transcriptomic dysregulation in PNS tissues exhibited enrichment for neuronal subtype markers associated with peptidergic nociceptors and myelinated sensory neurons, which are known to be affected in FD. In summary, this study has identified critical tissue-specific gene networks underlying the etiology of FD and provides new insights into the molecular basis of the disease.


Dysautonomia, Familial , Humans , Mice , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Carrier Proteins/metabolism , Peripheral Nervous System/metabolism , Sensory Receptor Cells/metabolism , Gene Expression Profiling , Gene Expression
3.
Sci Rep ; 13(1): 18600, 2023 10 30.
Article En | MEDLINE | ID: mdl-37903840

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.


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
4.
Nat Commun ; 13(1): 7032, 2022 11 17.
Article En | MEDLINE | ID: mdl-36396637

Familial dysautonomia (FD), a rare neurodevelopmental and neurodegenerative disorder affects the sympathetic and sensory nervous system. Although almost all patients harbor a mutation in ELP1, it remains unresolved exactly how function of sympathetic neurons (symNs) is affected; knowledge critical for understanding debilitating disease hallmarks, including cardiovascular instability or dysautonomic crises, that result from dysregulated sympathetic activity. Here, we employ the human pluripotent stem cell (hPSC) system to understand symN disease mechanisms and test candidate drugs. FD symNs are intrinsically hyperactive in vitro, in cardiomyocyte co-cultures, and in animal models. We report reduced norepinephrine transporter expression, decreased intracellular norepinephrine (NE), decreased NE re-uptake, and excessive extracellular NE in FD symNs. SymN hyperactivity is not a direct ELP1 mutation result, but may connect to NET via RAB proteins. We found that candidate drugs lowered hyperactivity independent of ELP1 modulation. Our findings may have implications for other symN disorders and may allow future drug testing and discovery.


Dysautonomia, Familial , Animals , Humans , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Norepinephrine Plasma Membrane Transport Proteins/genetics , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Neurons/metabolism , Norepinephrine/metabolism , Mutation
5.
Elife ; 112022 06 17.
Article En | MEDLINE | ID: mdl-35713404

Familial dysautonomia (FD) is a sensory and autonomic neuropathy caused by mutations in elongator complex protein 1 (ELP1). FD patients have small trigeminal nerves and impaired facial pain and temperature perception. These signals are relayed by nociceptive neurons in the trigeminal ganglion, a structure that is composed of both neural crest- and placode-derived cells. Mice lacking Elp1 in neural crest derivatives ('Elp1 CKO') are born with small trigeminal ganglia, suggesting Elp1 is important for trigeminal ganglion development, yet the function of Elp1 in this context is unknown. We demonstrate that Elp1, expressed in both neural crest- and placode-derived neurons, is not required for initial trigeminal ganglion formation. However, Elp1 CKO trigeminal neurons exhibit abnormal axon outgrowth and deficient target innervation. Developing nociceptors expressing the receptor TrkA undergo early apoptosis in Elp1 CKO, while TrkB- and TrkC-expressing neurons are spared, indicating Elp1 supports the target innervation and survival of trigeminal nociceptors. Furthermore, we demonstrate that specific TrkA deficits in the Elp1 CKO trigeminal ganglion reflect the neural crest lineage of most TrkA neurons versus the placodal lineage of most TrkB and TrkC neurons. Altogether, these findings explain defects in cranial gangliogenesis that may lead to loss of facial pain and temperature sensation in FD.


Dysautonomia, Familial , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Facial Pain/metabolism , Mice , Neural Crest/metabolism , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Trigeminal Ganglion
6.
Hum Mol Genet ; 31(20): 3521-3538, 2022 10 10.
Article En | MEDLINE | ID: mdl-35708500

Recent research on familial dysautonomia (FD) has focused on the development of therapeutics that facilitate the production of the correctly spliced, exon 20-containing, transcript in cells and individuals bearing the splice-altering, FD-causing mutation in the elongator acetyltransferase complex subunit I (ELP1) gene. We report here the ability of carnosol, a diterpene present in plant species of the Lamiaceae family, including rosemary, to enhance the cellular presence of the correctly spliced ELP1 transcript in FD patient-derived fibroblasts by upregulating transcription of the ELP1 gene and correcting the aberrant splicing of the ELP1 transcript. Carnosol treatment also elevates the level of the RNA binding motif protein 24 (RBM24) and RNA binding motif protein 38 (RBM38) proteins, two multifunctional RNA-binding proteins. Transfection-mediated expression of either of these RNA binding motif (RBMs) facilitates the inclusion of exon 20 sequence into the transcript generated from a minigene-bearing ELP1 genomic sequence containing the FD-causing mutation. Suppression of the carnosol-mediated induction of either of these RBMs, using targeting siRNAs, limited the carnosol-mediated inclusion of the ELP1 exon 20 sequence. Carnosol treatment of FD patient peripheral blood mononuclear cells facilitates the inclusion of exon 20 into the ELP1 transcript. The increased levels of the ELP1 and RBM38 transcripts and the alternative splicing of the sirtuin 2 (SIRT2) transcript, a sentinel for exon 20 inclusion in the FD-derived ELP1 transcript, are observed in RNA isolated from whole blood of healthy adults following the ingestion of carnosol-containing rosemary extract. These findings and the excellent safety profile of rosemary together justify an expedited clinical study of the impact of carnosol on the FD patient population.


Dysautonomia, Familial , Rosmarinus , Transcriptional Elongation Factors/metabolism , Abietanes/pharmacology , Acetyltransferases , Adult , Carrier Proteins/genetics , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Humans , Leukocytes, Mononuclear/metabolism , RNA , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rosmarinus/genetics , Rosmarinus/metabolism , Sirtuin 2/metabolism , Transcriptional Elongation Factors/genetics
7.
J Genet Genomics ; 49(7): 654-665, 2022 07.
Article En | MEDLINE | ID: mdl-34896608

Familial dysautonomia (FD), a hereditary sensory and autonomic neuropathy, is caused by a mutation in the Elongator complex protein 1 (ELP1) gene that leads to a tissue-specific reduction of ELP1 protein. Our work to generate a phenotypic mouse model for FD headed to the discovery that homozygous deletion of the mouse Elp1 gene leads to embryonic lethality prior to mid-gestation. Given that FD is caused by a reduction, not loss, of ELP1, we generated two new mouse models by introducing different copy numbers of the human FD ELP1 transgene into the Elp1 knockout mouse (Elp1-/-) and observed that human ELP1 expression rescues embryonic development in a dose-dependent manner. We then conducted a comprehensive transcriptome analysis in mouse embryos to identify genes and pathways whose expression correlates with the amount of ELP1. We found that ELP1 is essential for the expression of genes responsible for nervous system development. Further, gene length analysis of the differentially expressed genes showed that the loss of Elp1 mainly impacts the expression of long genes and that by gradually restoring Elongator, their expression is progressively rescued. Finally, through evaluation of co-expression modules, we identified gene sets with unique expression patterns that depended on ELP1 expression.


Carrier Proteins , Dysautonomia, Familial , Animals , Carrier Proteins/genetics , Disease Models, Animal , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Gene Expression , Homozygote , Humans , Mice , Sequence Deletion
8.
Nat Commun ; 12(1): 5878, 2021 10 07.
Article En | MEDLINE | ID: mdl-34620845

Microtubule (MT)-based transport is an evolutionary conserved process finely tuned by posttranslational modifications. Among them, α-tubulin acetylation, primarily catalyzed by a vesicular pool of α-tubulin N-acetyltransferase 1 (Atat1), promotes the recruitment and processivity of molecular motors along MT tracks. However, the mechanism that controls Atat1 activity remains poorly understood. Here, we show that ATP-citrate lyase (Acly) is enriched in vesicles and provide Acetyl-Coenzyme-A (Acetyl-CoA) to Atat1. In addition, we showed that Acly expression is reduced upon loss of Elongator activity, further connecting Elongator to Atat1 in a pathway regulating α-tubulin acetylation and MT-dependent transport in projection neurons, across species. Remarkably, comparable defects occur in fibroblasts from Familial Dysautonomia (FD) patients bearing an autosomal recessive mutation in the gene coding for the Elongator subunit ELP1. Our data may thus shine light on the pathophysiological mechanisms underlying FD.


ATP Citrate (pro-S)-Lyase/metabolism , Axonal Transport/physiology , ATP Citrate (pro-S)-Lyase/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Acetyltransferases/genetics , Animals , Axonal Transport/genetics , Drosophila melanogaster , Dysautonomia, Familial/metabolism , Female , Fibroblasts/metabolism , Humans , Larva , Male , Mice , Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/metabolism
9.
Nat Commun ; 12(1): 4507, 2021 07 23.
Article En | MEDLINE | ID: mdl-34301951

Approximately half of genetic disease-associated mutations cause aberrant splicing. However, a widely applicable therapeutic strategy to splicing diseases is yet to be developed. Here, we analyze the mechanism whereby IKBKAP-familial dysautonomia (FD) exon 20 inclusion is specifically promoted by a small molecule splice modulator, RECTAS, even though IKBKAP-FD exon 20 has a suboptimal 5' splice site due to the IVS20 + 6 T > C mutation. Knockdown experiments reveal that exon 20 inclusion is suppressed in the absence of serine/arginine-rich splicing factor 6 (SRSF6) binding to an intronic splicing enhancer in intron 20. We show that RECTAS directly interacts with CDC-like kinases (CLKs) and enhances SRSF6 phosphorylation. Consistently, exon 20 splicing is bidirectionally manipulated by targeting cellular CLK activity with RECTAS versus CLK inhibitors. The therapeutic potential of RECTAS is validated in multiple FD disease models. Our study indicates that small synthetic molecules affecting phosphorylation state of SRSFs is available as a new therapeutic modality for mechanism-oriented precision medicine of splicing diseases.


Alternative Splicing/genetics , Dysautonomia, Familial/genetics , Mutation , Transcriptional Elongation Factors/genetics , Alternative Splicing/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/metabolism , Enhancer Elements, Genetic/genetics , Exons/genetics , HeLa Cells , Humans , Introns/genetics , Mice, Transgenic , Molecular Structure , Phosphoproteins/metabolism , Protein Binding/drug effects , RNA Splice Sites/genetics , Serine-Arginine Splicing Factors/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Transcriptional Elongation Factors/metabolism
10.
Hypertension ; 76(3): 724-731, 2020 09.
Article En | MEDLINE | ID: mdl-32654554

Afferent lesions of the arterial baroreflex occur in familial dysautonomia. This leads to excessive blood pressure variability with falls and frequent surges that damage the organs. These hypertensive surges are the result of excess peripheral catecholamine release and have no adequate treatment. Carbidopa is a selective DOPA-decarboxylase inhibitor that suppresses catecholamines production outside the brain. To learn whether carbidopa can inhibit catecholamine-induced hypertensive surges in patients with severe afferent baroreflex failure, we conducted a double-blind randomized crossover trial in which patients with familial dysautonomia received high dose carbidopa (600 mg/day), low-dose carbidopa (300 mg/day), or matching placebo in 3 4-week treatment periods. Among the 22 patients enrolled (13 females/8 males), the median age was 26 (range, 12-59 years). At enrollment, patients had hypertensive peaks to 164/116 (range, 144/92 to 213/150 mm Hg). Twenty-four hour urinary norepinephrine excretion, a marker of peripheral catecholamine release, was significantly suppressed on both high dose and low dose carbidopa, compared with placebo (P=0.0075). The 2 co-primary end points of the trial were met. The SD of systolic BP variability was reduced at both carbidopa doses (low dose: 17±4; high dose: 18±5 mm Hg) compared with placebo (23±7 mm Hg; P=0.0013), and there was a significant reduction in the systolic BP peaks on active treatment (P=0.0015). High- and low-dose carbidopa were similarly effective and well tolerated. This study provides class Ib evidence that carbidopa can reduce blood pressure variability in patients with congenital afferent baroreflex failure. Similar beneficial effects are observed in patients with acquired baroreflex lesions.


Baroreflex , Blood Pressure , Carbidopa , Dysautonomia, Familial , Hypertension , Adult , Afferent Pathways/drug effects , Afferent Pathways/metabolism , Afferent Pathways/physiopathology , Aromatic Amino Acid Decarboxylase Inhibitors/administration & dosage , Aromatic Amino Acid Decarboxylase Inhibitors/pharmacokinetics , Baroreflex/drug effects , Baroreflex/physiology , Blood Pressure/drug effects , Blood Pressure/physiology , Carbidopa/administration & dosage , Carbidopa/pharmacokinetics , Catecholamines/metabolism , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Drug Monitoring/methods , Dysautonomia, Familial/diagnosis , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/physiopathology , Female , Humans , Hypertension/drug therapy , Hypertension/etiology , Hypertension/physiopathology , Male , Treatment Outcome
11.
J Clin Invest ; 130(5): 2478-2487, 2020 05 01.
Article En | MEDLINE | ID: mdl-32281946

Familial dysautonomia (FD) is the most prevalent form of hereditary sensory and autonomic neuropathy (HSAN). In FD, a germline mutation in the Elp1 gene leads to Elp1 protein decrease that causes sympathetic neuron death and sympathetic nervous system dysfunction (dysautonomia). Elp1 is best known as a scaffolding protein within the nuclear hetero-hexameric transcriptional Elongator protein complex, but how it functions in sympathetic neuron survival is very poorly understood. Here, we identified a cytoplasmic function for Elp1 in sympathetic neurons that was essential for retrograde nerve growth factor (NGF) signaling and neuron target tissue innervation and survival. Elp1 was found to bind to internalized TrkA receptors in an NGF-dependent manner, where it was essential for maintaining TrkA receptor phosphorylation (activation) by regulating PTPN6 (Shp1) phosphatase activity within the signaling complex. In the absence of Elp1, Shp1 was hyperactivated, leading to premature TrkA receptor dephosphorylation, which resulted in retrograde signaling failure and neuron death. Inhibiting Shp1 phosphatase activity in the absence of Elp1 rescued NGF-dependent retrograde signaling, and in an animal model of FD it rescued abnormal sympathetic target tissue innervation. These results suggest that regulation of retrograde NGF signaling in sympathetic neurons by Elp1 may explain sympathetic neuron loss and physiologic dysautonomia in patients with FD.


Dysautonomia, Familial/metabolism , Nerve Growth Factor/metabolism , Neurons/metabolism , Signal Transduction , Sympathetic Nervous System/metabolism , Animals , Dysautonomia, Familial/genetics , Dysautonomia, Familial/pathology , Germ-Line Mutation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Nerve Growth Factor/genetics , Neurons/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Sympathetic Nervous System/pathology
12.
PLoS One ; 14(3): e0211602, 2019.
Article En | MEDLINE | ID: mdl-30889183

Familial Dysautonomia (FD) is an autosomal recessive congenital neuropathy that results from a point mutation at the 5' splice site of intron 20 in the IKBKAP gene. This mutation decreases production of the IKAP protein, and treatments that increase the level of the full-length IKBKAP transcript are likely to be of therapeutic value. We previously found that phosphatidylserine (PS), an FDA-approved food supplement, elevates IKAP levels in cells generated from FD patients. Here we demonstrate that combined treatment of cells generated from FD patients with PS and kinetin or PS and the histone deacetylase inhibitor trichostatin A (TSA) resulted in an additive elevation of IKAP compared to each drug alone. This indicates that the compounds influence different pathways. We also found that pridopidine enhances production of IKAP in cells generated from FD patients. Pridopidine has an additive effect on IKAP levels when used in combination with kinetin or TSA, but not with PS; suggesting that PS and pridopidine influence IKBKAP levels through the same mechanism. Indeed, we demonstrate that the effect of PS and pridopidine is through sigma-1 receptor-mediated activation of the BDNF signaling pathway. A combination treatment with any of these drugs with different mechanisms has potential to benefit FD patients.


Carrier Proteins/metabolism , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Carrier Proteins/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Drug Therapy, Combination , Dysautonomia, Familial/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Kinetin/pharmacology , MAP Kinase Signaling System/drug effects , Phosphatidylserines/pharmacology , Piperidines/pharmacology , Transcriptional Elongation Factors , Treatment Outcome , Tubulin/metabolism
13.
J Pediatr Gastroenterol Nutr ; 68(3): 422-427, 2019 03.
Article En | MEDLINE | ID: mdl-30334929

OBJECTIVES: Familial dysautonomia (FD) is a rare hereditary sensory and autonomic neuropathy characterized by chronic lung disease and cyclic vomiting due to hyper-adrenergic crises. Most FD patients are in a depleted nutritional state; however, the phenotype of the disease is quite different between patients, as for the severity of lung disease and the intensity and frequency of these pathognomonic crises. In this study we wanted to investigate whether resting energy expenditure (REE) levels are increased in this population, and if correlations exist between REE levels and phenotype severity. METHODS: Data was collected from 12 FD patients (6/6 m/f). REE measurements were conducted by indirect calorimeter. Measured REE % predicted were correlated with pulmonary function, severity of the scoliosis, serum C-reactive protein, yearly frequency of hyperadrenergic crisis, hospital admissions and the use of nocturnal noninvasive positive pressure ventilation. RESULTS: Mean REE was 112 ±13% predicted with 50% being in a hypermetabolic state (REE/HB > 110%). Body mass index (BMI) was below normal range in 75% of patients, and reduced energy intake was also decreased in 75%. No significant correlations to disease severity factors were found. When dividing the subjects to REE levels above or below 125% predicted, Patients with REE above 125% predicted presented with significantly lower inspiratory capacity (42.7% predicted vs 62.8% predicted; P = 0.04). CONCLUSIONS: Hypermetabolic state was described in 50% of FD patients. The Low BMI is explained by combination of relative anorexia and increased REE. The REE levels are related to the underling respiratory disease.


Dysautonomia, Familial/metabolism , Energy Metabolism , Adult , Body Mass Index , Calorimetry, Indirect , Dysautonomia, Familial/physiopathology , Female , Humans , Lung/physiopathology , Male , Nutritional Status , Retrospective Studies , Skinfold Thickness , Thinness/metabolism
14.
Nat Commun ; 9(1): 889, 2018 03 01.
Article En | MEDLINE | ID: mdl-29497044

Familial dysautonomia (FD) results from mutation in IKBKAP/ELP1, a gene encoding the scaffolding protein for the Elongator complex. This highly conserved complex is required for the translation of codon-biased genes in lower organisms. Here we investigate whether Elongator serves a similar function in mammalian peripheral neurons, the population devastated in FD. Using codon-biased eGFP sensors, and multiplexing of codon usage with transcriptome and proteome analyses of over 6,000 genes, we identify two categories of genes, as well as specific gene identities that depend on Elongator for normal expression. Moreover, we show that multiple genes in the DNA damage repair pathway are codon-biased, and that with Elongator loss, their misregulation is correlated with elevated levels of DNA damage. These findings link Elongator's function in the translation of codon-biased genes with both the developmental and neurodegenerative phenotypes of FD, and also clarify the increased risk of cancer associated with the disease.


Codon/genetics , Dysautonomia, Familial/metabolism , Neurons/metabolism , Peptide Chain Elongation, Translational , Peripheral Nerves/metabolism , Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Codon/metabolism , Dysautonomia, Familial/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Neurons/cytology , Peripheral Nerves/cytology , Proteins/genetics
15.
RNA ; 23(9): 1393-1403, 2017 09.
Article En | MEDLINE | ID: mdl-28592461

The 5' splice site mutation (IVS20+6T>C) of the inhibitor of κ light polypeptide gene enhancer in B cells, kinase complex-associated protein (IKBKAP) gene in familial dysautonomia (FD) is at the sixth intronic nucleotide of the 5' splice site. It is known to weaken U1 snRNP recognition and result in an aberrantly spliced mRNA product in neuronal tissue, but normally spliced mRNA in other tissues. Aberrantly spliced IKBKAP mRNA abrogates IKK complex-associated protein (IKAP)/elongator protein 1 (ELP1) expression and results in a defect of neuronal cell development in FD. To elucidate the tissue-dependent regulatory mechanism, we screened an expression library of major RNA-binding proteins (RBPs) with our mammalian dual-color splicing reporter system and identified RBM24 as a regulator. RBM24 functioned as a cryptic intronic splicing enhancer binding to an element (IVS20+13-29) downstream from the intronic 5' splice site mutation in the IKBKAP gene and promoted U1 snRNP recognition only to the mutated 5' splice site (and not the wild-type 5' splice site). Our results show that tissue-specific expression of RBM24 can explain the neuron-specific aberrant splicing of IKBKAP exon 20 in familial dysautonomia, and that ectopic expression of RBM24 in neuronal tissue could be a novel therapeutic target of the disease.


Carrier Proteins/genetics , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Mutation , RNA Splice Sites , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Alternative Splicing , Animals , Cell Line , Exons , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , High-Throughput Screening Assays , Humans , Introns , Mice , Models, Biological , Organ Specificity/genetics , Protein Binding , RNA Precursors/chemistry , RNA Precursors/genetics , Regulatory Sequences, Nucleic Acid , Transcriptional Elongation Factors
16.
Proc Natl Acad Sci U S A ; 114(19): 5035-5040, 2017 05 09.
Article En | MEDLINE | ID: mdl-28439028

Hereditary sensory and autonomic neuropathy type III, or familial dysautonomia [FD; Online Mendelian Inheritance in Man (OMIM) 223900], affects the development and long-term viability of neurons in the peripheral nervous system (PNS) and retina. FD is caused by a point mutation in the gene IKBKAP/ELP1 that results in a tissue-specific reduction of the IKAP/ELP1 protein, a subunit of the Elongator complex. Hallmarks of the disease include vasomotor and cardiovascular instability and diminished pain and temperature sensation caused by reductions in sensory and autonomic neurons. It has been suggested but not demonstrated that mitochondrial function may be abnormal in FD. We previously generated an Ikbkap/Elp1 conditional-knockout mouse model that recapitulates the selective death of sensory (dorsal root ganglia) and autonomic neurons observed in FD. We now show that in these mice neuronal mitochondria have abnormal membrane potentials, produce elevated levels of reactive oxygen species, are fragmented, and do not aggregate normally at axonal branch points. The small hydroxylamine compound BGP-15 improved mitochondrial function, protecting neurons from dying in vitro and in vivo, and promoted cardiac innervation in vivo. Given that impairment of mitochondrial function is a common pathological component of neurodegenerative diseases such as amyotrophic lateral sclerosis and Alzheimer's, Parkinson's, and Huntington's diseases, our findings identify a therapeutic approach that may have efficacy in multiple degenerative conditions.


Axons/metabolism , Dysautonomia, Familial , Ganglia, Spinal/metabolism , Oximes/pharmacology , Piperidines/pharmacology , Animals , Axons/pathology , Carrier Proteins/genetics , Cell Death/drug effects , Cell Death/genetics , Disease Models, Animal , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/genetics , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Ganglia, Spinal/pathology , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains
17.
Neurobiol Dis ; 103: 113-122, 2017 Jul.
Article En | MEDLINE | ID: mdl-28404519

FD is a rare neurodegenerative disorder caused by a mutation of the IKBKAP gene, which induces low expression levels of the Elongator subunit IKAP/hELP1 protein. A rational strategy for FD treatment could be to identify drugs increasing IKAP/hELP1 expression levels by blocking protein degradation pathways such as the 26S proteasome. Proteasome inhibitors are promising molecules emerging in cancer treatment and could thus constitute an enticing pharmaceutical strategy for FD treatment. Therefore, we tested three proteasome inhibitors on FD human olfactory ecto-mesenchymal stem cells (hOE-MSCs): two approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA), bortezomib and carfilzomib, as well as epoxomicin. Although all 3 inhibitors demonstrated activity in correcting IKBKAP mRNA aberrant splicing, carfilzomib was superior in enhancing IKAP/hELP1 quantity. Moreover, we observed a synergistic effect of suboptimal doses of carfilzomib on kinetin in improving IKBKAP isoforms ratio and IKAP/hELP1 expression levels allowing to counterbalance carfilzomib toxicity. Finally, we identified several dysregulated miRNAs after carfilzomib treatment that target proteasome-associated mRNAs and determined that IKAP/hELP1 deficiency in FD pathology is correlated to an overactivity of the 26S proteasome. Altogether, these results reinforce the rationale for using chemical compounds inhibiting the 26S proteasome as an innovative option for FD and a promising therapeutic pathway for many other neurodegenerative diseases.


Carrier Proteins/metabolism , Dysautonomia, Familial/metabolism , Proteasome Inhibitors/pharmacology , RNA Splicing/physiology , RNA, Messenger/metabolism , Bortezomib/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dysautonomia, Familial/genetics , Dysautonomia, Familial/pathology , Humans , RNA Splicing/drug effects , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , Transcriptional Elongation Factors
18.
eNeuro ; 3(5)2016.
Article En | MEDLINE | ID: mdl-27699209

Familial dysautonomia (FD) is an autosomal recessive congenital neuropathy that is caused by a mutation in the gene for inhibitor of kappa B kinase complex-associated protein (IKBKAP). Although FD patients suffer from multiple neuropathies, a major debilitation that affects their quality of life is progressive blindness. To determine the requirement for Ikbkap in the developing and adult retina, we generated Ikbkap conditional knockout (CKO) mice using a TUBA1a promoter-Cre (Tα1-Cre). In the retina, Tα1-Cre expression is detected predominantly in retinal ganglion cells (RGCs). At 6 months, significant loss of RGCs had occurred in the CKO retinas, with the greatest loss in the temporal retina, which is the same spatial phenotype observed in FD, Leber hereditary optic neuropathy, and dominant optic atrophy. Interestingly, the melanopsin-positive RGCs were resistant to degeneration. By 9 months, signs of photoreceptor degeneration were observed, which later progressed to panretinal degeneration, including RGC and photoreceptor loss, optic nerve thinning, Müller glial activation, and disruption of layers. Taking these results together, we conclude that although Ikbkap is not required for normal development of RGCs, its loss causes a slow, progressive RGC degeneration most severely in the temporal retina, which is later followed by indirect photoreceptor loss and complete retinal disorganization. This mouse model of FD is not only useful for identifying the mechanisms mediating retinal degeneration, but also provides a model system in which to attempt to test therapeutics that may mitigate the loss of vision in FD patients.


Carrier Proteins/metabolism , Dysautonomia, Familial/metabolism , Retinal Degeneration/metabolism , Animals , Carrier Proteins/genetics , Disease Models, Animal , Disease Progression , Dysautonomia, Familial/pathology , Female , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins , Male , Mice, Knockout , Neuroglia/metabolism , Neuroglia/pathology , Optic Nerve/metabolism , Optic Nerve/pathology , Optic Neuritis/metabolism , Optic Neuritis/pathology , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Rod Opsins/metabolism , Time Factors
19.
Hum Mol Genet ; 25(7): 1307-17, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26769675

Familial dysautonomia (FD) is a genetic disorder manifested due to abnormal development and progressive degeneration of the sensory and autonomic nervous system. FD is caused by a point mutation in the IKBKAP gene encoding the IKAP protein, resulting in decreased protein levels. A promising potential treatment for FD is phosphatidylserine (PS); however, the manner by which PS elevates IKAP levels has yet to be identified. Analysis of ChIP-seq results of the IKBKAP promoter region revealed binding of the transcription factors CREB and ELK1, which are regulated by the mitogen-activated protein kinase (MAPK)/extracellular-regulated kinase (ERK) signaling pathway. We show that PS treatment enhanced ERK phosphorylation in cells derived from FD patients. ERK activation resulted in elevated IKBKAP transcription and IKAP protein levels, whereas pretreatment with the MAPK inhibitor U0126 blocked elevation of the IKAP protein level. Overexpression of either ELK1 or CREB activated the IKBKAP promoter, whereas downregulation of these transcription factors resulted in a decrease of the IKAP protein. Additionally, we show that PS improves cell migration, known to be enhanced by MAPK/ERK activation and abrogated in FD cells. In conclusion, our results demonstrate that PS activates the MAPK/ERK signaling pathway, resulting in activation of transcription factors that bind the promoter region of IKBKAP and thus enhancing its transcription. Therefore, compounds that activate the MAPK/ERK signaling pathway could constitute potential treatments for FD.


Carrier Proteins/genetics , Dysautonomia, Familial/drug therapy , MAP Kinase Signaling System/drug effects , Phosphatidylserines/pharmacology , Transcriptional Activation , Carrier Proteins/drug effects , Cyclic AMP Response Element-Binding Protein , Dysautonomia, Familial/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Female , Humans , Phosphatidylserines/therapeutic use , Transcriptional Elongation Factors , ets-Domain Protein Elk-1
20.
Hum Mol Genet ; 25(6): 1116-28, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26769677

Familial dysautonomia (FD) is an autosomal recessive neurodegenerative disease that affects the development and survival of sensory and autonomic neurons. FD is caused by an mRNA splicing mutation in intron 20 of the IKBKAP gene that results in a tissue-specific skipping of exon 20 and a corresponding reduction of the inhibitor of kappaB kinase complex-associated protein (IKAP), also known as Elongator complex protein 1. To date, several promising therapeutic candidates for FD have been identified that target the underlying mRNA splicing defect, and increase functional IKAP protein. Despite these remarkable advances in drug discovery for FD, we lacked a phenotypic mouse model in which we could manipulate IKBKAP mRNA splicing to evaluate potential efficacy. We have, therefore, engineered a new mouse model that, for the first time, will permit to evaluate the phenotypic effects of splicing modulators and provide a crucial platform for preclinical testing of new therapies. This new mouse model, TgFD9; Ikbkap(Δ20/flox) was created by introducing the complete human IKBKAP transgene with the major FD splice mutation (TgFD9) into a mouse that expresses extremely low levels of endogenous Ikbkap (Ikbkap(Δ20/flox)). The TgFD9; Ikbkap(Δ20/flox) mouse recapitulates many phenotypic features of the human disease, including reduced growth rate, reduced number of fungiform papillae, spinal abnormalities, and sensory and sympathetic impairments, and recreates the same tissue-specific mis-splicing defect seen in FD patients. This is the first mouse model that can be used to evaluate in vivo the therapeutic effect of increasing IKAP levels by correcting the underlying FD splicing defect.


Disease Models, Animal , Dysautonomia, Familial/metabolism , Dysautonomia, Familial/pathology , Alternative Splicing , Animals , Autonomic Pathways/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dysautonomia, Familial/genetics , Exons , Humans , Intracellular Signaling Peptides and Proteins , Introns , Male , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , RNA Splicing/genetics , RNA, Messenger/metabolism , Sensory Receptor Cells/metabolism
...