Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Cell Stem Cell ; 31(5): 734-753.e8, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38608707

RESUMO

Autonomic parasympathetic neurons (parasymNs) control unconscious body responses, including "rest-and-digest." ParasymN innervation is important for organ development, and parasymN dysfunction is a hallmark of autonomic neuropathy. However, parasymN function and dysfunction in humans are vastly understudied due to the lack of a model system. Human pluripotent stem cell (hPSC)-derived neurons can fill this void as a versatile platform. Here, we developed a differentiation paradigm detailing the derivation of functional human parasymNs from Schwann cell progenitors. We employ these neurons (1) to assess human autonomic nervous system (ANS) development, (2) to model neuropathy in the genetic disorder familial dysautonomia (FD), (3) to show parasymN dysfunction during SARS-CoV-2 infection, (4) to model the autoimmune disease Sjögren's syndrome (SS), and (5) to show that parasymNs innervate white adipocytes (WATs) during development and promote WAT maturation. Our model system could become instrumental for future disease modeling and drug discovery studies, as well as for human developmental studies.


Assuntos
Diferenciação Celular , Disautonomia Familiar , Células-Tronco Pluripotentes , Humanos , Células-Tronco Pluripotentes/citologia , Disautonomia Familiar/patologia , Neurônios , Síndrome de Sjogren/patologia , COVID-19/virologia , COVID-19/patologia , Animais , Sistema Nervoso Parassimpático , Células de Schwann , Camundongos , SARS-CoV-2/fisiologia
2.
Sci Rep ; 14(1): 570, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38177237

RESUMO

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.


Assuntos
Disautonomia Familiar , Humanos , Camundongos , Animais , Disautonomia Familiar/genética , Disautonomia Familiar/metabolismo , Disautonomia Familiar/patologia , Proteínas de Transporte/metabolismo , Sistema Nervoso Periférico/metabolismo , Células Receptoras Sensoriais/metabolismo , Perfilação da Expressão Gênica , Expressão Gênica
3.
Hum Mol Genet ; 31(11): 1776-1787, 2022 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-34908112

RESUMO

Familial dysautonomia (FD) is an autosomal recessive neurodegenerative disease caused by a splicing mutation in the gene encoding Elongator complex protein 1 (ELP1, also known as IKBKAP). This mutation results in tissue-specific skipping of exon 20 with a corresponding reduction of ELP1 protein, predominantly in the central and peripheral nervous system. Although FD patients have a complex neurological phenotype caused by continuous depletion of sensory and autonomic neurons, progressive visual decline leading to blindness is one of the most problematic aspects of the disease, as it severely affects their quality of life. To better understand the disease mechanism as well as to test the in vivo efficacy of targeted therapies for FD, we have recently generated a novel phenotypic mouse model, TgFD9; IkbkapΔ20/flox. This mouse exhibits most of the clinical features of the disease and accurately recapitulates the tissue-specific splicing defect observed in FD patients. Driven by the dire need to develop therapies targeting retinal degeneration in FD, herein, we comprehensively characterized the progression of the retinal phenotype in this mouse, and we demonstrated that it is possible to correct ELP1 splicing defect in the retina using the splicing modulator compound (SMC) BPN-15477.


Assuntos
Disautonomia Familiar , Peptídeos e Proteínas de Sinalização Intracelular , Doenças Neurodegenerativas , Doenças do Nervo Óptico , Células Ganglionares da Retina , Animais , Modelos Animais de Doenças , Disautonomia Familiar/patologia , Humanos , Camundongos , Doenças Neurodegenerativas/patologia , Doenças do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia
4.
J Neuropathol Exp Neurol ; 79(10): 1072-1083, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32954436

RESUMO

Von Economo neurons (VENs) and fork cells are principally located in the anterior cingulate cortex (ACC) and the frontoinsular cortex (FI). Both of these regions integrate inputs from the autonomic nervous system (ANS) and are involved in decision-making and perception of the emotional states of self and others. Familial dysautonomia (FD) is an orphan disorder characterized by autonomic dysfunction and behavioral abnormalities including repetitive behavior and emotional rigidity, which are also seen in autism spectrum disorder. To understand a possible link between the ANS and the cortical regions implicated in emotion regulation we studied VENs and fork cells in an autonomic disorder. We determined the densities of VENs, fork cells, and pyramidal neurons and the ratio of VENs and fork cells to pyramidal neurons in ACC and FI in 4 FD patient and 6 matched control brains using a stereologic approach. We identified alterations in densities of VENs and pyramidal neurons and their distributions in the ACC and FI in FD brains. These data suggest that alterations in migration and numbers of VENs may be involved in FD pathophysiology thereby supporting the notion of a functional link between VENs, the ANS and the peripheral nervous system in general.


Assuntos
Disautonomia Familiar/patologia , Neocórtex/patologia , Neurônios/patologia , Adulto , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
5.
J Clin Invest ; 130(5): 2478-2487, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32281946

RESUMO

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.


Assuntos
Disautonomia Familiar/metabolismo , Fator de Crescimento Neural/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/metabolismo , Animais , Disautonomia Familiar/genética , Disautonomia Familiar/patologia , Mutação em Linhagem Germinativa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/genética , Neurônios/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Sistema Nervoso Simpático/patologia
6.
Dis Model Mech ; 11(7)2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-29929962

RESUMO

Familial dysautonomia (FD) is an autosomal recessive disorder marked by developmental and progressive neuropathies. It is caused by an intronic point-mutation in the IKBKAP/ELP1 gene, which encodes the inhibitor of κB kinase complex-associated protein (IKAP, also called ELP1), a component of the elongator complex. Owing to variation in tissue-specific splicing, the mutation primarily affects the nervous system. One of the most debilitating hallmarks of FD that affects patients' quality of life is progressive blindness. To determine the pathophysiological mechanisms that are triggered by the absence of IKAP in the retina, we generated retina-specific Ikbkap conditional knockout (CKO) mice using Pax6-Cre, which abolished Ikbkap expression in all cell types of the retina. Although sensory and autonomic neuropathies in FD are known to be developmental in origin, the loss of IKAP in the retina did not affect its development, demonstrating that IKAP is not required for retinal development. The loss of IKAP caused progressive degeneration of retinal ganglion cells (RGCs) by 1 month of age. Mitochondrial membrane integrity was breached in RGCs, and later in other retinal neurons. In Ikbkap CKO retinas, mitochondria were depolarized, and complex I function and ATP were significantly reduced. Although mitochondrial impairment was detected in all Ikbkap-deficient retinal neurons, RGCs were the only cell type to degenerate; the survival of other retinal neurons was unaffected. This retina-specific FD model is a useful in vivo model for testing potential therapeutics for mitigating blindness in FD. Moreover, our data indicate that RGCs and mitochondria are promising targets.


Assuntos
Proteínas de Transporte/metabolismo , Disautonomia Familiar/patologia , Disautonomia Familiar/fisiopatologia , Mitocôndrias/patologia , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Trifosfato de Adenosina/metabolismo , Animais , Modelos Animais de Doenças , Peptídeos e Proteínas de Sinalização Intracelular , Potencial da Membrana Mitocondrial , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Especificidade de Órgãos , Células Ganglionares da Retina/ultraestrutura
7.
Nucleic Acids Res ; 46(10): 4833-4844, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29672717

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Disautonomia Familiar/genética , Disautonomia Familiar/terapia , Oligonucleotídeos Antissenso/farmacologia , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Disautonomia Familiar/patologia , Elementos Facilitadores Genéticos , Éxons , Fibroblastos , Humanos , Camundongos Transgênicos , Oligonucleotídeos Antissenso/química , Sítios de Splice de RNA , Splicing de RNA , Fatores de Elongação da Transcrição
8.
PLoS One ; 13(4): e0196599, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29698477

RESUMO

INTRODUCTION AND OBJECTIVES: Progressive ataxic gait is a common symptom in individuals with Familial Dysautonomia (FD). At least 50% of adults with FD require assistance with walking. Our aims were to describe the medical condition of individuals with FD (ii) compare their gait characteristics to healthy individuals, and (iii) assess correlations between gait measures, presence of unstable gait pattern and frequency of falls. METHODS: Twelve subjects with FD (7 males, age 25.3±10.6 years) and 16 healthy participants (6 males, age 35.9±11.9 years) were recruited. Gait kinematics, gait symmetry, dynamic muscle activity, and foot deep vibration sensation were recorded. RESULTS: Ataxic gait degrees were: severe (6 out of 12), moderate (4 out of 12) and low (2 out of 12). The number of falls correlated with base width asymmetry. Crouch gait was noted in 3 out of 12 of the subjects. CONCLUSIONS: In-depth quantitative gait analysis of individuals with FD revealed ataxic gait. The ataxic pattern might be a result of combined neurological deficiencies and osseous deformities. Increasing the base of support of patients with FD might increase the symmetry of the base width during gait and decrease the number of falls. Additionally, perturbation treatment and dynamic balance exercises may be recommended in order to improve compensatory strategies. Future investigation of this population should include quantification of osseous rotations of the lower limb in order to fully understand its effect on their gait pattern and falls.


Assuntos
Disautonomia Familiar/patologia , Marcha Atáxica/fisiopatologia , Marcha/fisiologia , Acidentes por Quedas , Adolescente , Adulto , Fenômenos Biomecânicos , Índice de Massa Corporal , Estudos de Casos e Controles , Criança , Disautonomia Familiar/complicações , Eletromiografia , Feminino , Marcha Atáxica/complicações , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X , Adulto Jovem
9.
Clin Auton Res ; 27(4): 235-243, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28667575

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Disautonomia Familiar/patologia , Células-Tronco/fisiologia , Animais , Disautonomia Familiar/genética , Disautonomia Familiar/terapia , Humanos , Camundongos
10.
Proc Natl Acad Sci U S A ; 114(19): 5035-5040, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439028

RESUMO

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.


Assuntos
Axônios/metabolismo , Disautonomia Familiar , Gânglios Espinais/metabolismo , Oximas/farmacologia , Piperidinas/farmacologia , Animais , Axônios/patologia , Proteínas de Transporte/genética , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Disautonomia Familiar/tratamento farmacológico , Disautonomia Familiar/genética , Disautonomia Familiar/metabolismo , Disautonomia Familiar/patologia , Gânglios Espinais/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Mutantes
11.
J Neuropathol Exp Neurol ; 76(3): 238-244, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28395083

RESUMO

Clinical data suggest that optic neuropathy and retinal ganglion cell loss are the main cause of visual decline in patients with familial dysautonomia, but this has not previously been confirmed by pathological analyses. We studied retinas and optic nerves in 6 eyes from 3 affected patients obtained at autopsy. Analyses included routine neurohistology and immunohistochemistry for neurofilaments, cytochrome c oxidase (COX), and melanopsin-containing ganglion cells. We observed profound axon loss in the temporal portions of optic nerves with relative preservation in the nasal portions; this correlated with clinical and optical coherence tomography findings in 1 patient. Retinal ganglion cell layers were markedly reduced in the central retina, whereas melanopsin-containing ganglion cells were relatively spared. COX staining was reduced in the temporal portions of the optic nerve indicating reduced mitochondrial density. Axonal swelling with degenerating lysosomes and mitochondria were observed by electron microscopy. These findings support the concept that there is a specific optic neuropathy and retinopathy in patients with familial dysautonomia similar to that seen in other optic neuropathies with mitochondrial dysfunction. This raises the possibility that defective expression of the IkB kinase complex-associated protein (IKAP) resulting from mutations in IKBKAP affects mitochondrial function in the metabolism-dependent retinal parvocellular ganglion cells in this condition.


Assuntos
Disautonomia Familiar/complicações , Disautonomia Familiar/patologia , Doenças do Nervo Óptico/complicações , Doenças do Nervo Óptico/patologia , Adolescente , Criança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nervo Óptico/patologia , Células Ganglionares da Retina/patologia
12.
Neurobiol Dis ; 103: 113-122, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28404519

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Disautonomia Familiar/metabolismo , Inibidores de Proteassoma/farmacologia , Splicing de RNA/fisiologia , RNA Mensageiro/metabolismo , Bortezomib/farmacologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Disautonomia Familiar/genética , Disautonomia Familiar/patologia , Humanos , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , Fatores de Elongação da Transcrição
13.
PLoS Genet ; 12(12): e1006486, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27997532

RESUMO

Familial Dysautonomia (FD) is a neurodegenerative disease in which aberrant tissue-specific splicing of IKBKAP exon 20 leads to reduction of IKAP protein levels in neuronal tissues. Here we generated a conditional knockout (CKO) mouse in which exon 20 of IKBKAP is deleted in the nervous system. The CKO FD mice exhibit developmental delays, sensory abnormalities, and less organized dorsal root ganglia (DRGs) with attenuated axons compared to wild-type mice. Furthermore, the CKO FD DRGs show elevated HDAC6 levels, reduced acetylated α-tubulin, unstable microtubules, and impairment of axonal retrograde transport of nerve growth factor (NGF). These abnormalities in DRG properties underlie neuronal degeneration and FD symptoms. Phosphatidylserine treatment decreased HDAC6 levels and thus increased acetylation of α-tubulin. Further PS treatment resulted in recovery of axonal outgrowth and enhanced retrograde axonal transport by decreasing histone deacetylase 6 (HDAC6) levels and thus increasing acetylation of α-tubulin levels. Thus, we have identified the molecular pathway that leads to neurodegeneration in FD and have demonstrated that phosphatidylserine treatment has the potential to slow progression of neurodegeneration.


Assuntos
Transporte Axonal/efeitos dos fármacos , Disautonomia Familiar/genética , Histona Desacetilases/genética , Fosfatidilserinas/administração & dosagem , Tubulina (Proteína)/genética , Processamento Alternativo/genética , Animais , Transporte Axonal/genética , Axônios/efeitos dos fármacos , Modelos Animais de Doenças , Disautonomia Familiar/tratamento farmacológico , Disautonomia Familiar/patologia , Éxons/genética , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/patologia , Desacetilase 6 de Histona , Histona Desacetilases/biossíntese , Humanos , Camundongos , Camundongos Knockout , Degeneração Neural/tratamento farmacológico , Degeneração Neural/genética , Degeneração Neural/patologia , Fator de Crescimento Neural/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fosfatidilserinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
14.
eNeuro ; 3(5)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27699209

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Disautonomia Familiar/metabolismo , Degeneração Retiniana/metabolismo , Animais , Proteínas de Transporte/genética , Modelos Animais de Doenças , Progressão da Doença , Disautonomia Familiar/patologia , Feminino , Técnicas de Inativação de Genes , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos Knockout , Neuroglia/metabolismo , Neuroglia/patologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Neurite Óptica/metabolismo , Neurite Óptica/patologia , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Degeneração Retiniana/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Opsinas de Bastonetes/metabolismo , Fatores de Tempo
15.
Dis Model Mech ; 9(8): 899-909, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27483351

RESUMO

Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a mutation in intron 20 of the IKBKAP gene (c.2204+6T>C), leading to tissue-specific skipping of exon 20 and a decrease in the synthesis of the encoded protein IKAP (also known as ELP1). Small non-coding RNAs known as microRNAs (miRNAs) are important post-transcriptional regulators of gene expression and play an essential role in the nervous system development and function. To better understand the neuronal specificity of IKAP loss, we examined expression of miRNAs in human olfactory ecto-mesenchymal stem cells (hOE-MSCs) from five control individuals and five FD patients. We profiled the expression of 373 miRNAs using microfluidics and reverse transcription coupled to quantitative PCR (RT-qPCR) on two biological replicate series of hOE-MSC cultures from healthy controls and FD patients. This led to the total identification of 26 dysregulated miRNAs in FD, validating the existence of a miRNA signature in FD. We then selected the nine most discriminant miRNAs for further analysis. The signaling pathways affected by these dysregulated miRNAs were largely within the nervous system. In addition, many targets of these dysregulated miRNAs had been previously demonstrated to be affected in FD models. Moreover, we found that four of our nine candidate miRNAs target the neuron-specific splicing factor NOVA1. We demonstrated that overexpression of miR-203a-3p leads to a decrease of NOVA1, counter-balanced by an increase of IKAP, supporting a potential interaction between NOVA1 and IKAP. Taken together, these results reinforce the choice of miRNAs as potential therapeutic targets and suggest that NOVA1 could be a regulator of FD pathophysiology.


Assuntos
Proteínas de Transporte/metabolismo , Disautonomia Familiar/genética , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo/genética , Proteínas de Transporte/genética , Estudos de Casos e Controles , Disautonomia Familiar/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Antígeno Neuro-Oncológico Ventral , Bulbo Olfatório/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Transdução de Sinais/genética , Fatores de Elongação da Transcrição
16.
Hum Mol Genet ; 25(6): 1116-28, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26769677

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Disautonomia Familiar/metabolismo , Disautonomia Familiar/patologia , Processamento Alternativo , Animais , Vias Autônomas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Disautonomia Familiar/genética , Éxons , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Íntrons , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Splicing de RNA/genética , RNA Mensageiro/metabolismo , Células Receptoras Sensoriais/metabolismo
17.
PLoS One ; 10(10): e0138807, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26437462

RESUMO

A splicing mutation in the IKBKAP gene causes Familial Dysautonomia (FD), affecting the IKAP protein expression levels and proper development and function of the peripheral nervous system (PNS). Here we found new molecular insights for the IKAP role and the impact of the FD mutation in the human PNS lineage by using a novel and unique human embryonic stem cell (hESC) line homozygous to the FD mutation originated by pre implantation genetic diagnosis (PGD) analysis. We found that IKBKAP downregulation during PNS differentiation affects normal migration in FD-hESC derived neural crest cells (NCC) while at later stages the PNS neurons show reduced intracellular colocalization between vesicular proteins and IKAP. Comparative wide transcriptome analysis of FD and WT hESC-derived neurons together with the analysis of human brains from FD and WT 12 weeks old embryos and experimental validation of the results confirmed that synaptic vesicular and neuronal transport genes are directly or indirectly affected by IKBKAP downregulation in FD neurons. Moreover we show that kinetin (a drug that corrects IKBKAP alternative splicing) promotes the recovery of IKAP expression and these IKAP functional associated genes identified in the study. Altogether, these results support the view that IKAP might be a vesicular like protein that might be involved in neuronal transport in hESC derived PNS neurons. This function seems to be mostly affected in FD-hESC derived PNS neurons probably reflecting some PNS neuronal dysfunction observed in FD.


Assuntos
Proteínas de Transporte/metabolismo , Regulação para Baixo , Disautonomia Familiar/genética , Células-Tronco Embrionárias Humanas/patologia , Neurônios/metabolismo , Sistema Nervoso Periférico/patologia , Vesículas Sinápticas/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Proteínas de Transporte/genética , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Disautonomia Familiar/metabolismo , Disautonomia Familiar/patologia , Feto , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Cinetina/farmacologia , Masculino , Mutação , Crista Neural/efeitos dos fármacos , Crista Neural/patologia , Neurônios/efeitos dos fármacos , Sistema Nervoso Periférico/efeitos dos fármacos , Fenótipo , Vesículas Sinápticas/efeitos dos fármacos , Fatores de Elongação da Transcrição
18.
Nat Neurosci ; 18(1): 17-24, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25420066

RESUMO

Reprogramming somatic cells from one cell fate to another can generate specific neurons suitable for disease modeling. To maximize the utility of patient-derived neurons, they must model not only disease-relevant cell classes, but also the diversity of neuronal subtypes found in vivo and the pathophysiological changes that underlie specific clinical diseases. We identified five transcription factors that reprogram mouse and human fibroblasts into noxious stimulus-detecting (nociceptor) neurons. These recapitulated the expression of quintessential nociceptor-specific functional receptors and channels found in adult mouse nociceptor neurons, as well as native subtype diversity. Moreover, the derived nociceptor neurons exhibited TrpV1 sensitization to the inflammatory mediator prostaglandin E2 and the chemotherapeutic drug oxaliplatin, modeling the inherent mechanisms underlying inflammatory pain hypersensitivity and painful chemotherapy-induced neuropathy. Using fibroblasts from patients with familial dysautonomia (hereditary sensory and autonomic neuropathy type III), we found that the technique was able to reveal previously unknown aspects of human disease phenotypes in vitro.


Assuntos
Fibroblastos , Modelos Neurológicos , Nociceptores , Dor/fisiopatologia , Células Receptoras Sensoriais , Animais , Disautonomia Familiar/patologia , Fenômenos Eletrofisiológicos/fisiologia , Humanos , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Doenças do Sistema Nervoso Periférico/patologia , Fatores de Transcrição
19.
Biochem Biophys Res Commun ; 454(3): 441-5, 2014 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-25450681

RESUMO

Familial dysautonomia (FD) is a recessive neurodegenerative genetic disease. FD is caused by a mutation in the IKBKAP gene resulting in a splicing defect and reduced levels of full length IKAP protein. IKAP homologues can be found in all eukaryotes and are part of a conserved six subunit protein complex, Elongator complex. Inactivation of any Elongator subunit gene in multicellular organisms cause a wide range of phenotypes, suggesting that Elongator has a pivotal role in several cellular processes. In yeast, there is convincing evidence that the main role of Elongator complex is in formation of modified wobble uridine nucleosides in tRNA and that their absence will influence translational efficiency. To date, no study has explored the possibility that FD patients display defects in formation of modified wobble uridine nucleosides as a consequence of reduced IKAP levels. In this study, we show that brain tissue and fibroblast cell lines from FD patients have reduced levels of the wobble uridine nucleoside 5-methoxycarbonylmethyl-2-thiouridine (mcm(5)s(2)U). Our findings indicate that FD could be caused by inefficient translation due to lower levels of wobble uridine nucleosides.


Assuntos
Encéfalo/patologia , Disautonomia Familiar/patologia , Fibroblastos/patologia , RNA de Transferência/química , Tiouridina/análogos & derivados , Encéfalo/metabolismo , Linhagem Celular , Disautonomia Familiar/metabolismo , Fibroblastos/metabolismo , Humanos , RNA de Transferência/metabolismo , Tiouridina/análise , Tiouridina/metabolismo
20.
PLoS One ; 9(11): e113428, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25409162

RESUMO

A splicing mutation in the ikbkap gene causes Familial Dysautonomia (FD), affecting the IKAP protein expression levels and proper development and function of the peripheral nervous system (PNS). Here we attempted to elucidate the role of IKAP in PNS development in the chick embryo and found that IKAP is required for proper axonal outgrowth, branching, and peripheral target innervation. Moreover, we demonstrate that IKAP colocalizes with activated JNK (pJNK), dynein, and ß-tubulin at the axon terminals of dorsal root ganglia (DRG) neurons, and may be involved in transport of specific target derived signals required for transcription of JNK and NGF responsive genes in the nucleus. These results suggest the novel role of IKAP in neuronal transport and specific signaling mediated transcription, and provide, for the first time, the basis for a molecular mechanism behind the FD phenotype.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fator de Crescimento Neural/metabolismo , Sistema Nervoso Periférico/patologia , Animais , Axônios/metabolismo , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Movimento Celular , Células Cultivadas , Embrião de Galinha , Galinhas , Dineínas/metabolismo , Disautonomia Familiar/genética , Disautonomia Familiar/patologia , Gânglios Espinais/citologia , Microscopia de Fluorescência , Neurônios/citologia , Neurônios/metabolismo , Sistema Nervoso Periférico/crescimento & desenvolvimento , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...