Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 18600, 2023 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-37903840

RESUMEN

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.


Asunto(s)
Disautonomía Familiar , Enfermedades Neurodegenerativas , Ratones , Animales , Humanos , Células Ganglionares de la Retina/metabolismo , Disautonomía Familiar/genética , Disautonomía Familiar/terapia , Disautonomía Familiar/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Empalme del ARN , Terapia Genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
2.
bioRxiv ; 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37293016

RESUMEN

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 data foundation for translation to FD patients.

3.
Metabolites ; 13(3)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36984872

RESUMEN

Central metabolism has a profound impact on the clinical phenotypes and penetrance of neurological diseases such as Alzheimer's (AD) and Parkinson's (PD) diseases, Amyotrophic Lateral Sclerosis (ALS) and Autism Spectrum Disorder (ASD). In contrast to the multifactorial origin of these neurological diseases, neurodevelopmental impairment and neurodegeneration in Familial Dysautonomia (FD) results from a single point mutation in the ELP1 gene. FD patients represent a well-defined population who can help us better understand the cellular networks underlying neurodegeneration, and how disease traits are affected by metabolic dysfunction, which in turn may contribute to dysregulation of the gut-brain axis of FD. Here, 1H NMR spectroscopy was employed to characterize the serum and fecal metabolomes of FD patients, and to assess similarities and differences in the polar metabolite profiles between FD patients and healthy relative controls. Findings from this work revealed noteworthy metabolic alterations reflected in energy (ATP) production, mitochondrial function, amino acid and nucleotide catabolism, neurosignaling molecules, and gut-microbial metabolism. These results provide further evidence for a close interconnection between metabolism, neurodegeneration, and gut microbiome dysbiosis in FD, and create an opportunity to explore whether metabolic interventions targeting the gut-brain-metabolism axis of FD could be used to redress or slow down the progressive neurodegeneration observed in FD patients.

4.
Nat Commun ; 14(1): 218, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-36639365

RESUMEN

Familial dysautonomia (FD) is a rare genetic neurologic disorder caused by impaired neuronal development and progressive degeneration of both the peripheral and central nervous systems. FD is monogenic, with >99.4% of patients sharing an identical point mutation in the elongator acetyltransferase complex subunit 1 (ELP1) gene, providing a relatively simple genetic background in which to identify modifiable factors that influence pathology. Gastrointestinal symptoms and metabolic deficits are common among FD patients, which supports the hypothesis that the gut microbiome and metabolome are altered and dysfunctional compared to healthy individuals. Here we show significant differences in gut microbiome composition (16 S rRNA gene sequencing of stool samples) and NMR-based stool and serum metabolomes between a cohort of FD patients (~14% of patients worldwide) and their cohabitating, healthy relatives. We show that key observations in human subjects are recapitulated in a neuron-specific Elp1-deficient mouse model, and that cohousing mutant and littermate control mice ameliorates gut microbiome dysbiosis, improves deficits in gut transit, and reduces disease severity. Our results provide evidence that neurologic deficits in FD alter the structure and function of the gut microbiome, which shifts overall host metabolism to perpetuate further neurodegeneration.


Asunto(s)
Disautonomía Familiar , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Disautonomía Familiar/genética , Disbiosis/metabolismo , Neuronas/metabolismo , Sistema Nervioso Central/metabolismo
5.
Nat Commun ; 13(1): 7032, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396637

RESUMEN

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.


Asunto(s)
Disautonomía Familiar , Animales , Humanos , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Neuronas/metabolismo , Norepinefrina/metabolismo , Mutación
6.
Elife ; 112022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35713404

RESUMEN

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.


Asunto(s)
Disautonomía Familiar , Animales , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Dolor Facial/metabolismo , Ratones , Cresta Neural/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Ganglio del Trigémino
7.
Dis Model Mech ; 15(5)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35481599

RESUMEN

Cardiovascular instability and a blunted respiratory drive in hypoxic conditions are hallmark features of the genetic sensory and autonomic neuropathy, familial dysautonomia (FD). FD results from a mutation in the gene ELP1, the encoded protein of which is a scaffolding subunit of the six-subunit Elongator complex. In mice, we and others have shown that Elp1 is essential for the normal development of neural crest-derived dorsal root ganglia sensory neurons. Whether Elp1 is also required for development of ectodermal placode-derived visceral sensory receptors, which are required for normal baroreception and chemosensory responses, has not been investigated. Using mouse models for FD, we here show that the entire circuitry underlying baroreception and chemoreception is impaired due to a requirement for Elp1 in the visceral sensory neuron ganglia, as well as for normal peripheral target innervation, and in their central nervous system synaptic partners in the medulla. Thus, Elp1 is required in both placode- and neural crest-derived sensory neurons, and its reduction aborts the normal development of neuronal circuitry essential for autonomic homeostasis and interoception. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas Portadoras , Disautonomía Familiar , Animales , Proteínas Portadoras/metabolismo , Sistema Nervioso Central/metabolismo , Disautonomía Familiar/genética , Ganglios Espinales/metabolismo , Humanos , Ratones , Cresta Neural/metabolismo
8.
Biol Open ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34590699

RESUMEN

Elongator dysfunction is increasingly recognized as a contributor to multiple neurodevelopmental and neurodegenerative disorders including familial dysautonomia, intellectual disability, amyotrophic lateral sclerosis, and autism spectrum disorder. Although numerous cellular processes are perturbed in the context of Elongator loss, converging evidence from multiple studies has resolved Elongator's primary function in the cell to the modification of tRNA wobble uridines and the translational regulation of codon-biased genes. Here we characterize H2a.z, encoding the variant H2a histone H2A.Z, as an indirect Elongator target. We further show that canonical Notch signaling, a pathway directed by H2A.Z, is perturbed as a consequence of Elp1 loss. Finally, we demonstrate that hyperacetylation of H2A.Z and other histones via exposure to the histone deacetylase inhibitor Trichostatin A during neurogenesis corrects the expression of Notch3 and rescues the development of sensory neurons in embryos lacking the Elp1 Elongator subunit.


Asunto(s)
Histonas/metabolismo , Enfermedades Neurodegenerativas/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Factores de Elongación Transcripcional/genética , Humanos , Mutación con Pérdida de Función/genética
9.
Semin Neurol ; 40(5): 473-484, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32927484

RESUMEN

Investigations of the cellular and molecular mechanisms that mediate the development of the autonomic nervous system have identified critical genes and signaling pathways that, when disrupted, cause disorders of the autonomic nervous system. This review summarizes our current understanding of how the autonomic nervous system emerges from the organized spatial and temporal patterning of precursor cell migration, proliferation, communication, and differentiation, and discusses potential clinical implications for developmental disorders of the autonomic nervous system, including familial dysautonomia, Hirschsprung disease, Rett syndrome, and congenital central hypoventilation syndrome.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo , Sistema Nervioso Autónomo/crecimiento & desarrollo , Disautonomía Familiar , Enfermedad de Hirschsprung , Hipoventilación/congénito , Síndrome de Rett , Apnea Central del Sueño , Humanos
10.
Dev Dyn ; 249(9): 1077-1097, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32390246

RESUMEN

BACKGROUND: Neural crest cells (NCCs) delaminate from the neural tube (NT) and migrate ventrally to generate the trunk peripheral nervous system (PNS). Although several signaling pathways have been identified that steer NCCs once they are on their ventral trajectory, no molecules have been identified that are required for the initial migration between the NT and the dorsal root ganglion. Given the critical role of fibroblast growth factor (FGF) signaling in embryogenesis, we investigated its function in this initial migration. RESULTS: FGFR1 signaling is required for the migration of newly delaminated NCCs onto the ventral pathway. Live imaging of migrating NCCs revealed that inhibition of FGFR1 signaling caused the dorsally stalled NCCs to lose their dorsal/ventral oriented polarity and instead adopt a rounded morphology while dynamically extending filopodia. FGF8, an FGFR1 ligand, increased motility of NCCs away from the NT by acting chemokinetically. Finally, we provide evidence that inhibition of FGFR1-mediated chemokinesis is partially rescued by increasing Akt signaling, inhibiting RhoA, and activation of N-cadherin signaling. CONCLUSION: These data support a model in which NCCs are stimulated chemokinetically by FGF:FGFR1 signaling, and that this activation positions and orients NCCs on their ventral migratory route-a process that is essential for patterning the trunk PNS.


Asunto(s)
Movimiento Celular , Quimiocinas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Cresta Neural/metabolismo , Transducción de Señal , Animales , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Ratones , Cresta Neural/citología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
11.
Dis Model Mech ; 11(7)2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-29929962

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Disautonomía Familiar/patología , Disautonomía Familiar/fisiopatología , Mitocondrias/patología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Adenosina Trifosfato/metabolismo , Animales , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular , Potencial de la Membrana Mitocondrial , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Especificidad de Órganos , Células Ganglionares de la Retina/ultraestructura
12.
Nat Commun ; 9(1): 889, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29497044

RESUMEN

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.


Asunto(s)
Codón/genética , Disautonomía Familiar/metabolismo , Neuronas/metabolismo , Extensión de la Cadena Peptídica de Translación , Nervios Periféricos/metabolismo , Proteínas/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Codón/metabolismo , Disautonomía Familiar/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Neuronas/citología , Nervios Periféricos/citología , Proteínas/genética
14.
Clin Auton Res ; 27(4): 235-243, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28667575

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Disautonomía Familiar/patología , Células Madre/fisiología , Animales , Disautonomía Familiar/genética , Disautonomía Familiar/terapia , Humanos , Ratones
16.
Proc Natl Acad Sci U S A ; 114(19): 5035-5040, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28439028

RESUMEN

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.


Asunto(s)
Axones/metabolismo , Disautonomía Familiar , Ganglios Espinales/metabolismo , Oximas/farmacología , Piperidinas/farmacología , Animales , Axones/patología , Proteínas Portadoras/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Modelos Animales de Enfermedad , Disautonomía Familiar/tratamiento farmacológico , Disautonomía Familiar/genética , Disautonomía Familiar/metabolismo , Disautonomía Familiar/patología , Ganglios Espinales/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes
17.
Dis Model Mech ; 10(5): 605-618, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28167615

RESUMEN

Hereditary sensory and autonomic neuropathies (HSANs) are a genetically and clinically diverse group of disorders defined by peripheral nervous system (PNS) dysfunction. HSAN type III, known as familial dysautonomia (FD), results from a single base mutation in the gene IKBKAP that encodes a scaffolding unit (ELP1) for a multi-subunit complex known as Elongator. Since mutations in other Elongator subunits (ELP2 to ELP4) are associated with central nervous system (CNS) disorders, the goal of this study was to investigate a potential requirement for Ikbkap in the CNS of mice. The sensory and autonomic pathophysiology of FD is fatal, with the majority of patients dying by age 40. While signs and pathology of FD have been noted in the CNS, the clinical and research focus has been on the sensory and autonomic dysfunction, and no genetic model studies have investigated the requirement for Ikbkap in the CNS. Here, we report, using a novel mouse line in which Ikbkap is deleted solely in the nervous system, that not only is Ikbkap widely expressed in the embryonic and adult CNS, but its deletion perturbs both the development of cortical neurons and their survival in adulthood. Primary cilia in embryonic cortical apical progenitors and motile cilia in adult ependymal cells are reduced in number and disorganized. Furthermore, we report that, in the adult CNS, both autonomic and non-autonomic neuronal populations require Ikbkap for survival, including spinal motor and cortical neurons. In addition, the mice developed kyphoscoliosis, an FD hallmark, indicating its neuropathic etiology. Ultimately, these perturbations manifest in a developmental and progressive neurodegenerative condition that includes impairments in learning and memory. Collectively, these data reveal an essential function for Ikbkap that extends beyond the peripheral nervous system to CNS development and function. With the identification of discrete CNS cell types and structures that depend on Ikbkap, novel strategies to thwart the progressive demise of CNS neurons in FD can be developed.


Asunto(s)
Proteínas Portadoras/genética , Sistema Nervioso Central/metabolismo , Disautonomía Familiar/genética , Animales , Conducta Animal , Supervivencia Celular/genética , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/patología , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Mutación , Neuronas/patología
18.
eNeuro ; 3(5)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27699209

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Disautonomía Familiar/metabolismo , Degeneración Retiniana/metabolismo , Animales , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Disautonomía Familiar/patología , Femenino , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones Noqueados , Neuroglía/metabolismo , Neuroglía/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Neuritis Óptica/metabolismo , Neuritis Óptica/patología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Opsinas de Bastones/metabolismo , Factores de Tiempo
19.
Dev Biol ; 413(1): 70-85, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-26988118

RESUMEN

During amniote embryogenesis the nervous and vascular systems interact in a process that significantly affects the respective morphogenesis of each network by forming a "neurovascular" link. The importance of neurovascular cross-talk in the central nervous system has recently come into focus with the growing awareness that these two systems interact extensively both during development, in the stem-cell niche, and in neurodegenerative conditions such as Alzheimer's Disease and Amyotrophic Lateral Sclerosis. With respect to the peripheral nervous system, however, there have been no live, real-time investigations of the potential relationship between these two developing systems. To address this deficit, we used multispectral 4D time-lapse imaging in a transgenic quail model in which endothelial cells (ECs) express a yellow fluorescent marker, while neural crest cells (NCCs) express an electroporated red fluorescent marker. We monitored EC and NCC migration in real-time during formation of the peripheral nervous system. Our time-lapse recordings indicate that NCCs and ECs are physically juxtaposed and dynamically interact at multiple locations along their trajectories. These interactions are stereotypical and occur at precise anatomical locations along the NCC migratory pathway. NCCs migrate alongside the posterior surface of developing intersomitic vessels, but fail to cross these continuous streams of motile ECs. NCCs change their morphology and migration trajectory when they encounter gaps in the developing vasculature. Within the nascent dorsal root ganglion, proximity to ECs causes filopodial retraction which curtails forward persistence of NCC motility. Overall, our time-lapse recordings support the conclusion that primary vascular networks substantially influence the distribution and migratory behavior of NCCs and the patterned formation of dorsal root and sympathetic ganglia.


Asunto(s)
Células Endoteliales/citología , Ganglios Espinales/embriología , Microscopía/métodos , Cresta Neural/embriología , Sistema Nervioso Periférico/embriología , Sistema Nervioso Simpático/embriología , Imagen de Lapso de Tiempo/métodos , Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Bacterianas/metabolismo , Tipificación del Cuerpo , Comunicación Celular , Movimiento Celular , Coturnix , Ganglios Espinales/citología , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Proteínas Luminiscentes/metabolismo , Cresta Neural/citología , Células Madre/citología
20.
Nat Commun ; 6: 8281, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26404565

RESUMEN

The sympathetic nervous system is essential for maintaining mammalian homeostasis. How this intricately connected network, composed of preganglionic neurons that reside in the spinal cord and post-ganglionic neurons that comprise a chain of vertebral sympathetic ganglia, arises developmentally is incompletely understood. This problem is especially complex given the vertebral chain of sympathetic ganglia derive secondarily from the dorsal migration of 'primary' sympathetic ganglia that are initially located several hundred microns ventrally from their future pre-synaptic partners. Here we report that the dorsal migration of discrete ganglia is not a simple migration of individual cells but a much more carefully choreographed process that is mediated by extensive interactions of pre-and post-ganglionic neurons. Dorsal migration does not occur in the absence of contact with preganglionic axons, and this is mediated by BDNF/TrkB signalling. Thus BDNF released by preganglionic axons acts chemotactically on TrkB-positive sympathetic neurons, to pattern the developing peripheral nervous system.


Asunto(s)
Fibras Autónomas Preganglionares/metabolismo , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Quimiotaxis , Ganglios Simpáticos/metabolismo , Receptor trkB/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Movimiento Celular , Embrión de Pollo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Microscopía Confocal , Receptor trkB/genética , Transducción de Señal , Médula Espinal , Sistema Nervioso Simpático , Imagen de Lapso de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA