Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Neurosci ; 41(17): 3932-3943, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33741721

RESUMO

The Abelson-helper integration site 1 (AHI1) gene encodes for a ciliary transition zone localizing protein that when mutated causes the human ciliopathy, Joubert syndrome. We prepared and examined neuronal cultures derived from male and female embryonic Ahi1+/+ and Ahi1-/- mice (littermates) and found that the distribution of ciliary melanin-concentrating hormone receptor-1 (MchR1) was significantly reduced in Ahi1-/- neurons; however, the total and surface expression of MchR1 on Ahi1-/- neurons was similar to controls (Ahi1+/+). This indicates that a pathway for MchR1 trafficking to the surface plasma membrane is intact, but the process of targeting MchR1 into cilia is impaired in Ahi1-deficient mouse neurons, indicating a role for Ahi1 in localizing MchR1 to the cilium. Mouse Ahi1-/- neurons that fail to accumulate MchR1 in the ciliary membrane have significant decreases in two downstream MchR1 signaling pathways [cAMP and extracellular signal-regulated kinase (Erk)] on MCH stimulation. These results suggest that the ciliary localization of MchR1 is necessary and critical for MchR1 signaling, with Ahi1 participating in regulating MchR1 localization to cilia, and further supporting cilia as critical signaling centers in neurons.SIGNIFICANCE STATEMENT Our work here demonstrates that neuronal primary cilia are powerful and focused signaling centers for the G-protein-coupled receptor (GPCR), melanin-concentrating hormone receptor-1 (MCHR1), with a role for the ciliary transition zone protein, Abelson-helper integration site 1 (AHI1), in mediating ciliary trafficking of MCHR1. Moreover, our manuscript further expands the repertoire of cilia functions on neurons, a cell type that has not received significant attention in the cilia field. Lastly, our work demonstrates the significant influence of ciliary GPCR signaling in the overall signaling of neurons.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Cílios/fisiologia , Neurônios/fisiologia , Receptores de Somatostatina/fisiologia , Transdução de Sinais/fisiologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Membrana Celular/fisiologia , Cerebelo/anormalidades , Cerebelo/fisiopatologia , AMP Cíclico/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Feminino , Doenças Renais Císticas/genética , Doenças Renais Císticas/fisiopatologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Gravidez , Receptores de Somatostatina/genética , Retina/anormalidades , Retina/fisiopatologia , Transdução de Sinais/genética
2.
FASEB J ; 35(10): e21869, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34469026

RESUMO

The leucine-rich repeat-containing family 8 member A (LRRC8A) is an essential subunit of the volume-regulated anion channel (VRAC). VRAC is critical for cell volume control, but its broader physiological functions remain under investigation. Recent studies in the field indicate that Lrrc8a disruption in the brain astrocytes reduces neuronal excitability, impairs synaptic plasticity and memory, and protects against cerebral ischemia. In the present work, we generated brain-wide conditional LRRC8A knockout mice (LRRC8A bKO) using NestinCre -driven Lrrc8aflox/flox excision in neurons, astrocytes, and oligodendroglia. LRRC8A bKO animals were born close to the expected Mendelian ratio and developed without overt histological abnormalities, but, surprisingly, all died between 5 and 9 weeks of age with a seizure phenotype, which was confirmed by video and EEG recordings. Brain slice electrophysiology detected changes in the excitability of pyramidal cells and modified GABAergic inputs in the hippocampal CA1 region of LRRC8A bKO. LRRC8A-null hippocampi showed increased immunoreactivity of the astrocytic marker GFAP, indicating reactive astrogliosis. We also found decreased whole-brain protein levels of the GABA transporter GAT-1, the glutamate transporter GLT-1, and the astrocytic enzyme glutamine synthetase. Complementary HPLC assays identified reduction in the tissue levels of the glutamate and GABA precursor glutamine. Together, these findings suggest that VRAC provides vital control of brain excitability in mouse adolescence. VRAC deletion leads to a lethal phenotype involving progressive astrogliosis and dysregulation of astrocytic uptake and supply of amino acid neurotransmitters and their precursors.


Assuntos
Astrócitos/patologia , Gliose/mortalidade , Ácido Glutâmico/metabolismo , Proteínas de Membrana/fisiologia , Convulsões/mortalidade , Animais , Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Feminino , Gliose/etiologia , Gliose/patologia , Transporte de Íons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Convulsões/etiologia , Convulsões/patologia
3.
J Cell Sci ; 132(17)2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31391239

RESUMO

Mutations in the Abelson-helper integration site 1 (AHI1) gene are associated with neurological/neuropsychiatric disorders, and cause the neurodevelopmental ciliopathy Joubert syndrome (JBTS). Here, we show that deletion of the transition zone (TZ) protein Ahi1 in mouse embryonic fibroblasts (MEFs) has a small effect on cilia formation. However, Ahi1 loss in these cells results in: (1) reduced localization of the JBTS-associated protein Arl13b to the ciliary membrane, (2) decreased sonic hedgehog signaling, (3) and an abnormally elongated ciliary axoneme accompanied by an increase in ciliary IFT88 concentrations. While no changes in Arl13b levels are detected in crude cell membrane extracts, loss of Ahi1 significantly reduced the level of non-membrane-associated Arl13b and its stability via the proteasome pathway. Exogenous expression of Ahi1-GFP in Ahi1-/- MEFs restored ciliary length, increased ciliary recruitment of Arl13b and augmented Arl13b stability. Finally, Ahi1-/- MEFs displayed defects in cell motility and Pdgfr-α-dependent migration. Overall, our findings support molecular mechanisms underlying JBTS etiology that involve: (1) disruptions at the TZ resulting in defects of membrane- and non-membrane-associated proteins to localize to primary cilia, and (2) defective cell migration.This article has an associated First Person interview with the first author of the paper.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Cílios/metabolismo , Fatores de Ribosilação do ADP/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Movimento Celular/fisiologia , Fibroblastos/metabolismo , Camundongos , Camundongos Knockout , Mutação , Transporte Proteico , Transdução de Sinais
4.
Dev Biol ; 448(1): 36-47, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30695685

RESUMO

Joubert syndrome (JBTS) is a predominantly autosomal recessive neurodevelopmental disorder that presents with characteristic malformations of the cerebellar vermis, superior cerebellar peduncles and midbrain in humans. Accompanying these malformations are a heterogeneous set of clinical symptoms, which frequently include deficits in motor and muscle function, such as hypotonia (low muscle tone) and ataxia (clumsiness). These symptoms are attributed to improper development of the hindbrain, but no direct evidence has been reported linking these in JBTS. Here, we describe muscle developmental defects in a mouse with a targeted deletion of the Abelson helper integration site 1 gene, Ahi1, one of the genes known to cause JBTS in humans. While FVB/NJ Ahi1-/- mice display no gross malformations of the cerebellum, deficits are observed in several measures of motor function, strength, and body development. Specifically, Ahi1-/- mice show delayed physical development, delays in surface reflex righting as neonates, and reductions in grip strength and spontaneous locomotor activity as adults. Additionally, Ahi1-/- mice showed evidence of muscle-specific contributions to this phenotype, such as reductions in 1) myoblast differentiation potential in vitro, 2) muscle desmin expression, and 3) overall muscle mass, myonuclear domain, and muscle fiber cross-sectional area. Together, these data suggest that loss of Ahi1 may cause abnormalities in the differentiation of myoblasts to mature muscle cells. Moreover, Ahi1 loss impacts muscle development directly, outside of any indirect impact of cerebellar malformations, revealing a novel myogenic cause for hypotonia in JBTS.


Assuntos
Anormalidades Múltiplas/embriologia , Diferenciação Celular , Cerebelo/anormalidades , Anormalidades do Olho/embriologia , Doenças Renais Císticas/embriologia , Desenvolvimento Muscular , Transtornos do Neurodesenvolvimento/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Retina/anormalidades , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Proteínas Adaptadoras de Transporte Vesicular , Animais , Cerebelo/embriologia , Cerebelo/patologia , Desmina/genética , Desmina/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Locomoção/genética , Camundongos , Camundongos Knockout , Força Muscular/genética , Mioblastos/metabolismo , Mioblastos/patologia , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Proteínas Proto-Oncogênicas/metabolismo , Reflexo de Endireitamento/genética , Retina/embriologia , Retina/patologia
5.
Bioessays ; 40(8): e1700132, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29882973

RESUMO

Once dismissed as vestigial organelles, primary cilia have garnered the interest of scientists, given their importance in development/signaling, and for their implication in a new disease category known as ciliopathies. However, many, if not all, "cilia" proteins also have locations/functions outside of the primary cilium. These extraciliary functions can complicate the interpretation of a particular ciliopathy phenotype: it may be a result of defects at the cilium and/or at extraciliary locations, and it could be broadly related to a unifying cellular process for these proteins, such as polarity. Assembly of a cilium has many similarities to the development of other polarized structures. This evolutionarily preserved process for the assembly of polarized cell structures offers a perspective on how the cilium may have evolved. We hypothesize that cilia proteins are critical for cell polarity, and that core polarity proteins may have been specialized to form various cellular protrusions, including primary cilia.


Assuntos
Polaridade Celular/fisiologia , Cílios/metabolismo , Cílios/patologia , Ciliopatias/patologia , Proteínas/metabolismo , Animais , Evolução Biológica , Centrossomo/metabolismo , Ciliopatias/etiologia , Citoesqueleto/metabolismo , Dendritos/metabolismo , Humanos , Neuritos/fisiologia , Fosfatidilinositóis/metabolismo , Transdução de Sinais
6.
Cell Mol Life Sci ; 75(9): 1521-1540, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29305615

RESUMO

Primary cilia are immotile organelles known for their roles in development and cell signaling. Defects in primary cilia result in a range of disorders named ciliopathies. Because this organelle can be found singularly on almost all cell types, its importance extends to most organ systems. As such, elucidating the importance of the primary cilium has attracted researchers from all biological disciplines. As the primary cilia field expands, caution is warranted in attributing biological defects solely to the function of this organelle, since many of these "ciliary" proteins are found at other sites in cells and likely have non-ciliary functions. Indeed, many, if not all, cilia proteins have locations and functions outside the primary cilium. Extraciliary functions are known to include cell cycle regulation, cytoskeletal regulation, and trafficking. Cilia proteins have been observed in the nucleus, at the Golgi apparatus, and even in immune synapses of T cells (interestingly, a non-ciliated cell). Given the abundance of extraciliary sites and functions, it can be difficult to definitively attribute an observed phenotype solely to defective cilia rather than to some defective extraciliary function or a combination of both. Thus, extraciliary sites and functions of cilia proteins need to be considered, as well as experimentally determined. Through such consideration, we will understand the true role of the primary cilium in disease as compared to other cellular processes' influences in mediating disease (or through a combination of both). Here, we review a compilation of known extraciliary sites and functions of "cilia" proteins as a means to demonstrate the potential non-ciliary roles for these proteins.


Assuntos
Cílios/metabolismo , Proteínas/metabolismo , Animais , Movimento Celular/fisiologia , Humanos , Fenótipo , Transdução de Sinais/fisiologia
7.
J Neurosci ; 36(28): 7485-96, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27413158

RESUMO

UNLABELLED: The occurrence of recurrent, unprovoked seizures is the hallmark of human epilepsy. Currently, only two-thirds of this patient population has adequate seizure control. New epilepsy models provide the potential for not only understanding the development of spontaneous seizures, but also for testing new strategies to treat this disorder. Here, we characterize a primary generalized seizure model of epilepsy following repeated exposure to the GABAA receptor antagonist, flurothyl, in which mice develop spontaneous seizures that remit within 1 month. In this model, we expose C57BL/6J mice to flurothyl until they experience a generalized seizure. Each of these generalized seizures typically lasts <30 s. We induce one seizure per day for 8 d followed by 24 h video-electroencephalographic recordings. Within 1 d following the last of eight flurothyl-induced seizures, ∼50% of mice have spontaneous seizures. Ninety-five percent of mice tested have seizures within the first week of the recording period. Of the spontaneous seizures recorded, the majority are generalized clonic seizures, with the remaining 7-12% comprising generalized clonic seizures that transition into brainstem seizures. Over the course of an 8 week recording period, spontaneous seizure episodes remit after ∼4 weeks. Overall, the repeated flurothyl paradigm is a model of epileptogenesis with spontaneous seizures that remit. This model provides an additional tool in our armamentarium for understanding the mechanisms underlying epileptogenesis and may provide insights into why spontaneous seizures remit without anticonvulsant treatment. Elucidating these processes could lead to the development of new epilepsy therapeutics. SIGNIFICANCE STATEMENT: Epilepsy is a chronic disorder characterized by the occurrence of recurrent, unprovoked seizures in which the individual seizure-ictal events are self-limiting. Remission of recurrent, unprovoked seizures can be achieved in two-thirds of cases by treatment with anticonvulsant medication, surgical resection, and/or nerve/brain electrode stimulation. However, there are examples in humans of epilepsy with recurrent, unprovoked seizures remitting without any intervention. While elucidating how recurrent, unprovoked seizures develop is critical for understanding epileptogenesis, an understanding of how and why recurrent, unprovoked seizures remit may further our understanding and treatment of epilepsy. Here, we describe a new model of recurrent, unprovoked spontaneous seizures in which the occurrence of spontaneous seizures naturally remits over time without any therapeutic intervention.


Assuntos
Convulsivantes/toxicidade , Flurotila/toxicidade , Convulsões/induzido quimicamente , Análise de Variância , Animais , Anticonvulsivantes/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Eletroencefalografia , Fluoresceínas/metabolismo , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/tratamento farmacológico , Convulsões/patologia , Fatores de Tempo , Gravação em Vídeo
8.
Am J Hum Genet ; 94(1): 62-72, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24360808

RESUMO

Joubert syndrome (JBTS) is a recessive ciliopathy in which a subset of affected individuals also have the skeletal dysplasia Jeune asphyxiating thoracic dystrophy (JATD). Here, we have identified biallelic truncating CSPP1 (centrosome and spindle pole associated protein 1) mutations in 19 JBTS-affected individuals, four of whom also have features of JATD. CSPP1 mutations explain ∼5% of JBTS in our cohort, and despite truncating mutations in all affected individuals, the range of phenotypic severity is broad. Morpholino knockdown of cspp1 in zebrafish caused phenotypes reported in other zebrafish models of JBTS (curved body shape, pronephric cysts, and cerebellar abnormalities) and reduced ciliary localization of Arl13b, further supporting loss of CSPP1 function as a cause of JBTS. Fibroblasts from affected individuals with CSPP1 mutations showed reduced numbers of primary cilia and/or short primary cilia, as well as reduced axonemal localization of ciliary proteins ARL13B and adenylyl cyclase III. In summary, CSPP1 mutations are a major cause of the Joubert-Jeune phenotype in humans; however, the mechanism by which these mutations lead to both JBTS and JATD remains unknown.


Assuntos
Proteínas de Ciclo Celular/genética , Doenças Cerebelares/genética , Cílios/genética , Síndrome de Ellis-Van Creveld/genética , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação , Retina/anormalidades , Anormalidades Múltiplas , Adolescente , Animais , Cerebelo/anormalidades , Criança , Pré-Escolar , Cílios/patologia , Éxons , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Lactente , Masculino , Fenótipo , Análise de Sequência de DNA , Adulto Jovem , Peixe-Zebra/genética
9.
Epilepsy Behav ; 73: 214-235, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28651171

RESUMO

It is becoming increasingly clear that the genetic background of mice and rats, even in inbred strains, can have a profound influence on measures of seizure susceptibility and epilepsy. These differences can be capitalized upon through genetic mapping studies to reveal genes important for seizures and epilepsy. However, strain background and particularly mixed genetic backgrounds of transgenic animals need careful consideration in both the selection of strains and in the interpretation of results and conclusions. For instance, mice with targeted deletions of genes involved in epilepsy can have profoundly disparate phenotypes depending on the background strain. In this review, we discuss findings related to how this genetic heterogeneity has and can be utilized in the epilepsy field to reveal novel insights into seizures and epilepsy. Moreover, we discuss how caution is needed in regards to rodent strain or even animal vendor choice, and how this can significantly influence seizure and epilepsy parameters in unexpected ways. This is particularly critical in decisions regarding the strain of choice used in generating mice with targeted deletions of genes. Finally, we discuss the role of environment (at vendor and/or laboratory) and epigenetic factors for inter- and intrastrain differences and how such differences can affect the expression of seizures and the animals' performance in behavioral tests that often accompany acute and chronic seizure testing.


Assuntos
Modelos Animais de Doenças , Epilepsia/genética , Convulsões/genética , Animais , Masculino , Camundongos , Ratos , Especificidade da Espécie
10.
Hum Mol Genet ; 23(17): 4663-73, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24760772

RESUMO

Filamin B (FlnB) is an actin-binding protein thought to transduce signals from various membrane receptors and intracellular proteins onto the actin cytoskeleton. Formin1 (Fmn1) is an actin-nucleating protein, implicated in actin assembly and intracellular signaling. Human mutations in FLNB cause several skeletal disorders associated with dwarfism and early bone fusion. Mouse mutations in Fmn1 cause aberrant fusion of carpal digits. We report here that FlnB and Fmn1 physically interact, are co-expressed in chondrocytes in the growth plate and share overlapping expression in the cell cytoplasm and nucleus. Loss of FlnB leads to a dramatic decrease in Fmn1 expression at the hypertrophic-to-ossification border. Loss of Fmn1-FlnB in mice leads to a more severe reduction in body size, weight and growth plate length, than observed in mice following knockout of either gene alone. Shortening of the long bone is associated with a decrease in chondrocyte proliferation and an overall delay in ossification in the double-knockout mice. In contrast to FlnB null, Fmn1 loss results in a decrease in the width of the prehypertrophic zone. Loss of both proteins, however, causes an overall decrease in the width of the proliferation zone and an increase in the differentiated hypertrophic zone. The current findings suggest that Fmn1 and FlnB have shared and independent functions. FlnB loss promotes prehypertrophic differentiation whereas Fmn1 leads to a delay. Both proteins, however, regulate chondrocyte proliferation, and FlnB may regulate Fmn1 function at the hypertrophic-to-ossification border, thereby explaining the overall delay in ossification.


Assuntos
Diferenciação Celular , Condrócitos/metabolismo , Condrócitos/patologia , Proteínas Fetais/metabolismo , Filaminas/metabolismo , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/patologia , Proteínas dos Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Animais , Calcificação Fisiológica , Proliferação de Células , Proteínas Fetais/deficiência , Filaminas/deficiência , Forminas , Humanos , Hipertrofia , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas Nucleares/deficiência , Ligação Proteica , Transporte Proteico , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
11.
J Biol Chem ; 288(19): 13676-94, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23532844

RESUMO

BACKGROUND: Missense mutations in AHI1 result in the neurodevelopmental ciliopathy called Joubert syndrome. RESULTS: Mutations in AHI1 decrease cilia formation, alter its localization and stability, and change its binding to HAP1 and NPHP1. CONCLUSION: Mutations in AHI1 affect ciliogenesis, AHI1 protein localization, and AHI1-protein interactions. SIGNIFICANCE: This study begins to describe how missense mutations in AHI1 can cause Joubert syndrome. Mutations in AHI1 cause Joubert syndrome (JBTS), a neurodevelopmental ciliopathy, characterized by midbrain-hindbrain malformations and motor/cognitive deficits. Here, we show that primary cilia (PC) formation is decreased in fibroblasts from individuals with JBTS and AHI1 mutations. Most missense mutations in AHI1, causing JBTS, occur in known protein domains, however, a common V443D mutation in AHI1 is found in a region with no known protein motifs. We show that cells transfected with AHI1-V443D, or a new JBTS-causing mutation, AHI1-R351L, have aberrant localization of AHI1 at the basal bodies of PC and at cell-cell junctions, likely through decreased binding of mutant AHI1 to NPHP1 (another JBTS-causing protein). The AHI1-V443D mutation causes decreased AHI1 stability because there is a 50% reduction in AHI1-V443D protein levels compared with wild type AHI1. Huntingtin-associated protein-1 (Hap1) is a regulatory protein that binds Ahi1, and Hap1 knock-out mice have been reported to have JBTS-like phenotypes, suggesting a role for Hap1 in ciliogenesis. Fibroblasts and neurons with Hap1 deficiency form PC with normal growth factor-induced ciliary signaling, indicating that the Hap1 JBTS phenotype is likely not through effects at PC. These results also suggest that the binding of Ahi1 and Hap1 may not be critical for ciliary function. However, we show that HAP1 has decreased binding to AHI1-V443D indicating that this altered binding could be responsible for the JBTS-like phenotype through an unknown pathway. Thus, these JBTS-associated missense mutations alter their subcellular distribution and protein interactions, compromising functions of AHI1 in cell polarity and cilium-mediated signaling, thereby contributing to JBTS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doenças Cerebelares/genética , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Mutação de Sentido Incorreto , Anormalidades Múltiplas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Polaridade Celular , Células Cultivadas , Doenças Cerebelares/metabolismo , Doenças Cerebelares/patologia , Cerebelo/anormalidades , Cílios/metabolismo , Cílios/patologia , Sequência Conservada , Proteínas do Citoesqueleto , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Junções Intercelulares/metabolismo , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapas de Interação de Proteínas , Estabilidade Proteica , Transporte Proteico , Retina/anormalidades , Retina/metabolismo , Retina/patologia , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
12.
J Neurosci ; 32(36): 12619-29, 2012 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-22956851

RESUMO

Periventricular heterotopia (PH) is a human malformation of cortical development associated with gene mutations in ADP-ribosylation factor guanine exchange factor 2 (ARFGEF2 encodes for Big2 protein) and Filamin A (FLNA). PH is thought to derive from neuroependymal disruption, but the extent to which neuronal migration contributes to this phenotype is unknown. Here, we show that Arfgef2 null mice develop PH and exhibit impaired neural migration with increased protein expression for both FlnA and phosphoFlnA at Ser2152. Big2 physically interacts with FlnA and overexpression of phosphomimetic Ser2512 FLNA impairs neuronal migration. FlnA phosphorylation directs FlnA localization toward the cell cytoplasm, diminishes its binding affinity to actin skeleton, and alters the number and size of paxillin focal adhesions. Collectively, our results demonstrate a molecular mechanism whereby Big2 inhibition promotes phosphoFlnA (Ser2152) expression, and increased phosphoFlnA impairs its actin binding affinity and the distribution of focal adhesions, thereby disrupting cell intrinsic neuronal migration.


Assuntos
Movimento Celular/fisiologia , Proteínas Contráteis/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Proteínas dos Microfilamentos/metabolismo , Neurônios/fisiologia , Animais , Feminino , Filaminas , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/fisiologia
13.
J Neurosci ; 32(22): 7672-84, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649246

RESUMO

Cytoskeleton-associated proteins play key roles not only in regulating cell morphology and migration but also in proliferation. Mutations in the cytoskeleton-associated gene filamin A (FlnA) cause the human disorder periventricular heterotopia (PH). PH is a disorder of neural stem cell development that is characterized by disruption of progenitors along the ventricular epithelium and subsequent formation of ectopic neuronal nodules. FlnA-dependent regulation of cytoskeletal dynamics is thought to direct neural progenitor migration and proliferation. Here we show that embryonic FlnA-null mice exhibited a reduction in brain size and decline in neural progenitor numbers over time. The drop in the progenitor population was not attributable to cell death or changes in premature differentiation, but to prolonged cell cycle duration. Suppression of FlnA led to prolongation of the entire cell cycle length, principally in M phase. FlnA loss impaired degradation of cyclin B1-related proteins, thereby delaying the onset and progression through mitosis. We found that the cdk1 kinase Wee1 bound FlnA, demonstrated increased expression levels after loss of FlnA function, and was associated with increased phosphorylation of cdk1. Phosphorylation of cdk1 inhibited activation of the anaphase promoting complex degradation system, which was responsible for cyclin B1 degradation and progression through mitosis. Collectively, our results demonstrate a molecular mechanism whereby FlnA loss impaired G2 to M phase entry, leading to cell cycle prolongation, compromised neural progenitor proliferation, and reduced brain size.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Córtex Cerebral/fisiologia , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Proteína Quinase CDC2/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Proteínas Contráteis/deficiência , Ciclina B1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos , Filaminas , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica/genética , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67 , Camundongos , Camundongos Transgênicos , Microcefalia/genética , Proteínas dos Microfilamentos/deficiência , Heterotopia Nodular Periventricular/genética , Heterotopia Nodular Periventricular/patologia , Fosforilação/genética , Proteínas com Domínio T/metabolismo , Tirosina/metabolismo
14.
Nat Genet ; 36(9): 1008-13, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15322546

RESUMO

Joubert syndrome is a congenital brain malformation of the cerebellar vermis and brainstem with abnormalities of axonal decussation (crossing in the brain) affecting the corticospinal tract and superior cerebellar peduncles. Individuals with Joubert syndrome have motor and behavioral abnormalities, including an inability to walk due to severe clumsiness and 'mirror' movements, and cognitive and behavioral disturbances. Here we identified a locus associated with Joubert syndrome, JBTS3, on chromosome 6q23.2-q23.3 and found three deleterious mutations in AHI1, the first gene to be associated with Joubert syndrome. AHI1 is most highly expressed in brain, particularly in neurons that give rise to the crossing axons of the corticospinal tract and superior cerebellar peduncles. Comparative genetic analysis of AHI1 indicates that it has undergone positive evolutionary selection along the human lineage. Therefore, changes in AHI1 may have been important in the evolution of human-specific motor behaviors.


Assuntos
Anormalidades Múltiplas/genética , Cerebelo/anormalidades , Deficiências do Desenvolvimento/genética , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Animais , Encéfalo/anormalidades , Encéfalo/embriologia , Encéfalo/metabolismo , Tronco Encefálico/anormalidades , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Filogenia , Síndrome
15.
Methods Cell Biol ; 176: 43-57, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37164542

RESUMO

Primary cilia are complex organelles, usually singularly located on cell surfaces that are now known to be important for signaling and whose defect is implicated in a category of developmental diseases known as ciliopathies. They are composed of a microtubule axoneme and contain a cilia membrane that is unique and distinct from the plasma membrane. Primary cilia also have their own transport system termed the intraflagellar transport (IFT) system that allows for proteins to be trafficked along the microtubule axoneme in either an anterograde or retrograde manner. Proteins that localize to the primary cilium are referred to as ciliary proteins and have been implicated directly or indirectly in ciliogenesis or ciliary function. It is now recognized that cilia proteins can localize to different compartments of cilia, but can also localize to multiple sites outside of cilia (extraciliary sites). This complexity results in a need for a better understanding of ciliary protein fixation and immunolabeling protocols, as different methods are required to visualize different cilia proteins and reveal novel or unique localizations. Here, we detail a variety of fixation methods and their effects on ciliary protein immunolabeling.


Assuntos
Cílios , Proteínas , Cílios/metabolismo , Transporte Biológico , Microtúbulos/metabolismo , Membrana Celular/metabolismo
16.
J Neurosci ; 30(26): 8759-68, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20592197

RESUMO

Vertebrate photoreceptors have a modified cilium composed of a basal body, axoneme and outer segment. The outer segment includes stacked membrane discs, containing opsin and the signal transduction apparatus mediating phototransduction. In photoreceptors, two distinct classes of vesicles are trafficked. Synaptic vesicles are transported down the axon to the synapse, whereas opsin-containing vesicles are transported to the outer segment. The continuous replacement of the outer segments imposes a significant biosynthetic and trafficking burden on the photoreceptors. Here, we show that Ahi1, a gene that when mutated results in the neurodevelopmental disorder, Joubert syndrome (JBTS), is required for photoreceptor sensory cilia formation and the development of photoreceptor outer segments. In mice with a targeted deletion of Ahi1, photoreceptors undergo early degeneration. Whereas synaptic proteins are correctly trafficked, photoreceptor outer segment proteins fail to be transported appropriately or are significantly reduced in their expression levels (i.e., transducin and Rom1) in Ahi1(-/-) mice. We show that vesicular targeting defects in Ahi1(-/-) mice are cilium specific, and our evidence suggests that the defects are caused by a decrease in expression of the small GTPase Rab8a, a protein required for accurate polarized vesicular trafficking. Thus, our results suggest that Ahi1 plays a role in stabilizing the outer segment proteins, transducin and Rom1, and that Ahi1 is an important component of Rab8a-mediated vesicular trafficking in photoreceptors. The retinal degeneration observed in Ahi1(-/-) mice recapitulates aspects of the retinal phenotype observed in patients with JBTS and suggests the importance of Ahi1 in photoreceptor function.


Assuntos
Proteínas Proto-Oncogênicas/metabolismo , Degeneração Retiniana/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Encefalopatias , Cílios/metabolismo , Proteínas do Olho/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Deleção de Genes , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Retina/metabolismo , Vesículas Sinápticas/metabolismo , Síndrome , Tetraspaninas , Transducina/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
17.
Hum Mol Genet ; 18(24): 4853-67, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19783549

RESUMO

Historical episodes of natural selection can skew the frequencies of genetic variants, leaving a signature that can persist for many tens or even hundreds of thousands of years. However, formal tests for selection based on allele frequency skew require strong assumptions about demographic history and mutation, which are rarely well understood. Here, we develop an empirical approach to test for signals of selection that compares patterns of genetic variation at a candidate locus with matched random regions of the genome collected in the same way. We apply this approach to four genes that have been implicated in syndromes of impaired neurological development, comparing the pattern of variation in our re-sequencing data with a large-scale, genomic data set that provides an empirical null distribution. We confirm a previously reported signal at FOXP2, and find a novel signal of selection centered at AHI1, a gene that is involved in motor and behavior abnormalities. The locus is marked by many high frequency derived alleles in non-Africans that are of low frequency in Africans, suggesting that selection at this or a closely neighboring gene occurred in the ancestral population of non-Africans. Our study also provides a prototype for how empirical scans for ancient selection can be carried out once many genomes are sequenced.


Assuntos
Simulação por Computador , Genoma Humano , Modelos Genéticos , Polimorfismo de Nucleotídeo Único , Seleção Genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular , Fatores de Transcrição Forkhead/genética , Frequência do Gene , Haplótipos , Humanos , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Receptores Acoplados a Proteínas G/genética , Análise de Sequência de DNA
18.
Hum Mol Genet ; 18(20): 3926-41, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19625297

RESUMO

The primary non-motile cilium, a membrane-ensheathed, microtubule-bundled organelle, extends from virtually all cells and is important for development. Normal functioning of the cilium requires proper axoneme assembly, membrane biogenesis and ciliary protein localization, in tight coordination with the intraflagellar transport system and vesicular trafficking. Disruptions at any level can induce severe alterations in cell function, giving rise to a myriad of human genetic diseases known as ciliopathies. Here we show that the Abelson helper integration site 1 (Ahi1) gene, whose human ortholog is mutated in Joubert syndrome, regulates cilium formation via its interaction with Rab8a, a small GTPase critical for polarized membrane trafficking. We find that the Ahi1 protein localizes to a single centriole, the mother centriole, which becomes the basal body of the primary cilium. In order to determine whether Ahi1 functions in ciliogenesis, loss of function analysis of Ahi1 was performed in cell culture models of ciliogenesis. Knockdown of Ahi1 expression by shRNAi in cells or targeted deletion of Ahi1 (Ahi1 knockout mouse) leads to impairments in ciliogenesis. In Ahi1-knockdown cells, Rab8a is destabilized and does not properly localize to the basal body. Since Rab8a is implicated in vesicular trafficking, we next examined this process in Ahi1-knockdown cells. Defects in the trafficking of endocytic vesicles from the plasma membrane to the Golgi and back to the plasma membrane were observed in Ahi1-knockdown cells. Overall, our data indicate that the distribution and functioning of Rab8a is regulated by Ahi1, not only affecting cilium formation, but also vesicle transport.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Cílios/metabolismo , Mutação , Doenças do Sistema Nervoso/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Vesículas Transportadoras/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Linhagem Celular , Células Cultivadas , Cílios/genética , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças do Sistema Nervoso/genética , Ligação Proteica , Transporte Proteico , Proteínas Proto-Oncogênicas/genética , Proteínas rab de Ligação ao GTP/genética
19.
Hum Mol Genet ; 18(3): 497-516, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18996916

RESUMO

Periventricular heterotopia (PH) is a disorder characterized by neuronal nodules, ectopically positioned along the lateral ventricles of the cerebral cortex. Mutations in either of two human genes, Filamin A (FLNA) or ADP-ribosylation factor guanine exchange factor 2 (ARFGEF2), cause PH (Fox et al. in 'Mutations in filamin 1 prevent migration of cerebral cortical neurons in human periventricular heterotopia'. Neuron, 21, 1315-1325, 1998; Sheen et al. in 'Mutations in ARFGEF2 implicate vesicle trafficking in neural progenitor proliferation and migration in the human cerebral cortex'. Nat. Genet., 36, 69-76, 2004). Recent studies have shown that mutations in mitogen-activated protein kinase kinase kinase-4 (Mekk4), an indirect interactor with FlnA, also lead to periventricular nodule formation in mice (Sarkisian et al. in 'MEKK4 signaling regulates filamin expression and neuronal migration'. Neuron, 52, 789-801, 2006). Here we show that neurons in post-mortem human PH brains migrated appropriately into the cortex, that periventricular nodules were primarily composed of later-born neurons, and that the neuroependyma was disrupted in all PH cases. As studied in the mouse, loss of FlnA or Big2 function in neural precursors impaired neuronal migration from the germinal zone, disrupted cell adhesion and compromised neuroepithelial integrity. Finally, the hydrocephalus with hop gait (hyh) mouse, which harbors a mutation in Napa [encoding N-ethylmaleimide-sensitive factor attachment protein alpha (alpha-SNAP)], also develops a progressive denudation of the neuroepithelium, leading to periventricular nodule formation. Previous studies have shown that Arfgef2 and Napa direct vesicle trafficking and fusion, whereas FlnA associates dynamically with the Golgi membranes during budding and trafficking of transport vesicles. Our current findings suggest that PH formation arises from a final common pathway involving disruption of vesicle trafficking, leading to impaired cell adhesion and loss of neuroependymal integrity.


Assuntos
Ventrículos Cerebrais/citologia , Heterotopia Nodular Periventricular/patologia , Células-Tronco/citologia , Adulto , Idoso de 80 Anos ou mais , Animais , Adesão Celular , Movimento Celular , Ventrículos Cerebrais/fisiopatologia , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Feminino , Filaminas , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Recém-Nascido , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Neurônios/fisiologia , Heterotopia Nodular Periventricular/fisiopatologia , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/genética , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo
20.
J Neurosci Methods ; 331: 108529, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31760060

RESUMO

BACKGROUND: Sholl analysis has been used to analyze neuronal morphometry and dendritic branching and complexity for many years. While the process has become semi-automated in recent years, existing software packages are still dependent on user tracing and hence are subject to observer bias, variability, and increased user times for analyses. Commercial software packages have the same issues as they also rely on user tracing. In addition, these packages are also expensive and require extensive user training. NEW METHOD: To address these issues, we have developed a broadly applicable, no-cost ImageJ plugin, we call AutoSholl, to perform Sholl analysis on pre-processed and 'thresholded' images. This algorithm extends the already existing plugin in Fiji ImageJ for Sholl analysis by allowing for secondary analysis techniques, such as determining number and length of root, intermediate, and terminal dendrites; functions not currently supported in the existing Sholl Analysis plugin in Fiji ImageJ. RESULTS: The algorithm allows for rapid Sholl analysis in both 2-dimensional and 3-dimensional data sets independent of user tracing. COMPARISON WITH EXISTING METHODS: We validated the performance of AutoSholl against pre-existing software packages using trained human observers and images of neurons. We found that our algorithm outputs similar results as available software (i.e., Bonfire), but allows for faster analysis times and unbiased quantification. CONCLUSIONS: As such, AutoSholl allows inexperienced observers to output results like more trained observers efficiently, thereby increasing the consistency, speed, and reliability of Sholl analyses.


Assuntos
Neurônios , Software , Algoritmos , Automação , Humanos , Processamento de Imagem Assistida por Computador , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA