Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
STAR Protoc ; 5(1): 102904, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38427568

RESUMEN

Neurodevelopmental disorders are characterized by complex phenotypes that often result from concomitant dysregulation of cell proliferation, differentiation, or other crucial developmental processes. Here, we present a protocol to quantify differentiation of progenitor populations during early stages of neurogenesis in induced pluripotent stem cell (iPSC)-derived cerebral organoids. We describe steps for organoid differentiation and maturation, sample preparation, immunofluorescence, and imaging and analysis using epifluorescence microscopy. This protocol can be used to compare cerebral organoids from control and patient-derived iPSCs. For complete details on the use and execution of this protocol, please refer to Rakotomamonjy et al. (2023).1.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trastornos del Neurodesarrollo , Humanos , Diferenciación Celular/genética , Neurogénesis/genética , Organoides
2.
Nat Commun ; 15(1): 827, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-38280846

RESUMEN

PACS1 syndrome is a neurodevelopmental disorder characterized by intellectual disability and distinct craniofacial abnormalities resulting from a de novo p.R203W variant in phosphofurin acidic cluster sorting protein 1 (PACS1). PACS1 is known to have functions in the endosomal pathway and nucleus, but how the p.R203W variant affects developing neurons is not fully understood. Here we differentiated stem cells towards neuronal models including cortical organoids to investigate the impact of the PACS1 syndrome-causing variant on neurodevelopment. While few deleterious effects were detected in PACS1(+/R203W) neural precursors, mature PACS1(+/R203W) glutamatergic neurons exhibited impaired expression of genes involved in synaptic signaling processes. Subsequent characterization of neural activity using calcium imaging and multielectrode arrays revealed the p.R203W PACS1 variant leads to a prolonged neuronal network burst duration mediated by an increased interspike interval. These findings demonstrate the impact of the PACS1 p.R203W variant on developing human neural tissue and uncover putative electrophysiological underpinnings of disease.


Asunto(s)
Anomalías Craneofaciales , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Neuronas , Discapacidad Intelectual/genética , Anomalías Craneofaciales/genética , Proteínas de Transporte Vesicular/genética
3.
Cell Rep ; 42(8): 112845, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37480564

RESUMEN

Protocadherins (PCDHs) are cell adhesion molecules that regulate many essential neurodevelopmental processes related to neuronal maturation, dendritic arbor formation, axon pathfinding, and synaptic plasticity. Biallelic loss-of-function variants in PCDH12 are associated with several neurodevelopmental disorders (NDDs). Despite the highly deleterious outcome resulting from loss of PCDH12, little is known about its role during brain development and disease. Here, we show that PCDH12 loss severely impairs cerebral organoid development, with reduced proliferative areas and disrupted laminar organization. 2D models further show that neural progenitor cells lacking PCDH12 prematurely exit the cell cycle and differentiate earlier when compared with wild type. Furthermore, we show that PCDH12 regulates neuronal migration and suggest that this could be through a mechanism requiring ADAM10-mediated ectodomain shedding and/or membrane recruitment of cytoskeleton regulators. Our results demonstrate a critical involvement of PCDH12 in cortical organoid development, suggesting a potential cause for the pathogenic mechanisms underlying PCDH12-related NDDs.


Asunto(s)
Orientación del Axón , Organoides , Proteína ADAM10 , Ciclo Celular , División Celular
4.
bioRxiv ; 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36711630

RESUMEN

Protocadherins (PCDHs) are cell adhesion molecules that regulate many essential neurodevelopmental processes related to neuronal maturation, dendritic arbor formation, axon pathfinding, and synaptic plasticity. Bi-allelic loss-of-function variants in PCDH12 are associated with several neurodevelopmental disorders (NDDs) such as diencephalic-mesencephalic dysplasia syndrome, cerebral palsy, cerebellar ataxia, and microcephaly. Despite the highly deleterious outcome resulting from loss of PCDH12, little is known about its role during brain development and disease. Here, we show that PCDH12 loss severely impairs cerebral organoid development with reduced proliferative areas and disrupted laminar organization. 2D models further show that neural progenitor cells lacking PCDH12 prematurely exit cell cycle and differentiate earlier when compared to wildtype. Furthermore, we show that PCDH12 regulates neuronal migration through a mechanism requiring ADAM10-mediated ectodomain shedding and membrane recruitment of cytoskeleton regulators. Our data demonstrate a critical and broad involvement of PCDH12 in cortical development, revealing the pathogenic mechanisms underlying PCDH12-related NDDs.

5.
Trends Genet ; 38(2): 109-112, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34836651

RESUMEN

Rare diseases affect nearly 400 million people worldwide and have a devastating impact on patients and families. Although these diseases are collectively common, they are often overlooked by the research community. We present the ongoing work of the PACS1 Syndrome Research Foundation as a paradigm for approaching rare disease research.


Asunto(s)
Enfermedades Raras , Proteínas de Transporte Vesicular , Humanos , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Enfermedades Raras/genética , Enfermedades Raras/terapia , Síndrome
7.
Ann Neurol ; 90(2): 274-284, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34185323

RESUMEN

OBJECTIVE: The MAST family of microtubule-associated serine-threonine kinases (STKs) have distinct expression patterns in the developing and mature human and mouse brain. To date, only MAST1 has been conclusively associated with neurological disease, with de novo variants in individuals with a neurodevelopmental disorder, including a mega corpus callosum. METHODS: Using exome sequencing, we identify MAST3 missense variants in individuals with epilepsy. We also assess the effect of these variants on the ability of MAST3 to phosphorylate the target gene product ARPP-16 in HEK293T cells. RESULTS: We identify de novo missense variants in the STK domain in 11 individuals, including 2 recurrent variants p.G510S (n = 5) and p.G515S (n = 3). All 11 individuals had developmental and epileptic encephalopathy, with 8 having normal development prior to seizure onset at <2 years of age. All patients developed multiple seizure types, 9 of 11 patients had seizures triggered by fever and 9 of 11 patients had drug-resistant seizures. In vitro analysis of HEK293T cells transfected with MAST3 cDNA carrying a subset of these patient-specific missense variants demonstrated variable but generally lower expression, with concomitant increased phosphorylation of the MAST3 target, ARPP-16, compared to wild-type. These findings suggest the patient-specific variants may confer MAST3 gain-of-function. Moreover, single-nuclei RNA sequencing and immunohistochemistry shows that MAST3 expression is restricted to excitatory neurons in the cortex late in prenatal development and postnatally. INTERPRETATION: In summary, we describe MAST3 as a novel epilepsy-associated gene with a potential gain-of-function pathogenic mechanism that may be primarily restricted to excitatory neurons in the cortex. ANN NEUROL 2021;90:274-284.


Asunto(s)
Epilepsia/diagnóstico por imagen , Epilepsia/genética , Variación Genética/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Niño , Estudios de Cohortes , Epilepsia/metabolismo , Femenino , Estudios de Seguimiento , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Adulto Joven
8.
Epilepsia ; 61(10): 2313-2320, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32944953

RESUMEN

OBJECTIVE: PCDH19-related epilepsy is characterized by a distinctive pattern of X-linked inheritance, where heterozygous females exhibit seizures and hemizygous males are asymptomatic. A cellular interference mechanism resulting from the presence of both wild-type and mutant PCDH19 neurons in heterozygous patients or mosaic carriers of PCDH19 variants has been hypothesized. We aim to investigate seizure susceptibility and progression in the Pchd19 mouse model. METHODS: We assessed seizure susceptibility and progression in the Pcdh19 mouse model using three acute seizure induction paradigms. We first induced focal, clonic seizures using the 6-Hz psychomotor test. Mice were stimulated with increasing current intensities and graded according to a modified Racine scale. We next induced generalized seizures using flurothyl or pentylenetetrazol (PTZ), both γ-aminobutyric acid type A receptor function inhibitors, and recorded latencies to myoclonic and generalized tonic-clonic seizures. RESULTS: Pcdh19 knockout and heterozygous females displayed increased seizure susceptibility across all current intensities in the 6-Hz psychomotor test, and increased severity overall. They also exhibited shorter latencies to generalized seizures following flurothyl, but not PTZ, seizure induction. Hemizygous males showed comparable seizure incidence and severity to their wild-type male littermates across all paradigms tested. SIGNIFICANCE: The heightened susceptibility observed in Pcdh19 knockout females suggests additional mechanisms other than cellular interference are at play in PCDH19-related epilepsy. Further experiments are needed to understand the variability in seizure susceptibility so that this model can be best utilized toward development of future therapeutic strategies for PCDH19-related epilepsy.


Asunto(s)
Cadherinas/deficiencia , Cadherinas/genética , Desempeño Psicomotor/fisiología , Convulsiones/genética , Convulsiones/metabolismo , Animales , Electroencefalografía/métodos , Femenino , Predisposición Genética a la Enfermedad/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Protocadherinas , Convulsiones/fisiopatología
9.
Neuron ; 107(1): 3-5, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32645307

RESUMEN

Loss-of-function variants in the PYRC2 gene cause hypomyelinating leukodystrophy 10 (HLD10), but the associated pathogenic mechanisms are unknown. In this issue of Neuron, Escande-Beillard et al. (2020) reveal that PYRC2 is a key enzyme for proper brain development and a regulator of glycine homeostasis, uncovering hyperglycinemia as a driver of HLD10 pathogenesis.


Asunto(s)
Encefalopatías/metabolismo , Glicina , Ubiquitina-Proteína Ligasas/metabolismo , Humanos , Pirrolina Carboxilato Reductasas
10.
Front Cell Neurosci ; 13: 385, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31481879

RESUMEN

Autism Spectrum Disorder (ASD) is one of the most prevalent neurodevelopmental disorders, affecting an estimated 1 in 59 children. ASD is highly genetically heterogeneous and may be caused by both inheritable and de novo gene variations. In the past decade, hundreds of genes have been identified that contribute to the serious deficits in communication, social cognition, and behavior that patients often experience. However, these only account for 10-20% of ASD cases, and patients with similar pathogenic variants may be diagnosed on very different levels of the spectrum. In this review, we will describe the genetic landscape of ASD and discuss how genetic modifiers such as copy number variation, single nucleotide polymorphisms, and epigenetic alterations likely play a key role in modulating the phenotypic spectrum of ASD patients. We also consider how genetic modifiers can alter convergent signaling pathways and lead to impaired neural circuitry formation. Lastly, we review sex-linked modifiers and clinical implications. Further understanding of these mechanisms is crucial for both comprehending ASD and for developing novel therapies.

11.
Neuron ; 101(6): 1089-1098.e4, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30713029

RESUMEN

Zika virus (ZIKV) targets neural progenitor cells in the brain, attenuates cell proliferation, and leads to cell death. Here, we describe a role for the ZIKV protease NS2B-NS3 heterodimer in mediating neurotoxicity through cleavage of a host protein required for neurogenesis. Similar to ZIKV infection, NS2B-NS3 expression led to cytokinesis defects and cell death in a protease activity-dependent fashion. Among binding partners, NS2B-NS3 cleaved Septin-2, a cytoskeletal factor involved in cytokinesis. Cleavage of Septin-2 occurred at residue 306 and forced expression of a non-cleavable Septin-2 restored cytokinesis, suggesting a direct mechanism of ZIKV-induced neural toxicity. VIDEO ABSTRACT.


Asunto(s)
Apoptosis , Citocinesis , Mitosis , Células-Madre Neurales/metabolismo , Septinas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Virus Zika/metabolismo , Citoesqueleto/metabolismo , Células HEK293 , Células HeLa , Humanos , Neurogénesis , ARN Helicasas/metabolismo , Serina Endopeptidasas/metabolismo
12.
J Vis Exp ; (154)2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31904025

RESUMEN

Organotypic slice culture is a powerful in vitro model that mimicks in vivo conditions more closely than dissociated primary cell cultures. In early postnatal development, cerebellar Purkinje cells are known to go through a vulnerable period, during which they undergo programmed cell death. Here, we provide a detailed protocol to perform mouse organotypic cerebellar slice culture during this critical time. The slices are further labeled to assess Purkinje cell survival and the efficacy of neuroprotective treatments. This method can be extremely valuable to screen for new neuroactive molecules.


Asunto(s)
Células de Purkinje/fisiología , Animales , Supervivencia Celular , Ratones , Técnicas de Cultivo de Órganos
13.
Ann Neurol ; 84(5): 638-647, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30178464

RESUMEN

OBJECTIVE: To identify causes of the autosomal-recessive malformation, diencephalic-mesencephalic junction dysplasia (DMJD) syndrome. METHODS: Eight families with DMJD were studied by whole-exome or targeted sequencing, with detailed clinical and radiological characterization. Patient-derived induced pluripotent stem cells were derived into neural precursor and endothelial cells to study gene expression. RESULTS: All patients showed biallelic mutations in the nonclustered protocadherin-12 (PCDH12) gene. The characteristic clinical presentation included progressive microcephaly, craniofacial dysmorphism, psychomotor disability, epilepsy, and axial hypotonia with variable appendicular spasticity. Brain imaging showed brainstem malformations and with frequent thinned corpus callosum with punctate brain calcifications, reflecting expression of PCDH12 in neural and endothelial cells. These cells showed lack of PCDH12 expression and impaired neurite outgrowth. INTERPRETATION: DMJD patients have biallelic mutations in PCDH12 and lack of protein expression. These patients present with characteristic microcephaly and abnormalities of white matter tracts. Such pathogenic variants predict a poor outcome as a result of brainstem malformation and evidence of white matter tract defects, and should be added to the phenotypic spectrum associated with PCDH12-related conditions. Ann Neurol 2018;84:646-655.


Asunto(s)
Tronco Encefálico/anomalías , Cadherinas/genética , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Protocadherinas
14.
Am J Hum Genet ; 101(3): 441-450, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28823706

RESUMEN

Pontocerebellar hypoplasia (PCH) represents a group of recessive developmental disorders characterized by impaired growth of the pons and cerebellum, which frequently follows a degenerative course. Currently, there are 10 partially overlapping clinical subtypes and 13 genes known mutated in PCH. Here, we report biallelic TBC1D23 mutations in six individuals from four unrelated families manifesting a non-degenerative form of PCH. In addition to reduced volume of pons and cerebellum, affected individuals had microcephaly, psychomotor delay, and ataxia. In zebrafish, tbc1d23 morphants replicated the human phenotype showing hindbrain volume loss. TBC1D23 localized at the trans-Golgi and was regulated by the small GTPases Arl1 and Arl8, suggesting a role in trans-Golgi membrane trafficking. Altogether, this study provides a causative link between TBC1D23 mutations and PCH and suggests a less severe clinical course than other PCH subtypes.


Asunto(s)
Enfermedades Cerebelosas/genética , Proteínas Activadoras de GTPasa/genética , Homocigoto , Microcefalia/genética , Mutación , Adolescente , Animales , Enfermedades Cerebelosas/patología , Niño , Preescolar , Femenino , Células HeLa , Humanos , Masculino , Microcefalia/patología , Linaje , Fenotipo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
15.
Nat Genet ; 49(3): 457-464, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28092684

RESUMEN

Deadenylases are best known for degrading the poly(A) tail during mRNA decay. The deadenylase family has expanded throughout evolution and, in mammals, consists of 12 Mg2+-dependent 3'-end RNases with substrate specificity that is mostly unknown. Pontocerebellar hypoplasia type 7 (PCH7) is a unique recessive syndrome characterized by neurodegeneration and ambiguous genitalia. We studied 12 human families with PCH7, uncovering biallelic, loss-of-function mutations in TOE1, which encodes an unconventional deadenylase. toe1-morphant zebrafish displayed midbrain and hindbrain degeneration, modeling PCH-like structural defects in vivo. Surprisingly, we found that TOE1 associated with small nuclear RNAs (snRNAs) incompletely processed spliceosomal. These pre-snRNAs contained 3' genome-encoded tails often followed by post-transcriptionally added adenosines. Human cells with reduced levels of TOE1 accumulated 3'-end-extended pre-snRNAs, and the immunoisolated TOE1 complex was sufficient for 3'-end maturation of snRNAs. Our findings identify the cause of a neurodegenerative syndrome linked to snRNA maturation and uncover a key factor involved in the processing of snRNA 3' ends.


Asunto(s)
Enfermedades Cerebelosas/genética , Exonucleasas/genética , Mutación/genética , Proteínas Nucleares/genética , ARN Nuclear Pequeño/genética , Alelos , Animales , Femenino , Humanos , Masculino , Ratones , Enfermedades Neurodegenerativas/genética , ARN Mensajero/genética , Empalmosomas/genética , Pez Cebra
16.
Nat Med ; 21(12): 1445-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26523971

RESUMEN

Focal malformations of cortical development (FMCDs) account for the majority of drug-resistant pediatric epilepsy. Postzygotic somatic mutations activating the phosphatidylinositol-4,5-bisphosphate-3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway are found in a wide range of brain diseases, including FMCDs. It remains unclear how a mutation in a small fraction of cells disrupts the architecture of the entire hemisphere. Within human FMCD-affected brain, we found that cells showing activation of the PI3K-AKT-mTOR pathway were enriched for the AKT3(E17K) mutation. Introducing the FMCD-causing mutation into mouse brain resulted in electrographic seizures and impaired hemispheric architecture. Mutation-expressing neural progenitors showed misexpression of reelin, which led to a non-cell autonomous migration defect in neighboring cells, due at least in part to derepression of reelin transcription in a manner dependent on the forkhead box (FOX) transcription factor FOXG1. Treatments aimed at either blocking downstream AKT signaling or inactivating reelin restored migration. These findings suggest a central AKT-FOXG1-reelin signaling pathway in FMCD and support pathway inhibitors as potential treatments or therapies for some forms of focal epilepsy.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Proteínas de la Matriz Extracelular/metabolismo , Factores de Transcripción Forkhead/metabolismo , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , Movimiento Celular/genética , Modelos Animales de Enfermedad , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Imagen por Resonancia Magnética , Malformaciones del Desarrollo Cortical/enzimología , Malformaciones del Desarrollo Cortical/cirugía , Ratones , Datos de Secuencia Molecular , Mosaicismo , Mutación/genética , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fenotipo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Recombinación Genética/genética , Proteína Reelina , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
17.
Nat Genet ; 47(7): 809-13, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26005868

RESUMEN

Docosahexanoic acid (DHA) is the most abundant omega-3 fatty acid in brain, and, although it is considered essential, deficiency has not been linked to disease. Despite the large mass of DHA in phospholipids, the brain does not synthesize it. DHA is imported across the blood-brain barrier (BBB) through the major facilitator superfamily domain-containing 2a (MFSD2A) protein. MFSD2A transports DHA as well as other fatty acids in the form of lysophosphatidylcholine (LPC). We identify two families displaying MFSD2A mutations in conserved residues. Affected individuals exhibited a lethal microcephaly syndrome linked to inadequate uptake of LPC lipids. The MFSD2A mutations impaired transport activity in a cell-based assay. Moreover, when expressed in mfsd2aa-morphant zebrafish, mutants failed to rescue microcephaly, BBB breakdown and lethality. Our results establish a link between transport of DHA and LPCs by MFSD2A and human brain growth and function, presenting the first evidence of monogenic disease related to transport of DHA in humans.


Asunto(s)
Encéfalo/metabolismo , Ácidos Grasos Omega-3/metabolismo , Microcefalia/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Consanguinidad , Femenino , Genes Letales , Estudios de Asociación Genética , Células HEK293 , Humanos , Lactante , Masculino , Ratones Noqueados , Mutación Missense , Simportadores , Síndrome , Pez Cebra
18.
Neuron ; 82(5): 1004-16, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24908484

RESUMEN

Activity-dependent neurotransmitter switching engages genetic programs regulating transmitter synthesis, but the mechanism by which activity is transduced is unknown. We suppressed activity in single neurons in the embryonic spinal cord to determine whether glutamate-gamma-aminobutyric acid (GABA) switching is cell autonomous. Transmitter respecification did not occur, suggesting that it is homeostatically regulated by the level of activity in surrounding neurons. Graded increase in the number of silenced neurons in cultures led to graded decrease in the number of neurons expressing GABA, supporting non-cell-autonomous transmitter switching. We found that brain-derived neurotrophic factor (BDNF) is expressed in the spinal cord during the period of transmitter respecification and that spike activity causes release of BDNF. Activation of TrkB receptors triggers a signaling cascade involving JNK-mediated activation of cJun that regulates tlx3, a glutamate/GABA selector gene, accounting for calcium-spike BDNF-dependent transmitter switching. Our findings identify a molecular mechanism for activity-dependent respecification of neurotransmitter phenotype in developing spinal neurons.


Asunto(s)
Ácido Glutámico/metabolismo , Neuronas/metabolismo , Transducción de Señal , Médula Espinal/embriología , Ácido gamma-Aminobutírico/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Calcio/metabolismo , Células Cultivadas , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-jun/metabolismo , Médula Espinal/metabolismo , Xenopus laevis
19.
Neuron ; 82(3): 511-21, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24811376

RESUMEN

Primary cilia were the largely neglected nonmotile counterparts of their better-known cousin, the motile cilia. For years these nonmotile cilia were considered evolutionary remnants of little consequence to cellular function. Fast forward 10 years and we now recognize primary cilia as key integrators of extracellular ligand-based signaling and cellular polarity, which regulate neuronal cell fate, migration, differentiation, as well as a host of adult behaviors. Important future questions will focus on structure-function relationships, their roles in signaling and disease and as areas of target for treatments.


Asunto(s)
Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Cilios/fisiología , Animales , Encefalopatías/metabolismo , Encefalopatías/patología , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Cilios/patología , Humanos , Transducción de Señal/fisiología
20.
Am J Hum Genet ; 94(1): 80-6, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24360807

RESUMEN

Joubert syndrome and related disorders (JSRDs) are genetically heterogeneous and characterized by a distinctive mid-hindbrain malformation. Causative mutations lead to primary cilia dysfunction, which often results in variable involvement of other organs such as the liver, retina, and kidney. We identified predicted null mutations in CSPP1 in six individuals affected by classical JSRDs. CSPP1 encodes a protein localized to centrosomes and spindle poles, as well as to the primary cilium. Despite the known interaction between CSPP1 and nephronophthisis-associated proteins, none of the affected individuals in our cohort presented with kidney disease, and further, screening of a large cohort of individuals with nephronophthisis demonstrated no mutations. CSPP1 is broadly expressed in neural tissue, and its encoded protein localizes to the primary cilium in an in vitro model of human neurogenesis. Here, we show abrogated protein levels and ciliogenesis in affected fibroblasts. Our data thus suggest that CSPP1 is involved in neural-specific functions of primary cilia.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/genética , Anomalías del Ojo/genética , Eliminación de Gen , Enfermedades Renales Quísticas/genética , Proteínas Asociadas a Microtúbulos/genética , Retina/anomalías , Anomalías Múltiples , Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Cerebelo/anomalías , Cilios/genética , Cilios/patología , Estudios de Cohortes , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Proteínas Asociadas a Microtúbulos/metabolismo , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA