Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38559165

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that primarily affects motor neurons, leading to progressive muscle weakness and loss of voluntary muscle control. While the exact cause of ALS is not fully understood, emerging research suggests that dysfunction of the nuclear envelope (NE) may contribute to disease pathogenesis and progression. The NE plays a role in ALS through several mechanisms, including nuclear pore defects, nucleocytoplasmic transport impairment, accumulation of mislocalized proteins, and nuclear morphology abnormalities. The LINC complex is the second biggest multi-protein complex in the NE and consists of the SUN1/2 proteins spanning the inner nuclear membrane and Nesprin proteins embedded in the outer membrane. The LINC complex, by interacting with both the nuclear lamina and the cytoskeleton, transmits mechanical forces to the nucleus regulating its morphology and functional homeostasis. In this study we show extensive alterations to the LINC complex in motor and cortical iPSC-derived neurons and spinal cord organoids carrying the ALS causative mutation in the C9ORF72 gene (C9). Importantly, we show that such alterations are present in vivo in a cohort of sporadic ALS and C9-ALS postmortem spinal cord and motor cortex biopsies. We also found that LINC complex disruption strongly correlated with nuclear morphological alterations occurring in ALS neurons, independently of TDP43 mislocalization. Altogether, our data establish morphological and functional alterations to the LINC complex as important events in ALS pathogenic cascade, making this pathway a possible target for both biomarker and therapy development.

2.
Acta Neuropathol Commun ; 12(1): 69, 2024 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664831

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder that primarily affects motor neurons, leading to progressive muscle weakness and loss of voluntary muscle control. While the exact cause of ALS is not fully understood, emerging research suggests that dysfunction of the nuclear envelope (NE) may contribute to disease pathogenesis and progression. The NE plays a role in ALS through several mechanisms, including nuclear pore defects, nucleocytoplasmic transport impairment, accumulation of mislocalized proteins, and nuclear morphology abnormalities. The LINC complex is the second biggest multi-protein complex in the NE and consists of the SUN1/2 proteins spanning the inner nuclear membrane and Nesprin proteins embedded in the outer membrane. The LINC complex, by interacting with both the nuclear lamina and the cytoskeleton, transmits mechanical forces to the nucleus regulating its morphology and functional homeostasis. In this study we show extensive alterations to the LINC complex in motor and cortical iPSC-derived neurons and spinal cord organoids carrying the ALS causative mutation in the C9ORF72 gene (C9). Importantly, we show that such alterations are present in vivo in a cohort of sporadic ALS and C9-ALS postmortem spinal cord and motor cortex specimens. We also found that LINC complex disruption strongly correlated with nuclear morphological alterations occurring in ALS neurons, independently of TDP43 mislocalization. Altogether, our data establish morphological and functional alterations to the LINC complex as important events in ALS pathogenic cascade, making this pathway a possible target for both biomarker and therapy development.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteína C9orf72 , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/metabolismo , Humanos , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Demencia Frontotemporal/metabolismo , Masculino , Neuronas Motoras/patología , Neuronas Motoras/metabolismo , Médula Espinal/patología , Médula Espinal/metabolismo , Membrana Nuclear/metabolismo , Membrana Nuclear/patología , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Persona de Mediana Edad , Anciano , Corteza Motora/patología , Corteza Motora/metabolismo
3.
bioRxiv ; 2024 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-38352403

RESUMEN

ALS and FTD are complex neurodegenerative disorders that primarily affects motor neurons in the brain and spinal cord, and cortical neurons in the frontal lobe. Although the pathogenesis of ALS/FTD is unclear, recent research spotlights nucleocytoplasmic transport impairment, DNA damage, and nuclear abnormalities as drivers of neuronal death. In this study, we show that loss of nuclear envelope (NE) integrity is a key pathology associated with nuclear pore complex (NPC) injury in C9ORF72 mutant neurons. Importantly, we show that mechanical stresses generated by cytoskeletal forces on the NE can lead to NPC injury, loss of nuclear integrity, and accumulation of DNA damage. Importantly, we demonstrate that restoring NE tensional homeostasis, by disconnecting the nucleus from the cytoskeleton, can rescue NPC injury and reduce DNA damage in C9ORF72 mutant cells. Together, our data suggest that modulation of NE homeostasis and repair may represent a novel and promising therapeutic target for ALS/FTD.

4.
Cell Rep ; 39(1): 110598, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385738

RESUMEN

Understanding the pathogenic mechanisms of disease mutations is critical to advancing treatments. ALS-associated mutations in the gene encoding the microtubule motor KIF5A result in skipping of exon 27 (KIF5AΔExon27) and the encoding of a protein with a novel 39 amino acid residue C-terminal sequence. Here, we report that expression of ALS-linked mutant KIF5A results in dysregulated motor activity, cellular mislocalization, altered axonal transport, and decreased neuronal survival. Single-molecule analysis revealed that the altered C terminus of mutant KIF5A results in a constitutively active state. Furthermore, mutant KIF5A possesses altered protein and RNA interactions and its expression results in altered gene expression/splicing. Taken together, our data support the hypothesis that causative ALS mutations result in a toxic gain of function in the intracellular motor KIF5A that disrupts intracellular trafficking and neuronal homeostasis.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/genética , Transporte Axonal/genética , Mutación con Ganancia de Función , Humanos , Cinesinas/genética , Mutación/genética
5.
JAMA Neurol ; 78(10): 1236-1248, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34459874

RESUMEN

Importance: Juvenile amyotrophic lateral sclerosis (ALS) is a rare form of ALS characterized by age of symptom onset less than 25 years and a variable presentation. Objective: To identify the genetic variants associated with juvenile ALS. Design, Setting, and Participants: In this multicenter family-based genetic study, trio whole-exome sequencing was performed to identify the disease-associated gene in a case series of unrelated patients diagnosed with juvenile ALS and severe growth retardation. The patients and their family members were enrolled at academic hospitals and a government research facility between March 1, 2016, and March 13, 2020, and were observed until October 1, 2020. Whole-exome sequencing was also performed in a series of patients with juvenile ALS. A total of 66 patients with juvenile ALS and 6258 adult patients with ALS participated in the study. Patients were selected for the study based on their diagnosis, and all eligible participants were enrolled in the study. None of the participants had a family history of neurological disorders, suggesting de novo variants as the underlying genetic mechanism. Main Outcomes and Measures: De novo variants present only in the index case and not in unaffected family members. Results: Trio whole-exome sequencing was performed in 3 patients diagnosed with juvenile ALS and their parents. An additional 63 patients with juvenile ALS and 6258 adult patients with ALS were subsequently screened for variants in the SPTLC1 gene. De novo variants in SPTLC1 (p.Ala20Ser in 2 patients and p.Ser331Tyr in 1 patient) were identified in 3 unrelated patients diagnosed with juvenile ALS and failure to thrive. A fourth variant (p.Leu39del) was identified in a patient with juvenile ALS where parental DNA was unavailable. Variants in this gene have been previously shown to be associated with autosomal-dominant hereditary sensory autonomic neuropathy, type 1A, by disrupting an essential enzyme complex in the sphingolipid synthesis pathway. Conclusions and Relevance: These data broaden the phenotype associated with SPTLC1 and suggest that patients presenting with juvenile ALS should be screened for variants in this gene.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Predisposición Genética a la Enfermedad/genética , Serina C-Palmitoiltransferasa/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Mutación , Secuenciación del Exoma , Adulto Joven
6.
Neurol Genet ; 7(3): e596, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34169147

RESUMEN

OBJECTIVE: Despite the strong genetic component of frontotemporal dementia (FTD), a substantial proportion of patients remain genetically unresolved. We performed an in-depth study of a family with an autosomal dominant form of FTD to investigate the underlying genetic cause. METHODS: Following clinical and pathologic characterization of the family, genetic studies included haplotype sharing analysis and exome sequencing. Subsequently, we performed immunohistochemistry, immunoblotting, and a microtubule repolymerization assay to investigate the potential impact of the candidate variant in tubulin alpha 4a (TUBA4A). RESULTS: The clinical presentation in this family is heterogeneous, including behavioral changes, parkinsonian features, and uncharacterized dementia. Neuropathologic examination of 2 patients revealed TAR DNA binding protein 43 (TDP-43) pathology with abundant dystrophic neurites and neuronal intranuclear inclusions, consistent with frontotemporal lobar degeneration-TDP type A. We identified a likely pathogenic variant in TUBA4A segregating with disease. TUBA4A encodes for α-tubulin, which is a major component of the microtubule network. Variants in TUBA4A have been suggested as a rare genetic cause of amyotrophic lateral sclerosis (ALS) and have sporadically been reported in patients with FTD without supporting genetic segregation. A decreased trend of TUBA4A protein abundance was observed in patients compared with controls, and a microtubule repolymerization assay demonstrated disrupted α-tubulin function. As opposed to variants found in ALS, TUBA4A variants associated with FTD appear more localized to the N-terminus, indicating different pathogenic mechanisms. CONCLUSIONS: Our findings support the role of TUBA4A variants as rare genetic cause of familial FTD.

7.
Neurobiol Dis ; 140: 104835, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32179176

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal late-onset neurodegenerative disease that specifically affects the function and survival of spinal and cortical motor neurons. ALS shares many genetic, clinical, and pathological characteristics with frontotemporal dementia (FTD), and these diseases are now recognized as presentations of a disease spectrum known as ALS/FTD. The molecular determinants of neuronal loss in ALS/FTD are still debated, but the recent discovery of nucleocytoplasmic transport defects as a common denominator of most if not all forms of ALS/FTD has dramatically changed our understanding of the pathogenic mechanisms of this disease. Loss of nuclear pores and nucleoporin aggregation, altered nuclear morphology, and impaired nuclear transport are some of the most prominent features that have been identified using a variety of animal, cellular, and human models of disease. Here, we review the experimental evidence linking nucleocytoplasmic transport defects to the pathogenesis of ALS/FTD and propose a unifying view on how these defects may lead to a vicious cycle that eventually causes neuronal death.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Demencia Frontotemporal/patología , Poro Nuclear/metabolismo , Transporte Activo de Núcleo Celular , Animales , Proteína C9orf72/metabolismo , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos
8.
Biochim Biophys Acta Gene Regul Mech ; 1862(9): 194413, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31382054

RESUMEN

The RNA-binding protein TDP-43, associated to amyotrophic lateral sclerosis and frontotemporal dementia, regulates the alternative splicing of several genes, including the skipping of TNIK exon 15. TNIK, a genetic risk factor for schizophrenia and causative for intellectual disability, encodes for a Ser/Thr kinase regulating negatively F-actin dynamics. Here we show that in the human adult nervous system TNIK exon 15 is mostly included compared to the other tissues and that, during neuronal differentiation of human induced pluripotent stem cells and of human neuroblastoma cells, TNIK exon 15 inclusion increases independently of TDP-43 protein content. By studying the possible molecular interplay of TDP-43 with brain-specific splicing factors, we found that the neuronal NOVA-1 protein competitively inhibits both TDP-43 and hnRNPA2/B1 skipping activity on TNIK by means of a RNA-dependent interaction and that this competitive mechanism is common to other TDP-43 RNA targets. We also show that the TNIK protein isoforms including/excluding exon 15 differently regulate cell spreading in non-neuronal cells and neuritogenesis in primary cortical neurons. Our data suggest a complex regulation between the ubiquitous TDP-43 and the neuron-specific NOVA-1 splicing factors in the brain that may help better understand the pathobiology of both neurodegenerative diseases and schizophrenia.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/genética , Esquizofrenia/genética , Empalme Alternativo/genética , Línea Celular , Proteínas de Unión al ADN/química , Exones/genética , Humanos , Antígeno Ventral Neuro-Oncológico , Neuronas/metabolismo , Neuronas/patología , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Proteínas Serina-Treonina Quinasas/química , ARN Mensajero/genética , Proteínas de Unión al ARN/química , Esquizofrenia/patología
9.
Nat Commun ; 10(1): 3827, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444357

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of unknown etiology. Although defects in nucleocytoplasmic transport (NCT) may be central to the pathogenesis of ALS and other neurodegenerative diseases, the molecular mechanisms modulating the nuclear pore function are still largely unknown. Here we show that genetic and pharmacological modulation of actin polymerization disrupts nuclear pore integrity, nuclear import, and downstream pathways such as mRNA post-transcriptional regulation. Importantly, we demonstrate that modulation of actin homeostasis can rescue nuclear pore instability and dysfunction caused by mutant PFN1 as well as by C9ORF72 repeat expansion, the most common mutation in ALS patients. Collectively, our data link NCT defects to ALS-associated cellular pathology and propose the regulation of actin homeostasis as a novel therapeutic strategy for ALS and other neurodegenerative diseases.


Asunto(s)
Actinas/metabolismo , Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/patología , Poro Nuclear/patología , Profilinas/metabolismo , Acrilamidas/farmacología , Actinas/ultraestructura , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Esclerosis Amiotrófica Lateral/genética , Biopsia , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Línea Celular , Corteza Cerebral/citología , Corteza Cerebral/patología , Embrión de Mamíferos , Fibroblastos , Humanos , Microscopía Electrónica de Transmisión , Neuronas Motoras/citología , Mutación , Poro Nuclear/efectos de los fármacos , Poro Nuclear/ultraestructura , Cultivo Primario de Células , Profilinas/genética , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética , Piel/citología , Piel/patología , Tiazoles/farmacología , Tiazolidinas/farmacología
10.
J Biol Chem ; 294(26): 10194-10210, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31092554

RESUMEN

Excitotoxic levels of glutamate represent a physiological stress that is strongly linked to amyotrophic lateral sclerosis (ALS) and other neurological disorders. Emerging evidence indicates a role for neurodegenerative disease-linked RNA-binding proteins (RBPs) in the cellular stress response. However, the relationships between excitotoxicity, RBP function, and disease have not been explored. Here, using primary cortical and motor neurons, we found that excitotoxicity induced the translocation of select ALS-linked RBPs from the nucleus to the cytoplasm within neurons. RBPs affected by excitotoxicity included TAR DNA-binding protein 43 (TDP-43) and, most robustly, fused in sarcoma/translocated in liposarcoma (FUS/TLS or FUS). We noted that FUS is translocated through a calcium-dependent mechanism and that its translocation coincides with striking alterations in nucleocytoplasmic transport. Furthermore, glutamate-induced up-regulation of glutamate ionotropic receptor α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type subunit 2 (GRIA2) in neurons depended on FUS expression, consistent with a functional role for FUS in excitotoxic stress. These findings reveal molecular links among prominent factors in neurodegenerative diseases, namely excitotoxicity, disease-associated RBPs, and nucleocytoplasmic transport.


Asunto(s)
Calcio/metabolismo , Núcleo Celular/metabolismo , Ácido Glutámico/efectos adversos , ARN Mensajero/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Receptores AMPA/metabolismo , Estrés Fisiológico , Transporte Activo de Núcleo Celular , Esclerosis Amiotrófica Lateral , Citoplasma , Demencia Frontotemporal , Humanos , Mutación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Proteína FUS de Unión a ARN/genética , Receptores AMPA/genética
11.
Sci Transl Med ; 9(388)2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28469040

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder. We screened 751 familial ALS patient whole-exome sequences and identified six mutations including p.D40G in the ANXA11 gene in 13 individuals. The p.D40G mutation was absent from 70,000 control whole-exome sequences. This mutation segregated with disease in two kindreds and was present in another two unrelated cases (P = 0.0102), and all mutation carriers shared a common founder haplotype. Annexin A11-positive protein aggregates were abundant in spinal cord motor neurons and hippocampal neuronal axons in an ALS patient carrying the p.D40G mutation. Transfected human embryonic kidney cells expressing ANXA11 with the p.D40G mutation and other N-terminal mutations showed altered binding to calcyclin, and the p.R235Q mutant protein formed insoluble aggregates. We conclude that mutations in ANXA11 are associated with ALS and implicate defective intracellular protein trafficking in disease pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Anexinas/genética , Anexinas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Mutación/genética , Unión Proteica , Transporte de Proteínas , Proteína A6 de Unión a Calcio de la Familia S100/metabolismo
12.
Hum Mol Genet ; 26(11): 2146-2155, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28379367

RESUMEN

Mutations in the profilin 1 (PFN1) gene are causative for familial amyotrophic lateral sclerosis (fALS). However, it is still not fully understood how these mutations lead to neurodegeneration. To address this question, we generated a novel Drosophila model expressing human wild-type and ALS-causative PFN1 mutants. We show that at larval neuromuscular junctions (NMJ), motor neuron expression of wild-type human PFN1 increases the number of ghost boutons, active zone density, F-actin content, and the formation of filopodia. In contrast, the expression of ALS-causative human PFN1 mutants causes a less pronounced phenotype, suggesting a loss of function of these mutants in promoting NMJ remodeling. Importantly, expression of human PFN1 in motor neurons results in progressive locomotion defects and shorter lifespan in adult flies, while ALS-causative PFN1 mutants display a less toxic effect. In summary, our study provides evidence that PFN1 is important in regulating NMJ morphology and influences survival and locomotion in Drosophila. Furthermore, our results suggest ALS-causative human PFN1 mutants display a partial loss of function relative to wild-type hPFN1 that may contribute to human disease pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Profilinas/genética , Profilinas/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Modelos Animales de Enfermedad , Drosophila/metabolismo , Regulación de la Expresión Génica , Humanos , Neuronas Motoras/metabolismo , Mutación , Unión Neuromuscular/metabolismo
13.
Sci Rep ; 7(1): 115, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28273913

RESUMEN

Mutations in Fused in Sarcoma/Translocated in Liposarcoma (FUS) cause familial forms of amyotrophic lateral sclerosis (ALS), a neurodegenerative disease characterized by progressive axonal degeneration mainly affecting motor neurons. Evidence from transgenic mouse models suggests mutant forms of FUS exert an unknown gain-of-toxic function in motor neurons, but mechanisms underlying this effect remain unknown. Towards this end, we studied the effect of wild type FUS (FUS WT) and three ALS-linked variants (G230C, R521G and R495X) on fast axonal transport (FAT), a cellular process critical for appropriate maintenance of axonal connectivity. All ALS-FUS variants impaired anterograde and retrograde FAT in squid axoplasm, whereas FUS WT had no effect. Misfolding of mutant FUS is implicated in this process, as the molecular chaperone Hsp110 mitigated these toxic effects. Interestingly, mutant FUS-induced impairment of FAT in squid axoplasm and of axonal outgrowth in mammalian primary motor neurons involved aberrant activation of the p38 MAPK pathway, as also reported for ALS-linked forms of Cu, Zn superoxide dismutase (SOD1). Accordingly, increased levels of active p38 MAPK were detected in post-mortem human ALS-FUS brain tissues. These data provide evidence for a novel gain-of-toxic function for ALS-linked FUS involving p38 MAPK activation.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Transporte Axonal , Neuronas Motoras/metabolismo , Proteína FUS de Unión a ARN/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Decapodiformes/crecimiento & desarrollo , Decapodiformes/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mutación , Pliegue de Proteína , Proteína FUS de Unión a ARN/química , Superóxido Dismutasa-1/metabolismo
14.
Cell Rep ; 18(7): 1660-1673, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199839

RESUMEN

Spinal muscular atrophy (SMA) is a motor neuron disease caused by reduced levels of the survival of motor neuron (SMN) protein. SMN is part of a multiprotein complex that facilitates the assembly of spliceosomal small nuclear ribonucleoproteins (snRNPs). SMN has also been found to associate with mRNA-binding proteins, but the nature of this association was unknown. Here, we have employed a combination of biochemical and advanced imaging methods to demonstrate that SMN promotes the molecular interaction between IMP1 protein and the 3' UTR zipcode region of ß-actin mRNA, leading to assembly of messenger ribonucleoprotein (mRNP) complexes that associate with the cytoskeleton to facilitate trafficking. We have identified defects in mRNP assembly in cells and tissues from SMA disease models and patients that depend on the SMN Tudor domain and explain the observed deficiency in mRNA localization and local translation, providing insight into SMA pathogenesis as a ribonucleoprotein (RNP)-assembly disorder.


Asunto(s)
Chaperonas Moleculares/metabolismo , Neuronas Motoras/metabolismo , Ribonucleoproteínas/metabolismo , Regiones no Traducidas 3'/fisiología , Actinas/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Humanos , Atrofia Muscular Espinal/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas del Complejo SMN/metabolismo
15.
Nat Genet ; 48(9): 1037-42, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27455347

RESUMEN

To identify genetic factors contributing to amyotrophic lateral sclerosis (ALS), we conducted whole-exome analyses of 1,022 index familial ALS (FALS) cases and 7,315 controls. In a new screening strategy, we performed gene-burden analyses trained with established ALS genes and identified a significant association between loss-of-function (LOF) NEK1 variants and FALS risk. Independently, autozygosity mapping for an isolated community in the Netherlands identified a NEK1 p.Arg261His variant as a candidate risk factor. Replication analyses of sporadic ALS (SALS) cases and independent control cohorts confirmed significant disease association for both p.Arg261His (10,589 samples analyzed) and NEK1 LOF variants (3,362 samples analyzed). In total, we observed NEK1 risk variants in nearly 3% of ALS cases. NEK1 has been linked to several cellular functions, including cilia formation, DNA-damage response, microtubule stability, neuronal morphology and axonal polarity. Our results provide new and important insights into ALS etiopathogenesis and genetic etiology.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Quinasa 1 Relacionada con NIMA/genética , Esclerosis Amiotrófica Lateral/epidemiología , Estudios de Casos y Controles , Estudios de Cohortes , Exoma/genética , Estudios de Asociación Genética , Humanos , Países Bajos/epidemiología
16.
J Neurosci ; 36(13): 3811-20, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27030765

RESUMEN

Spinal muscular atrophy (SMA) is a neurodegenerative disease primarily affecting spinal motor neurons. It is caused by reduced levels of the survival of motor neuron (SMN) protein, which plays an essential role in the biogenesis of spliceosomal small nuclear ribonucleoproteins in all tissues. The etiology of the specific defects in the motor circuitry in SMA is still unclear, but SMN has also been implicated in mediating the axonal localization of mRNA-protein complexes, which may contribute to the axonal degeneration observed in SMA. Here, we report that SMN deficiency severely disrupts local protein synthesis within neuronal growth cones. We also identify the cytoskeleton-associated growth-associated protein 43 (GAP43) mRNA as a new target of SMN and show that motor neurons from SMA mouse models have reduced levels ofGAP43mRNA and protein in axons and growth cones. Importantly, overexpression of two mRNA-binding proteins, HuD and IMP1, restoresGAP43mRNA and protein levels in growth cones and rescues axon outgrowth defects in SMA neurons. These findings demonstrate that SMN plays an important role in the localization and local translation of mRNAs with important axonal functions and suggest that disruption of this function may contribute to the axonal defects observed in SMA. SIGNIFICANCE STATEMENT: The motor neuron disease spinal muscular atrophy (SMA) is caused by reduced levels of the survival of motor neuron (SMN) protein, which plays a key role in assembling RNA/protein complexes that are essential for mRNA splicing. It remains unclear whether defects in this well characterized housekeeping function cause the specific degeneration of spinal motor neurons observed in SMA. Here, we describe an additional role of SMN in regulating the axonal localization and local translation of the mRNA encoding growth-associated protein 43 (GAP43). This study supports a model whereby SMN deficiency impedes transport and local translation of mRNAs important for neurite outgrowth and stabilization, thus contributing to axon degeneration, muscle denervation, and motor neuron cell death in SMA.


Asunto(s)
Conos de Crecimiento/fisiología , Neuronas Motoras/fisiología , ARN Mensajero/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Proteína 4 Similar a ELAV/metabolismo , Embrión de Mamíferos , Femenino , Proteína GAP-43/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Médula Espinal/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/deficiencia , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Transfección
17.
Neuron ; 84(2): 324-31, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25374358

RESUMEN

Exome sequencing is an effective strategy for identifying human disease genes. However, this methodology is difficult in late-onset diseases where limited availability of DNA from informative family members prohibits comprehensive segregation analysis. To overcome this limitation, we performed an exome-wide rare variant burden analysis of 363 index cases with familial ALS (FALS). The results revealed an excess of patient variants within TUBA4A, the gene encoding the Tubulin, Alpha 4A protein. Analysis of a further 272 FALS cases and 5,510 internal controls confirmed the overrepresentation as statistically significant and replicable. Functional analyses revealed that TUBA4A mutants destabilize the microtubule network, diminishing its repolymerization capability. These results further emphasize the role of cytoskeletal defects in ALS and demonstrate the power of gene-based rare variant analyses in situations where causal genes cannot be identified through traditional segregation analysis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Exoma/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Tubulina (Proteína)/genética , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Neuronas/metabolismo , Análisis de Secuencia de ADN , Tubulina (Proteína)/metabolismo
18.
Dev Neurobiol ; 74(3): 319-332, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23897586

RESUMEN

Spinal muscular atrophy (SMA) is a lethal neurodegenerative disease specifically affecting spinal motor neurons. SMA is caused by the homozygous deletion or mutation of the survival of motor neuron 1 (SMN1) gene. The SMN protein plays an essential role in the assembly of spliceosomal ribonucleoproteins. However, it is still unclear how low levels of the ubiquitously expressed SMN protein lead to the selective degeneration of motor neurons. An additional role for SMN in the regulation of the axonal transport of mRNA-binding proteins (mRBPs) and their target mRNAs has been proposed. Indeed, several mRBPs have been shown to interact with SMN, and the axonal levels of few mRNAs, such as the ß-actin mRNA, are reduced in SMA motor neurons. In this study we have identified the ß-actin mRNA-binding protein IMP1/ZBP1 as a novel SMN-interacting protein. Using a combination of biochemical assays and quantitative imaging techniques in primary motor neurons, we show that IMP1 associates with SMN in individual granules that are actively transported in motor neuron axons. Furthermore, we demonstrate that IMP1 axonal localization depends on SMN levels, and that SMN deficiency in SMA motor neurons leads to a dramatic reduction of IMP1 protein levels. In contrast, no difference in IMP1 protein levels was detected in whole brain lysates from SMA mice, further suggesting neuron specific roles of SMN in IMP1 expression and localization. Taken together, our data support a role for SMN in the regulation of mRNA localization and axonal transport through its interaction with mRBPs such as IMP1.


Asunto(s)
Axones/metabolismo , Neuronas Motoras/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Transporte Axonal , Transporte Biológico Activo , Encéfalo/metabolismo , Células Cultivadas , Gránulos Cromafines/metabolismo , Humanos , Ratones , Ratones Transgénicos , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/genética , Ratas , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética
19.
Nature ; 488(7412): 499-503, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22801503

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a late-onset neurodegenerative disorder resulting from motor neuron death. Approximately 10% of cases are familial (FALS), typically with a dominant inheritance mode. Despite numerous advances in recent years, nearly 50% of FALS cases have unknown genetic aetiology. Here we show that mutations within the profilin 1 (PFN1) gene can cause FALS. PFN1 is crucial for the conversion of monomeric (G)-actin to filamentous (F)-actin. Exome sequencing of two large ALS families showed different mutations within the PFN1 gene. Further sequence analysis identified 4 mutations in 7 out of 274 FALS cases. Cells expressing PFN1 mutants contain ubiquitinated, insoluble aggregates that in many cases contain the ALS-associated protein TDP-43. PFN1 mutants also display decreased bound actin levels and can inhibit axon outgrowth. Furthermore, primary motor neurons expressing mutant PFN1 display smaller growth cones with a reduced F/G-actin ratio. These observations further document that cytoskeletal pathway alterations contribute to ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Predisposición Genética a la Enfermedad/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Profilinas/genética , Profilinas/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Axones/metabolismo , Axones/patología , Células Cultivadas , Exoma/genética , Femenino , Conos de Crecimiento/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Judíos/genética , Masculino , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Proteínas Mutantes/genética , Linaje , Conformación Proteica , Ubiquitinación , Población Blanca/genética
20.
PLoS One ; 7(6): e38658, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761693

RESUMEN

TAR DNA-binding protein 43 (TDP-43) is a major component within ubiquitin-positive inclusions of a number of neurodegenerative diseases that increasingly are considered as TDP-43 proteinopathies. Identities of other inclusion proteins associated with TDP-43 aggregation remain poorly defined. In this study, we identify and quantitate 35 co-aggregating proteins in the detergent-resistant fraction of HEK-293 cells in which TDP-43 or a particularly aggregate prone variant, TDP-S6, were enriched following overexpression, using stable isotope-labeled (SILAC) internal standards and liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). We also searched for differential post-translational modification (PTM) sites of ubiquitination. Four sites of ubiquitin conjugation to TDP-43 or TDP-S6 were confirmed by dialkylated GST-TDP-43 external reference peptides, occurring on or near RNA binding motif (RRM) 1. RRM-containing proteins co-enriched in cytoplasmic granular structures in HEK-293 cells and primary motor neurons with insoluble TDP-S6, including cytoplasmic stress granule associated proteins G3BP, PABPC1, and eIF4A1. Proteomic evidence for TDP-43 co-aggregation with paraspeckle markers RBM14, PSF and NonO was also validated by western blot and by immunocytochemistry in HEK-293 cells. An increase in peptides from methylated arginine-glycine-glycine (RGG) RNA-binding motifs of FUS/TLS and hnRNPs was found in the detergent-insoluble fraction of TDP-overexpressing cells. Finally, TDP-43 and TDP-S6 detergent-insoluble species were reduced by mutagenesis of the identified ubiquitination sites, even following oxidative or proteolytic stress. Together, these findings define some of the aggregation partners of TDP-43, and suggest that TDP-43 ubiquitination influences TDP-43 oligomerization.


Asunto(s)
Secuencias de Aminoácidos/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteinopatías TDP-43/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Animales , Arginina/química , Western Blotting , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/genética , Glicina/química , Células HEK293 , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Fragmentos de Péptidos/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Proteómica , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleósido Difosfato Reductasa , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/patología , Espectrometría de Masas en Tándem , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...