Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Med ; 30(5): 1406-1415, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38745011

RESUMO

GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .


Assuntos
Dependovirus , Demência Frontotemporal , Terapia Genética , Progranulinas , Humanos , Demência Frontotemporal/genética , Demência Frontotemporal/terapia , Demência Frontotemporal/líquido cefalorraquidiano , Progranulinas/genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Dependovirus/genética , Pessoa de Meia-Idade , Feminino , Masculino , Idoso , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/líquido cefalorraquidiano , Vetores Genéticos , Animais , Resultado do Tratamento , Pesquisa Translacional Biomédica , Camundongos , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Proteínas de Neurofilamentos/sangue
2.
Sci Adv ; 7(51): eabg6363, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34910524

RESUMO

The pathogenesis of inherited genome instability neurodegenerative syndromes remains largely unknown. Here, we report new disease-relevant murine models of genome instability­driven neurodegeneration involving disabled ATM and APTX that develop debilitating ataxia. We show that neurodegeneration and ataxia result from transcriptional interference in the cerebellum via aberrant messenger RNA splicing. Unexpectedly, these splicing defects were restricted to only Purkinje cells, disrupting the expression of critical homeostatic regulators including ITPR1, GRID2, and CA8. Abundant genotoxic R loops were also found at these Purkinje cell gene loci, further exacerbating DNA damage and transcriptional disruption. Using ATAC-seq to profile global chromatin accessibility in the cerebellum, we found a notably unique chromatin conformation specifically in Purkinje chromatin at the affected gene loci, thereby promoting susceptibility to DNA damage. These data reveal the pathogenic basis of DNA damage in the nervous system and suggest chromatin conformation as a feature in directing genome instability­associated neuropathology.

3.
EMBO J ; 40(7): e106106, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33709453

RESUMO

A critical question in neurodegeneration is why the accumulation of disease-driving proteins causes selective neuronal loss despite their brain-wide expression. In Spinocerebellar ataxia type 1 (SCA1), accumulation of polyglutamine-expanded Ataxin-1 (ATXN1) causes selective degeneration of cerebellar and brainstem neurons. Previous studies revealed that inhibiting Msk1 reduces phosphorylation of ATXN1 at S776 as well as its levels leading to improved cerebellar function. However, there are no regulators that modulate ATXN1 in the brainstem-the brain region whose pathology is most closely linked to premature death. To identify new regulators of ATXN1, we performed genetic screens and identified a transcription factor-kinase axis (ZBTB7B-RSK3) that regulates ATXN1 levels. Unlike MSK1, RSK3 is highly expressed in the human and mouse brainstems where it regulates Atxn1 by phosphorylating S776. Reducing Rsk3 rescues brainstem-associated pathologies and deficits, and lowering Rsk3 and Msk1 together improves cerebellar and brainstem function in an SCA1 mouse model. Our results demonstrate that selective vulnerability of brain regions in SCA1 is governed by region-specific regulators of ATXN1, and targeting multiple regulators could rescue multiple degenerating brain areas.


Assuntos
Tronco Encefálico/metabolismo , Cerebelo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Ataxias Espinocerebelares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Ataxina-1/genética , Ataxina-1/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Proteínas de Ligação a DNA/genética , Drosophila melanogaster , Células HEK293 , Humanos , Camundongos , Fosforilação , Estabilidade Proteica , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Ataxias Espinocerebelares/genética , Fatores de Transcrição/genética
4.
Genes Dev ; 34(17-18): 1147-1160, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32763910

RESUMO

Identifying modifiers of dosage-sensitive genes involved in neurodegenerative disorders is imperative to discover novel genetic risk factors and potential therapeutic entry points. In this study, we focus on Ataxin-1 (ATXN1), a dosage-sensitive gene involved in the neurodegenerative disease spinocerebellar ataxia type 1 (SCA1). While the precise maintenance of ATXN1 levels is essential to prevent disease, the mechanisms that regulate ATXN1 expression remain largely unknown. We demonstrate that ATXN1's unusually long 5' untranslated region (5' UTR) negatively regulates its expression via posttranscriptional mechanisms. Based on recent reports that microRNAs (miRNAs) can interact with both 3' and 5' UTRs to regulate their target genes, we identify miR760 as a negative regulator that binds to a conserved site in ATXN1's 5' UTR to induce RNA degradation and translational inhibition. We found that delivery of Adeno-associated virus (AAV)-expressing miR760 in the cerebellum reduces ATXN1 levels in vivo and mitigates motor coordination deficits in a mouse model of SCA1. These findings provide new insights into the regulation of ATXN1 levels, present additional evidence for miRNA-mediated gene regulation via 5' UTR binding, and raise the possibility that noncoding mutations in the ATXN1 locus may act as risk factors for yet to be discovered progressive ataxias.


Assuntos
Regiões 5' não Traduzidas/genética , Ataxina-1/genética , Regulação da Expressão Gênica/genética , MicroRNAs/metabolismo , Ataxias Espinocerebelares/genética , Animais , Ataxina-1/metabolismo , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Mutação , Fatores de Risco , Ataxias Espinocerebelares/fisiopatologia
5.
Mov Disord ; 32(11): 1537-1545, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28843013

RESUMO

Dystonia is a common movement disorder that devastates the lives of many patients, but the etiology of this disorder remains poorly understood. Dystonia has traditionally been considered a disorder of the basal ganglia. However, growing evidence suggests that the cerebellum may be involved in certain types of dystonia, raising several questions. Can different types of dystonia be classified as either a basal ganglia disorder or a cerebellar disorder? Is dystonia a network disorder that involves the cerebellum and basal ganglia? If dystonia is a network disorder, how can we target treatments to alleviate symptoms in patients? A recent study by Chen et al, using the pharmacological mouse model of rapid-onset dystonia parkinsonism, has provided some insight into these important questions. They showed that the cerebellum can directly modulate basal ganglia activity through a short latency cerebello-thalamo-basal ganglia pathway. Further, this article and others have provided evidence that in some cases, aberrant cerebello-basal ganglia communication can be involved in dystonia. In this review we examine the evidence for the involvement of the cerebellum and cerebello-basal ganglia interactions in dystonia. We conclude that there is ample evidence to suggest that the cerebellum plays a role in some dystonias, including the early-onset primary torsion dystonia DYT1 and that further studies examining the role of this brain region and its interaction with the basal ganglia in dystonia are warranted. © 2017 International Parkinson and Movement Disorder Society.


Assuntos
Gânglios da Base/fisiopatologia , Cerebelo/fisiopatologia , Distúrbios Distônicos/fisiopatologia , Animais , Humanos
6.
Elife ; 62017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28198698

RESUMO

DYT1 is a debilitating movement disorder caused by loss-of-function mutations in torsinA. How these mutations cause dystonia remains unknown. Mouse models which have embryonically targeted torsinA have failed to recapitulate the dystonia seen in patients, possibly due to differential developmental compensation between rodents and humans. To address this issue, torsinA was acutely knocked down in select brain regions of adult mice using shRNAs. TorsinA knockdown in the cerebellum, but not in the basal ganglia, was sufficient to induce dystonia. In agreement with a potential developmental compensation for loss of torsinA in rodents, torsinA knockdown in the immature cerebellum failed to produce dystonia. Abnormal motor symptoms in knockdown animals were associated with irregular cerebellar output caused by changes in the intrinsic activity of both Purkinje cells and neurons of the deep cerebellar nuclei. These data identify the cerebellum as the main site of dysfunction in DYT1, and offer new therapeutic targets.


Assuntos
Cerebelo/patologia , Distonia Muscular Deformante/genética , Distonia Muscular Deformante/patologia , Chaperonas Moleculares/genética , Animais , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Camundongos
7.
Elife ; 42015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26705335

RESUMO

A new mutant mouse (lamb1t) exhibits intermittent dystonic hindlimb movements and postures when awake, and hyperextension when asleep. Experiments showed co-contraction of opposing muscle groups, and indicated that symptoms depended on the interaction of brain and spinal cord. SNP mapping and exome sequencing identified the dominant causative mutation in the Lamb1 gene. Laminins are extracellular matrix proteins, widely expressed but also known to be important in synapse structure and plasticity. In accordance, awake recording in the cerebellum detected abnormal output from a circuit of two Lamb1-expressing neurons, Purkinje cells and their deep cerebellar nucleus targets, during abnormal postures. We propose that dystonia-like symptoms result from lapses in descending inhibition, exposing excess activity in intrinsic spinal circuits that coordinate muscles. The mouse is a new model for testing how dysfunction in the CNS causes specific abnormal movements and postures.


Assuntos
Encéfalo/patologia , Laminina/genética , Laminina/metabolismo , Transtornos dos Movimentos/patologia , Mutação , Coluna Vertebral/patologia , Animais , Distonia/patologia , Locomoção , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Vias Neurais/patologia , Postura
8.
Neurobiol Dis ; 82: 200-212, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26093171

RESUMO

Loss-of-function mutations in the α3 isoform of the sodium pump are responsible for Rapid Onset Dystonia-Parkinsonism (RDP). A pharmacologic model of RDP replicates the most salient features of RDP, and implicates both the cerebellum and basal ganglia in the disorder; dystonia is associated with aberrant cerebellar output, and the parkinsonism-like features are attributable to the basal ganglia. The pharmacologic agent used to generate the model, ouabain, is selective for sodium pumps. However, close to the infusion sites in vivo it likely affects all sodium pump isoforms. Therefore, it remains to be established whether selective loss of α3-containing sodium pumps replicates the pharmacologic model. Moreover, while the pharmacologic model suggested that aberrant firing of Purkinje cells was the main cause of abnormal cerebellar output, it did not allow the scrutiny of this hypothesis. To address these questions RNA interference using small hairpin RNAs (shRNAs) delivered via adeno-associated viruses (AAV) was used to specifically knockdown α3-containing sodium pumps in different regions of the adult mouse brain. Knockdown of the α3-containing sodium pumps mimicked both the behavioral and electrophysiological changes seen in the pharmacologic model of RDP, recapitulating key aspects of the human disorder. Further, we found that knockdown of the α3 isoform altered the intrinsic pacemaking of Purkinje cells, but not the neurons of the deep cerebellar nuclei. Therefore, acute knockdown of proteins associated with inherited dystonias may be a good strategy for developing phenotypic genetic mouse models where traditional transgenic models have failed to produce symptomatic mice.


Assuntos
Cerebelo/fisiopatologia , Distonia/fisiopatologia , Distúrbios Distônicos/fisiopatologia , Células de Purkinje/fisiologia , ATPase Trocadora de Sódio-Potássio/genética , Animais , Cerebelo/metabolismo , Modelos Animais de Doenças , Distonia/genética , Distúrbios Distônicos/genética , Camundongos , Isoformas de Proteínas/genética , RNA Interferente Pequeno
9.
J Neurosci Res ; 90(8): 1547-56, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22437915

RESUMO

During their development as myelinating cells, oligodendrocyte progenitors (OPC) undergo dramatic changes in the organization of their cytoskeleton. These changes involve an increase in cell branching and in lamella extension, which is important for the ability of oligodendrocytes to myelinate multiple axons in the CNS. We have previously shown that the levels of the actin-associated motor protein nonmuscle myosin II (NMII) decrease as oligodendrocyte differentiate and that inhibition of NMII activity increases branching and myelination, suggesting that NMII is a negative regulator of oligodendrocyte differentiation. In agreement with this interpretation, we have found that overexpression of NMII prevents oligodendrocyte branching and differentiation and that OPC maturation is accelerated in NMII knockout mice as shown by a significant increase in the percentage of mature MBP(+) cells. Although several pathways have been implicated in oligodendrocyte morphogenesis, their specific contribution to the regulation of NMII activity has not been directly examined. We tested the hypothesis that the activity of NMII in OPC is controlled by Fyn kinase via downregulation of RhoA-ROCK-NMII phosphorylation. We found that treatment with PP2 or knockdown of Fyn using siRNA prevents the decrease in myosin phosphorylation normally observed during OPC differentiation and that the inhibition of branching induced by overexpression of constitutively active RhoA can be reversed by treatment with Y27632 or blebbistatin. Taken together, our results demonstrate that Fyn kinase downregulates NMII activity, thus promoting oligodendrocyte morphological differentiation.


Assuntos
Diferenciação Celular/fisiologia , Miosina Tipo II/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Transdução de Sinais/fisiologia , Animais , Citoesqueleto/metabolismo , Regulação para Baixo , Imunofluorescência , Camundongos , Camundongos Knockout , Microscopia Imunoeletrônica , Miosina Tipo II/deficiência , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Fosforilação , Ratos , Transfecção
10.
J Cell Sci ; 124(Pt 22): 3784-96, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22100921

RESUMO

Signaling through cyclic AMP (cAMP) has been implicated in the regulation of Schwann cell (SC) proliferation and differentiation. In quiescent SCs, elevation of cAMP promotes the expression of proteins associated with myelination such as Krox-20 and P0, and downregulation of markers associated with the non-myelinating SC phenotype. We have previously shown that the motor protein myosin II is required for the establishment of normal SC-axon interactions, differentiation and myelination, however, the mechanisms behind these effects are unknown. Here we report that the levels and activity of myosin light chain kinase (MLCK), an enzyme that regulates MLC phosphorylation in non-muscle cells, are dramatically downregulated in SCs after cAMP treatment, in a similar pattern to that of c-Jun, a known inhibitor of myelination. Knockdown of MLCK in SCs mimics the effect of cAMP elevation, inducing plasma membrane expansion and expression of Krox-20 and myelin proteins. Despite activation of myelin gene transcription these cells fail to make compact myelin when placed in contact with axons. Our data indicate that myosin II activity is differentially regulated at various stages during myelination and that in the absence of MLCK the processes of SC differentiation and compact myelin assembly are uncoupled.


Assuntos
Diferenciação Celular , Citoesqueleto/metabolismo , Bainha de Mielina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Células de Schwann/citologia , Células de Schwann/enzimologia , Animais , Células Cultivadas , Quinase de Cadeia Leve de Miosina/genética , Ratos , Células de Schwann/metabolismo
11.
Nat Neurosci ; 13(12): 1472-80, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21057508

RESUMO

Fast axonal conduction depends on myelin, which is formed by Schwann cells in the PNS. We found that the transcription factor Yin Yang 1 (YY1) is crucial for peripheral myelination. Conditional ablation of Yy1 in the Schwann cell lineage resulted in severe hypomyelination, which occurred independently of altered Schwann cell proliferation or apoptosis. In Yy1 mutant mice, Schwann cells established a 1:1 relationship with axons but were unable to myelinate them. The Schwann cells expressed low levels of myelin proteins and of Egr2 (also called Krox20), which is an important regulator of peripheral myelination. In vitro, Schwann cells that lacked Yy1 did not upregulate Egr2 in response to neuregulin1 and did not express myelin protein zero. This phenotype was rescued by overexpression of Egr2. In addition, neuregulin-induced phosphorylation of YY1 was required for transcriptional activation of Egr2. Thus, YY1 emerges as an important activator of peripheral myelination that links neuregulin signaling with Egr2 expression.


Assuntos
Proteína 2 de Resposta de Crescimento Precoce/fisiologia , Fibras Nervosas Mielinizadas/fisiologia , Neuregulina-1/fisiologia , Nervos Periféricos/fisiologia , Transcrição Gênica/fisiologia , Fator de Transcrição YY1/fisiologia , Animais , Células Cultivadas , Proteína 2 de Resposta de Crescimento Precoce/biossíntese , Proteína 2 de Resposta de Crescimento Precoce/genética , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Nervosas Mielinizadas/metabolismo , Neuregulina-1/biossíntese , Neuregulina-1/genética , Nervos Periféricos/metabolismo , Ratos , Células de Schwann/fisiologia , Fator de Transcrição YY1/biossíntese , Fator de Transcrição YY1/genética
12.
J Cell Biol ; 182(6): 1171-84, 2008 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-18794332

RESUMO

The myelin sheath forms by the spiral wrapping of a glial membrane around the axon. The mechanisms responsible for this process are unknown but are likely to involve coordinated changes in the glial cell cytoskeleton. We have found that inhibition of myosin II, a key regulator of actin cytoskeleton dynamics, has remarkably opposite effects on myelin formation by Schwann cells (SC) and oligodendrocytes (OL). Myosin II is necessary for initial interactions between SC and axons, and its inhibition or down-regulation impairs their ability to segregate axons and elongate along them, preventing the formation of a 1:1 relationship, which is critical for peripheral nervous system myelination. In contrast, OL branching, differentiation, and myelin formation are potentiated by inhibition of myosin II. Thus, by controlling the spatial and localized activation of actin polymerization, myosin II regulates SC polarization and OL branching, and by extension their ability to form myelin. Our data indicate that the mechanisms regulating myelination in the peripheral and central nervous systems are distinct.


Assuntos
Sistema Nervoso Central/metabolismo , Bainha de Mielina/metabolismo , Miosina Tipo II/metabolismo , Sistema Nervoso Periférico/metabolismo , Actinas/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Biomarcadores/metabolismo , Caderinas/metabolismo , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Citoesqueleto/metabolismo , Gânglios Espinais/citologia , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Proteína Básica da Mielina/metabolismo , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/genética , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Interferência de RNA , Ratos , Células de Schwann/citologia , Células de Schwann/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA