Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 152(3): 479-91, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374344

RESUMO

Fast axonal transport (FAT) requires consistent energy over long distances to fuel the molecular motors that transport vesicles. We demonstrate that glycolysis provides ATP for the FAT of vesicles. Although inhibiting ATP production from mitochondria did not affect vesicles motility, pharmacological or genetic inhibition of the glycolytic enzyme GAPDH reduced transport in cultured neurons and in Drosophila larvae. GAPDH localizes on vesicles via a huntingtin-dependent mechanism and is transported on fast-moving vesicles within axons. Purified motile vesicles showed GAPDH enzymatic activity and produced ATP. Finally, we show that vesicular GAPDH is necessary and sufficient to provide on-board energy for fast vesicular transport. Although detaching GAPDH from vesicles reduced transport, targeting GAPDH to vesicles was sufficient to promote FAT in GAPDH deficient neurons. This specifically localized glycolytic machinery may supply constant energy, independent of mitochondria, for the processive movement of vesicles over long distances in axons.


Assuntos
Transporte Axonal , Drosophila melanogaster/metabolismo , Glicólise , Neurônios/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Axônios/metabolismo , Encéfalo/citologia , Células Cultivadas , Drosophila melanogaster/crescimento & desenvolvimento , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Camundongos , Mitocôndrias/metabolismo , Ratos
2.
Traffic ; 25(1): e12926, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38084815

RESUMO

In neurons, fast axonal transport (FAT) of vesicles occurs over long distances and requires constant and local energy supply for molecular motors in the form of adenosine triphosphate (ATP). FAT is independent of mitochondrial metabolism. Indeed, the glycolytic machinery is present on vesicles and locally produces ATP, as well as nicotinamide adenine dinucleotide bonded with hydrogen (NADH) and pyruvate, using glucose as a substrate. It remains unclear whether pyruvate is transferred to mitochondria from the vesicles as well as how NADH is recycled into NAD+ on vesicles for continuous glycolysis activity. The optimization of a glycolytic activity test for subcellular compartments allowed the evaluation of the kinetics of vesicular glycolysis in the brain. This revealed that glycolysis is more efficient on vesicles than in the cytosol. We also found that lactate dehydrogenase (LDH) enzymatic activity is required for effective vesicular ATP production. Indeed, inhibition of LDH or the forced degradation of pyruvate inhibited ATP production from axonal vesicles. We found LDHA rather than the B isoform to be enriched on axonal vesicles suggesting a preferential transformation of pyruvate to lactate and a concomitant recycling of NADH into NAD+ on vesicles. Finally, we found that LDHA inhibition dramatically reduces the FAT of both dense-core vesicles and synaptic vesicle precursors in a reconstituted cortico-striatal circuit on-a-chip. Together, this shows that aerobic glycolysis is required to supply energy for vesicular transport in neurons, similar to the Warburg effect.


Assuntos
Glicólise , NAD , NAD/metabolismo , Glicólise/fisiologia , Axônios/metabolismo , Trifosfato de Adenosina/metabolismo , Piruvatos/metabolismo
3.
Mol Psychiatry ; 27(3): 1805-1815, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35165396

RESUMO

Sensorimotor information processing underlies normal cognitive and behavioral traits and has classically been evaluated through prepulse inhibition (PPI) of a startle reflex. PPI is a behavioral dimension deregulated in several neurological and psychiatric disorders, yet the mechanisms underlying the cross-diagnostic nature of PPI deficits across these conditions remain to be understood. To identify circuitry mechanisms for PPI, we performed circuitry recording over the prefrontal cortex and striatum, two brain regions previously implicated in PPI, using wild-type (WT) mice compared to Disc1-locus-impairment (LI) mice, a model representing neuropsychiatric conditions. We demonstrated that the corticostriatal projection regulates neurophysiological responses during the PPI testing in WT, whereas these circuitry responses were disrupted in Disc1-LI mice. Because our biochemical analyses revealed attenuated brain-derived neurotrophic factor (Bdnf) transport along the corticostriatal circuit in Disc1-LI mice, we investigated the potential role of Bdnf in this circuitry for regulation of PPI. Virus-mediated delivery of Bdnf into the striatum rescued PPI deficits in Disc1-LI mice. Pharmacologically augmenting Bdnf transport by chronic lithium administration, partly via phosphorylation of Huntingtin (Htt) serine-421 and its integration into the motor machinery, restored striatal Bdnf levels and rescued PPI deficits in Disc1-LI mice. Furthermore, reducing the cortical Bdnf expression negated this rescuing effect of lithium, confirming the key role of Bdnf in lithium-mediated PPI rescuing. Collectively, the data suggest that striatal Bdnf supply, collaboratively regulated by Htt and Disc1 along the corticostriatal circuit, is involved in sensorimotor gating, highlighting the utility of dimensional approach in investigating pathophysiological mechanisms across neuropsychiatric disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Corpo Estriado , Proteínas do Tecido Nervoso , Córtex Pré-Frontal , Inibição Pré-Pulso , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/metabolismo , Humanos , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/metabolismo , Inibição Pré-Pulso/fisiologia , Reflexo de Sobressalto/fisiologia , Filtro Sensorial/fisiologia
4.
Neurobiol Dis ; 173: 105857, 2022 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-36075537

RESUMO

Huntington disease (HD) is a neurodegenerative disorder caused by polyglutamine-encoding CAG repeat expansion in the huntingtin (HTT) gene. HTT is involved in the axonal transport of vesicles containing brain-derived neurotrophic factor (BDNF). In HD, diminished BDNF transport leads to reduced BDNF delivery to the striatum, contributing to striatal and cortical neuronal death. Pridopidine is a selective and potent sigma-1 receptor (S1R) agonist currently in clinical development for HD. The S1R is located at the endoplasmic reticulum (ER)-mitochondria interface, where it regulates key cellular pathways commonly impaired in neurodegenerative diseases. We used a microfluidic device that reconstitutes the corticostriatal network, allowing the investigation of presynaptic dynamics, synaptic morphology and transmission, and postsynaptic signaling. Culturing primary neurons from the HD mouse model HdhCAG140/+ provides a "disease-on-a-chip" platform ideal for investigating pathogenic mechanisms and drug activity. Pridopidine rescued the trafficking of BDNF and TrkB resulting in an increased neurotrophin signaling at the synapse. This increased the capacity of HD neurons to release glutamate and restored homeostasis at the corticostriatal synapse. These data suggest that pridopidine enhances the availability of corticostriatal BDNF via S1R activation, leading to neuroprotective effects.


Assuntos
Doença de Huntington , Fármacos Neuroprotetores , Animais , Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Glutamatos/farmacologia , Glutamatos/uso terapêutico , Homeostase , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Doença de Huntington/genética , Dispositivos Lab-On-A-Chip , Camundongos , Fármacos Neuroprotetores/farmacologia , Piperidinas , Sinapses/metabolismo
5.
Brain ; 142(8): 2432-2450, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31286142

RESUMO

Dysfunctions in brain cholesterol homeostasis have been extensively related to brain disorders. The main pathway for brain cholesterol elimination is its hydroxylation into 24S-hydroxycholesterol by the cholesterol 24-hydrolase, CYP46A1. Increasing evidence suggests that CYP46A1 has a role in the pathogenesis and progression of neurodegenerative disorders, and that increasing its levels in the brain is neuroprotective. However, the mechanisms underlying this neuroprotection remain to be fully understood. Huntington's disease is a fatal autosomal dominant neurodegenerative disease caused by an abnormal CAG expansion in huntingtin's gene. Among the multiple cellular and molecular dysfunctions caused by this mutation, altered brain cholesterol homeostasis has been described in patients and animal models as a critical event in Huntington's disease. Here, we demonstrate that a gene therapy approach based on the delivery of CYP46A1, the rate-limiting enzyme for cholesterol degradation in the brain, has a long-lasting neuroprotective effect in Huntington's disease and counteracts multiple detrimental effects of the mutated huntingtin. In zQ175 Huntington's disease knock-in mice, CYP46A1 prevented neuronal dysfunctions and restored cholesterol homeostasis. These events were associated to a specific striatal transcriptomic signature that compensates for multiple mHTT-induced dysfunctions. We thus explored the mechanisms for these compensations and showed an improvement of synaptic activity and connectivity along with the stimulation of the proteasome and autophagy machineries, which participate to the clearance of mutant huntingtin (mHTT) aggregates. Furthermore, BDNF vesicle axonal transport and TrkB endosome trafficking were restored in a cellular model of Huntington's disease. These results highlight the large-scale beneficial effect of restoring cholesterol homeostasis in neurodegenerative diseases and give new opportunities for developing innovative disease-modifying strategies in Huntington's disease.


Assuntos
Encéfalo/metabolismo , Colesterol 24-Hidroxilase/uso terapêutico , Colesterol/metabolismo , Terapia Genética , Vetores Genéticos/uso terapêutico , Doença de Huntington/terapia , Fármacos Neuroprotetores/uso terapêutico , Animais , Autofagia , Transporte Axonal , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Células Cultivadas , Córtex Cerebral/fisiopatologia , Colesterol 24-Hidroxilase/genética , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Dependovirus/genética , Endossomos/metabolismo , Técnicas de Introdução de Genes , Vetores Genéticos/genética , Humanos , Proteína Huntingtina/genética , Doença de Huntington/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/fisiopatologia , Fármacos Neuroprotetores/administração & dosagem , Oxisteróis/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Agregação Patológica de Proteínas , Proteínas Tirosina Quinases/fisiologia , Teste de Desempenho do Rota-Rod , Transmissão Sináptica , Transcriptoma
6.
EMBO J ; 34(17): 2255-71, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-26165689

RESUMO

Cleavage of mutant huntingtin (HTT) is an essential process in Huntington's disease (HD), an inherited neurodegenerative disorder. Cleavage generates N-ter fragments that contain the polyQ stretch and whose nuclear toxicity is well established. However, the functional defects induced by cleavage of full-length HTT remain elusive. Moreover, the contribution of non-polyQ C-terminal fragments is unknown. Using time- and site-specific control of full-length HTT proteolysis, we show that specific cleavages are required to disrupt intramolecular interactions within HTT and to cause toxicity in cells and flies. Surprisingly, in addition to the canonical pathogenic N-ter fragments, the C-ter fragments generated, that do not contain the polyQ stretch, induced toxicity via dilation of the endoplasmic reticulum (ER) and increased ER stress. C-ter HTT bound to dynamin 1 and subsequently impaired its activity at ER membranes. Our findings support a role for HTT on dynamin 1 function and ER homoeostasis. Proteolysis-induced alteration of this function may be relevant to disease.


Assuntos
Dinamina I/metabolismo , Doença de Huntington/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeos/metabolismo , Proteólise , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Animais , Proteínas de Drosophila , Drosophila melanogaster , Dinamina I/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/genética , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas do Tecido Nervoso/genética , Peptídeos/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
7.
Brain ; 141(5): 1434-1454, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29534157

RESUMO

The neurobiological functions of a number of kinases expressed in the brain are unknown. Here, we report new findings on DCLK3 (doublecortin like kinase 3), which is preferentially expressed in neurons in the striatum and dentate gyrus. Its function has never been investigated. DCLK3 expression is markedly reduced in Huntington's disease. Recent data obtained in studies related to cancer suggest DCLK3 could have an anti-apoptotic effect. Thus, we hypothesized that early loss of DCLK3 in Huntington's disease may render striatal neurons more susceptible to mutant huntingtin (mHtt). We discovered that DCLK3 silencing in the striatum of mice exacerbated the toxicity of an N-terminal fragment of mHtt. Conversely, overexpression of DCLK3 reduced neurodegeneration produced by mHtt. DCLK3 also produced beneficial effects on motor symptoms in a knock-in mouse model of Huntington's disease. Using different mutants of DCLK3, we found that the kinase activity of the protein plays a key role in neuroprotection. To investigate the potential mechanisms underlying DCLK3 effects, we studied the transcriptional changes produced by the kinase domain in human striatal neurons in culture. Results show that DCLK3 regulates in a kinase-dependent manner the expression of many genes involved in transcription regulation and nucleosome/chromatin remodelling. Consistent with this, histological evaluation showed DCLK3 is present in the nucleus of striatal neurons and, protein-protein interaction experiments suggested that the kinase domain interacts with zinc finger proteins, including the transcriptional activator adaptor TADA3, a core component of the Spt-ada-Gcn5 acetyltransferase (SAGA) complex which links histone acetylation to the transcription machinery. Our novel findings suggest that the presence of DCLK3 in striatal neurons may play a key role in transcription regulation and chromatin remodelling in these brain cells, and show that reduced expression of the kinase in Huntington's disease could render the striatum highly vulnerable to neurodegeneration.


Assuntos
Corpo Estriado/enzimologia , Proteína Huntingtina/genética , Doença de Huntington/terapia , Mutação/genética , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Quinases Semelhantes a Duplacortina , Regulação para Baixo/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Força da Mão/fisiologia , Doença de Huntington/genética , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Neurônios/metabolismo , Fosfopiruvato Hidratase/metabolismo , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Mov Disord ; 32(6): 932-936, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28436572

RESUMO

BACKGROUND: Cysteamine has been demonstrated as potentially effective in numerous animal models of Huntington's disease. METHODS: Ninety-six patients with early-stage Huntington's disease were randomized to 1200 mg delayed-release cysteamine bitartrate or placebo daily for 18 months. The primary end point was the change from baseline in the UHDRS Total Motor Score. A linear mixed-effects model for repeated measures was used to assess treatment effect, expressed as the least-squares mean difference of cysteamine minus placebo, with negative values indicating less deterioration relative to placebo. RESULTS: At 18 months, the treatment effect was not statistically significant - least-squares mean difference, -1.5 ± 1.71 (P = 0.385) - although this did represent less mean deterioration from baseline for the treated group relative to placebo. Treatment with cysteamine was safe and well tolerated. CONCLUSIONS: Efficacy of cysteamine was not demonstrated in this study population of patients with Huntington's disease. Post hoc analyses indicate the need for definitive future studies. © 2017 International Parkinson and Movement Disorder Society.


Assuntos
Cisteamina/farmacologia , Eliminadores de Cistina/farmacologia , Doença de Huntington/tratamento farmacológico , Adulto , Idoso , Cisteamina/administração & dosagem , Cisteamina/efeitos adversos , Eliminadores de Cistina/administração & dosagem , Eliminadores de Cistina/efeitos adversos , Preparações de Ação Retardada , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
9.
Proc Natl Acad Sci U S A ; 111(47): 16889-94, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385587

RESUMO

Although dominant gain-of-function triplet repeat expansions in the Huntingtin (HTT) gene are the underlying cause of Huntington disease (HD), understanding the normal functions of nonmutant HTT protein has remained a challenge. We report here findings that suggest that HTT plays a significant role in selective autophagy. Loss of HTT function in Drosophila disrupts starvation-induced autophagy in larvae and conditional knockout of HTT in the mouse CNS causes characteristic cellular hallmarks of disrupted autophagy, including an accumulation of striatal p62/SQSTM1 over time. We observe that specific domains of HTT have structural similarities to yeast Atg proteins that function in selective autophagy, and in particular that the C-terminal domain of HTT shares structural similarity to yeast Atg11, an autophagic scaffold protein. To explore possible functional similarity between HTT and Atg11, we investigated whether the C-terminal domain of HTT interacts with mammalian counterparts of yeast Atg11-interacting proteins. Strikingly, this domain of HTT coimmunoprecipitates with several key Atg11 interactors, including the Atg1/Unc-51-like autophagy activating kinase 1 kinase complex, autophagic receptor proteins, and mammalian Atg8 homologs. Mutation of a phylogenetically conserved WXXL domain in a C-terminal HTT fragment reduces coprecipitation with mammalian Atg8 homolog GABARAPL1, suggesting a direct interaction. Collectively, these data support a possible central role for HTT as an Atg11-like scaffold protein. These findings have relevance to both mechanisms of disease pathogenesis and to therapeutic intervention strategies that reduce levels of both mutant and normal HTT.


Assuntos
Autofagia , Proteínas Associadas aos Microtúbulos/fisiologia , Animais , Animais Geneticamente Modificados , Drosophila , Proteínas de Drosophila , Proteína Huntingtina , Camundongos , Proteínas Associadas aos Microtúbulos/genética
10.
J Neurosci ; 33(15): 6298-309, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575829

RESUMO

Huntingtin (HTT), the protein mutated in Huntington's disease (HD), controls transport of the neurotrophin, brain-derived neurotrophic factor (BDNF), within corticostriatal neurons. Transport and delivery of BDNF to the striatum are reduced in disease, which contributes to striatal neuron degeneration. BDNF released by cortical neurons activates TrkB receptors at striatal dendrites to promote striatum survival. However, it remains to be determined whether transport of TrkB, the BDNF receptor, depends on HTT and whether such transport is altered in mutant situation. Here we show that TrkB binds to and colocalizes with HTT and dynein. Silencing HTT reduces vesicular transport of TrkB in striatal neurons. In HD, the polyQ expansion in HTT alters the binding of TrkB-containing vesicles to microtubules and reduces transport. Using a combination of microfluidic devices that isolate dendrites from cell bodies and BDNF coupled to quantum dots, we selectively analyzed TrkB retrograde transport in response to BDNF stimulation at dendrite terminals. We show that the retrograde transport of TrkB vesicles within striatal dendrites and the BDNF/TrkB-induced signaling through ERK phosphorylation and c-fos induction are decreased in neurons from an HD mouse model. Together, our findings demonstrate that HTT is a crucial regulator of TrkB trafficking. Transport defects in HD are not restricted to BDNF transport in cortical neurons but also affect trafficking of its ligand-bound receptor in the striatal neurons. This transport alteration may further impair BDNF-TrkB survival signaling within the corticostriatal connection that is most affected in HD.


Assuntos
Corpo Estriado/metabolismo , Dendritos/metabolismo , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Receptor trkB/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Dineínas/metabolismo , Proteína Huntingtina , Doença de Huntington/genética , Camundongos , Microtúbulos/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Cultura Primária de Células , Transporte Proteico , Ratos , Transdução de Sinais/genética , Vesículas Transportadoras/genética , Vesículas Transportadoras/metabolismo
11.
J Neurosci ; 33(20): 8608-20, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678106

RESUMO

Huntington disease (HD) is associated with early psychiatric symptoms including anxiety and depression. Here, we demonstrate that wild-type huntingtin, the protein mutated in HD, modulates anxiety/depression-related behaviors according to its phosphorylation at serines 1181 and 1201. Genetic phospho-ablation at serines 1181 and 1201 in mouse reduces basal levels of anxiety/depression-like behaviors. We observe that the reduction in anxiety/depression-like phenotypes is associated with increased adult hippocampal neurogenesis. By improving the attachment of molecular motors to microtubules, huntingtin dephosphorylation increases axonal transport of BDNF, a crucial factor for hippocampal adult neurogenesis. Consequently, the huntingtin-mediated increased BDNF dynamics lead to an increased delivery and signaling of hippocampal BDNF. These results support the notion that huntingtin participates in anxiety and depression-like behavior and is thus relevant to the etiology of mood disorders and anxiety/depression in HD.


Assuntos
Ansiedade/patologia , Depressão/patologia , Hipocampo/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Neurogênese/genética , Proteínas Nucleares/metabolismo , Análise de Variância , Animais , Ansiedade/genética , Ansiedade/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Bromodesoxiuridina/metabolismo , Depressão/fisiopatologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Proteína Huntingtina , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese/fisiologia , Neuropeptídeos/metabolismo , Proteínas Nucleares/genética , Fosforilação/genética , Transporte Proteico/genética , Serina/genética , Serina/metabolismo
12.
Biomaterials ; 305: 122426, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38134473

RESUMO

Microglial cells, as the primary defense line in the central nervous system, play a crucial role in responding to various mechanical signals that can trigger their activation. Despite extensive research on the impact of chemical signaling on brain cells, the understanding of mechanical signaling in microglia remains limited. To bridge this gap, we subjected microglial cells to a singular mechanical stretch and compared their responses with those induced by lipopolysaccharide treatment, a well-established chemical activator. Here we show that stretching microglial cells leads to their activation, highlighting their significant mechanosensitivity. Stretched microglial cells exhibited distinct features, including elevated levels of Iba1 protein, a denser actin cytoskeleton, and increased persistence in migration. Unlike LPS-treated microglial cells, the secretory profile of chemokines and cytokines remained largely unchanged in response to stretching, except for TNF-α. Intriguingly, a single stretch injury resulted in more compacted chromatin and DNA damage, suggesting potential long-term genomic instabilities in stretched microglia. Using compartmentalized microfluidic chambers with neuronal networks, we observed that stretched microglial cells exhibited enhanced phagocytic and synaptic stripping activities. These findings collectively suggest that stretching events can unlock the immune potential of microglial cells, contributing to the maintenance of brain tissue homeostasis following mechanical injury.


Assuntos
Microglia , Fagócitos , Microglia/metabolismo , Sistema Nervoso Central , Encéfalo , Transdução de Sinais , Lipopolissacarídeos/farmacologia
13.
J Huntingtons Dis ; 13(1): 41-53, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38427495

RESUMO

Background: Mutations in the Huntingtin (HTT) gene cause Huntington's disease (HD), a neurodegenerative disorder. As a scaffold protein, HTT is involved in numerous cellular functions, but its normal and pathogenic functions during human forebrain development are poorly understood. Objective: To investigate the developmental component of HD, with a specific emphasis on understanding the functions of wild-type and mutant HTT alleles during forebrain neuron development in individuals carrying HD mutations. Methods: We used CRISPR/Cas9 gene-editing technology to disrupt the ATG region of the HTT gene via non-homologous end joining to produce mono- or biallelic HTT knock-out human induced pluripotent stem cell (iPSC) clones. Results: We showed that the loss of wild-type, mutant, or both HTT isoforms does not affect the pluripotency of iPSCs or their transition into neural cells. However, we observed that HTT loss causes division impairments in forebrain neuro-epithelial cells and alters maturation of striatal projection neurons (SPNs) particularly in the acquisition of DARPP32 expression, a key functional marker of SPNs. Finally, young post-mitotic neurons derived from HTT-/- human iPSCs display cellular dysfunctions observed in adult HD neurons. Conclusions: We described a novel collection of isogenic clones with mono- and biallelic HTT inactivation that complement existing HD-hiPSC isogenic series to explore HTT functions and test therapeutic strategies in particular HTT-lowering drugs. Characterizing neural and neuronal derivatives from human iPSCs of this collection, we show evidence that HTT loss or mutation has impacts on neuro-epithelial and striatal neurons maturation, and on basal DNA damage and BDNF axonal transport in post-mitotic neurons.


Assuntos
Doença de Huntington , Células-Tronco Pluripotentes Induzidas , Adulto , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Corpo Estriado/metabolismo , Alelos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
14.
Nat Chem Biol ; 7(12): 925-34, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037470

RESUMO

Polyglutamine (polyQ) stretches exceeding a threshold length confer a toxic function to proteins that contain them and cause at least nine neurological disorders. The basis for this toxicity threshold is unclear. Although polyQ expansions render proteins prone to aggregate into inclusion bodies, this may be a neuronal coping response to more toxic forms of polyQ. The exact structure of these more toxic forms is unknown. Here we show that the monoclonal antibody 3B5H10 recognizes a species of polyQ protein in situ that strongly predicts neuronal death. The epitope selectively appears among some of the many low-molecular-weight conformational states assumed by expanded polyQ and disappears in higher-molecular-weight aggregated forms, such as inclusion bodies. These results suggest that protein monomers and possibly small oligomers containing expanded polyQ stretches can adopt a conformation that is recognized by 3B5H10 and is toxic or closely related to a toxic species.


Assuntos
Doenças Neurodegenerativas/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Peptídeos/química , Peptídeos/toxicidade , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Epitopos/química , Epitopos/imunologia , Epitopos/toxicidade , Células HEK293 , Humanos , Corpos de Inclusão/química , Peso Molecular , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Peptídeos/imunologia , Relação Estrutura-Atividade , Expansão das Repetições de Trinucleotídeos
15.
Neuroscience ; 518: 162-177, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35995336

RESUMO

In several forms of dementia, such as Alzheimer's disease, the cytoskeleton-associated protein tau undergoes proteolysis, giving rise to fragments that have a toxic impact on neuronal homeostasis. How these fragments interact with cellular structures, in particular with the cytoskeleton, is currently incompletely understood. Here, we developed a method, derived from a Tobacco Etch Virus (TEV) protease system, to induce controlled cleavage of tau at specific sites. Five tau proteins containing specific TEV recognition sites corresponding to pathological proteolytic sites were engineered, and tagged with GFP at one end and mCherry at the other. After a controlled cleavage to produce GFP-N-terminal and C-terminal-mCherry fragments, we followed the fate of tau fragments in cells. Our results showed that whole engineered tau proteins associate with the cytoskeleton similarly to the non-modified tau, whereas tau fragments adopted different localizations with respect to the actin and microtubule cytoskeletons. These distinct localizations were confirmed by expressing each separate fragment in cells. Some cleavages - in particular cleavages at amino-acid positions 124 or 256 - displayed a certain level of cellular toxicity, with an unusual relocalization of the N-terminal fragments to the nucleus. Based on the data presented here, inducible cleavage of tau by the TEV protease appears to be a valuable tool to reproduce tau fragmentation in cells and study the resulting consequences on cell physiology.


Assuntos
Doença de Alzheimer , Proteínas tau , Humanos , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Proteólise , Neurônios/metabolismo , Núcleo Celular/metabolismo
16.
Elife ; 122023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37431882

RESUMO

Neurotransmitters are released at synapses by synaptic vesicles (SVs), which originate from SV precursors (SVPs) that have traveled along the axon. Because each synapse maintains a pool of SVs, only a small fraction of which are released, it has been thought that axonal transport of SVPs does not affect synaptic function. Here, studying the corticostriatal network both in microfluidic devices and in mice, we find that phosphorylation of the Huntingtin protein (HTT) increases axonal transport of SVPs and synaptic glutamate release by recruiting the kinesin motor KIF1A. In mice, constitutive HTT phosphorylation causes SV over-accumulation at synapses, increases the probability of SV release, and impairs motor skill learning on the rotating rod. Silencing KIF1A in these mice restored SV transport and motor skill learning to wild-type levels. Axonal SVP transport within the corticostriatal network thus influences synaptic plasticity and motor skill learning.

17.
Neuron ; 111(18): 2881-2898.e12, 2023 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-37442131

RESUMO

In the adult mammalian central nervous system (CNS), axons fail to regenerate spontaneously after injury because of a combination of extrinsic and intrinsic factors. Despite recent advances targeting the intrinsic regenerative properties of adult neurons, the molecular mechanisms underlying axon regeneration are not fully understood. Here, we uncover a regulatory mechanism that controls the expression of key proteins involved in regeneration at the translational level. Our results show that mRNA-specific translation is critical for promoting axon regeneration. Indeed, we demonstrate that specific ribosome-interacting proteins, such as the protein Huntingtin (HTT), selectively control the translation of a specific subset of mRNAs. Moreover, modulating the expression of these translationally regulated mRNAs is crucial for promoting axon regeneration. Altogether, our findings highlight that selective translation through the customization of the translational complex is a key mechanism of axon regeneration with major implications in the development of therapeutic strategies for CNS repair.


Assuntos
Axônios , Regeneração Nervosa , Animais , Axônios/metabolismo , Regeneração Nervosa/genética , Sistema Nervoso Central/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Mamíferos/metabolismo
18.
J Gen Physiol ; 155(1)2023 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-36409218

RESUMO

The expression of the Huntingtin protein, well known for its involvement in the neurodegenerative Huntington's disease, has been confirmed in skeletal muscle. The impact of HTT deficiency was studied in human skeletal muscle cell lines and in a mouse model with inducible and muscle-specific HTT deletion. Characterization of calcium fluxes in the knock-out cell lines demonstrated a reduction in excitation-contraction (EC) coupling, related to an alteration in the coupling between the dihydropyridine receptor and the ryanodine receptor, and an increase in the amount of calcium stored within the sarcoplasmic reticulum, linked to the hyperactivity of store-operated calcium entry (SOCE). Immunoprecipitation experiments demonstrated an association of HTT with junctophilin 1 (JPH1) and stromal interaction molecule 1 (STIM1), both providing clues on the functional effects of HTT deletion on calcium fluxes. Characterization of muscle strength and muscle anatomy of the muscle-specific HTT-KO mice demonstrated that HTT deletion induced moderate muscle weakness and mild muscle atrophy associated with histological abnormalities, similar to the phenotype observed in tubular aggregate myopathy. Altogether, this study points toward the hypotheses of the involvement of HTT in EC coupling via its interaction with JPH1, and on SOCE via its interaction with JPH1 and/or STIM1.


Assuntos
Cálcio , Retículo Sarcoplasmático , Camundongos , Humanos , Animais , Cálcio/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Retículo Sarcoplasmático/metabolismo , Músculo Esquelético/metabolismo , Acoplamento Excitação-Contração/fisiologia
19.
iScience ; 26(5): 106674, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-37182099

RESUMO

Non-lethal caspase activation (NLCA) has been linked to neurodevelopmental processes. However, how neurons control NLCA remains elusive. Here, we focused on Bcl-xL, a Bcl-2 homolog regulating caspase activation through the mitochondria. We generated a mouse model, referred to as ER-xL, in which Bcl-xL is absent in the mitochondria, yet present in the endoplasmic reticulum. Unlike bclx knockout mice that died at E13.5, ER-xL mice survived embryonic development but died post-partum because of altered feeding behavior. Enhanced caspase-3 activity was observed in the brain and the spinal cord white matter, but not the gray matter. No increase in cell death was observed in ER-xL cortical neurons, suggesting that the observed caspase-3 activation was apoptosis-independent. ER-xL neurons displayed increased caspase-3 activity in the neurites, resulting in impaired axon arborescence and synaptogenesis. Together, our findings suggest that mitochondrial Bcl-xL finely tunes caspase-3 through Drp-1-dependent mitochondrial fission, which is critical to neural network design.

20.
Neurobiol Dis ; 45(2): 786-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22127389

RESUMO

Mecp2 deficiency or overexpression causes a wide spectrum of neurological diseases in humans among which Rett Syndrome is the prototype. Pathogenic mechanisms are thought to involve transcriptional deregulation of target genes such as Bdnf together with defects in the general transcriptional program of affected cells. Here we found that two master genes, Huntingtin (Htt) and huntingtin-associated protein (Hap1), involved in the control of Bdnf axonal transport, are altered in the brain of Mecp2-deficient mice. We also revealed an in vivo defect of Bdnf transport throughout the cortico striatal pathway of Mecp2-deficient animals. We found that the velocity of Bdnf-containing vesicles is reduced in vitro in the Mecp2-deficient axons and this deficit can be rescued by the re-expression of Mecp2. The defect in axonal transport is not restricted to Bdnf since transport of the amyloid precursor protein (App) that is Htt and Hap1-dependent is also altered. Finally, treating Mecp2-deficient mice with cysteamine, a molecule increasing the secretion of Bdnf vesicles, improved the lifespan and reduced motor defects, suggesting a new therapeutic strategy for Rett syndrome.


Assuntos
Transporte Axonal/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Imunofluorescência , Perfilação da Expressão Gênica , Proteína Huntingtina , Imuno-Histoquímica , Masculino , Proteína 2 de Ligação a Metil-CpG/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA