Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(6): 107313, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38657864

RESUMO

Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.


Assuntos
Cricetulus , Proteínas Relacionadas a Receptor de LDL , Proteínas de Membrana Transportadoras , Proteínas tau , Humanos , Proteínas tau/metabolismo , Proteínas tau/genética , Animais , Células CHO , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Linhagem Celular Tumoral , Ligação Proteica , Transporte Proteico
2.
Brain ; 2023 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-37633260

RESUMO

Huntington's disease (HD) results from expansion of a polyglutamine tract (polyQ) in mutant huntingtin (mHTT) protein, but mechanisms underlying polyQ expansion-mediated toxic gain-of-mHTT function remain elusive. Here, deletion and antibody-based experiments revealed that a proline-rich domain (PRD) adjacent to the polyQ tract is necessary for mutant huntingtin (mHTT) to inhibit fast axonal transport and promote axonal pathology in cultured mammalian neurons. Further, polypeptides corresponding to subregions of the PRD sufficed to elicit the toxic effect on fast axonal transport, which was mediated by JNK kinases and involved PRD binding to one or more SH3-domain containing proteins. Collectively, these data suggested a mechanism whereby polyQ tract expansion in mHTT promotes aberrant PRD exposure and interactions of this domain with SH3 domain-containing proteins including some involved in activation of JNK kinases. In support, biochemical and immunohistochemical experiments linked aberrant PRD exposure to increased JNK activation in striatal tissues of the zQ175 mouse model and from post-mortem HD patients. Collectively, these findings support a critical role of PRD on mHTT toxicity, suggesting a novel framework for the potential development of therapies aimed to halt or reduce axonal pathology in HD.

3.
Neuroimage ; 225: 117485, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33132186

RESUMO

Compatible (positive approaching and negative avoiding) and incompatible (positive avoiding and negative approaching) behavior are of great significance for biological adaptation and survival. Previous research has found that reaction times of compatible behavior are shorter than the incompatible behavior, which is termed the stimulus-response compatibility (SRC) effect. However, the underlying neurophysiological mechanisms of the SRC effect applied to affective stimuli is still unclear. Here, we investigated preparatory activities in both the left and right primary motor cortex (M1) before the execution of an approaching-avoiding behavior using the right index finger in a manikin task based on self-identity. The results showed significantly shorter reaction times for compatible than incompatible behavior. Most importantly, motor-evoked potential (MEP) amplitudes from left M1 stimulation were significantly higher during compatible behavior than incompatible behavior at 150 and 200 ms after stimulus presentation, whereas the reversed was observed for right M1 stimulation with lower MEP amplitude in compatible compared to incompatible behavior at 150 ms. The current findings revealed the compatibility effect at both behavioral and neurophysiological levels, indicating that the affective SRC effect occurs early in the motor cortices during stimulus processing, and MEP modulation at this early processing stage could be a physiological marker of the affective SRC effect.


Assuntos
Córtex Motor/fisiologia , Desempenho Psicomotor/fisiologia , Potencial Evocado Motor/fisiologia , Feminino , Humanos , Masculino , Manequins , Tempo de Reação/fisiologia , Estimulação Magnética Transcraniana , Adulto Jovem
4.
Proc Natl Acad Sci U S A ; 115(48): E11406-E11414, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30429313

RESUMO

Drug receptor site occupancy is a central pharmacology parameter that quantitatively relates the biochemistry of drug binding to the biology of drug action. Taxanes and epothilones bind to overlapping sites in microtubules (MTs) and stabilize them. They are used to treat cancer and are under investigation for neurodegeneration. In cells, they cause concentration-dependent inhibition of MT dynamics and perturbation of mitosis, but the degree of site occupancy required to trigger different effects has not been measured. We report a live cell assay for taxane-site occupancy, and relationships between site occupancy and biological effects across four drugs and two cell lines. By normalizing to site occupancy, we were able to quantitatively compare drug activities and cell sensitivities independent of differences in drug affinity and uptake/efflux kinetics. Across all drugs and cells tested, we found that inhibition of MT dynamics, postmitotic micronucleation, and mitotic arrest required successively higher site occupancy. We also found interesting differences between cells and drugs, for example, insensitivity of the spindle assembly checkpoint to site occupancy. By extending our assay to a mouse xenograft tumor model, we estimated the initial site occupancy required for paclitaxel to completely prevent tumor growth as 80%. The most important cellular action of taxanes for cancer treatment may be formation of micronuclei, which occurs over a broad range of site occupancies.


Assuntos
Antineoplásicos/metabolismo , Hidrocarbonetos Aromáticos com Pontes/metabolismo , Taxoides/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Transporte Biológico , Hidrocarbonetos Aromáticos com Pontes/química , Hidrocarbonetos Aromáticos com Pontes/farmacologia , Linhagem Celular Tumoral , Epotilonas/química , Epotilonas/metabolismo , Epotilonas/farmacologia , Humanos , Cinética , Microscopia , Microtúbulos/química , Microtúbulos/metabolismo , Taxoides/química , Taxoides/farmacologia
5.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-32947863

RESUMO

Nucleoside diphosphate kinases (NDPK) are oligomeric proteins involved in the synthesis of nucleoside triphosphates. Their tridimensional structure has been solved by X-ray crystallography and shows that individual subunits present a conserved ferredoxin fold of about 140 residues in prokaryotes, archaea, eukaryotes and viruses. Monomers are functionally independent from each other inside NDPK complexes and the nucleoside kinase catalytic mechanism involves transient phosphorylation of the conserved catalytic histidine. To be active, monomers must assemble into conserved head to tail dimers, which further assemble into hexamers or tetramers. The interfaces between these oligomeric states are very different but, surprisingly, the assembly structure barely affects the catalytic efficiency of the enzyme. While it has been shown that assembly into hexamers induces full formation of the catalytic site and stabilizes the complex, it is unclear why assembly into tetramers is required for function. Several additional activities have been revealed for NDPK, especially in metastasis spreading, cytoskeleton dynamics, DNA binding and membrane remodeling. However, we still lack the high resolution structural data of NDPK in complex with different partners, which is necessary for deciphering the mechanism of these diverse functions. In this review we discuss advances in the structure, folding and stability of NDPKs.


Assuntos
Proteínas de Bactérias/química , Núcleosídeo-Difosfato Quinase/química , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Humanos , Modelos Moleculares , Mutação , Nucleosídeo NM23 Difosfato Quinases/química , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Estabilidade Proteica , Proteínas de Protozoários/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
6.
Hum Mol Genet ; 26(12): 2321-2334, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28398512

RESUMO

Mutations of various genes cause hereditary spastic paraplegia (HSP), a neurological disease involving dying-back degeneration of upper motor neurons. From these, mutations in the SPAST gene encoding the microtubule-severing protein spastin account for most HSP cases. Cumulative genetic and experimental evidence suggests that alterations in various intracellular trafficking events, including fast axonal transport (FAT), may contribute to HSP pathogenesis. However, the mechanisms linking SPAST mutations to such deficits remain largely unknown. Experiments presented here using isolated squid axoplasm reveal inhibition of FAT as a common toxic effect elicited by spastin proteins with different HSP mutations, independent of microtubule-binding or severing activity. Mutant spastin proteins produce this toxic effect only when presented as the tissue-specific M1 isoform, not when presented as the ubiquitously-expressed shorter M87 isoform. Biochemical and pharmacological experiments further indicate that the toxic effects of mutant M1 spastins on FAT involve casein kinase 2 (CK2) activation. In mammalian cells, expression of mutant M1 spastins, but not their mutant M87 counterparts, promotes abnormalities in the distribution of intracellular organelles that are correctable by pharmacological CK2 inhibition. Collectively, these results demonstrate isoform-specific toxic effects of mutant M1 spastin on FAT, and identify CK2 as a critical mediator of these effects.


Assuntos
Adenosina Trifosfatases/genética , Transporte Axonal/genética , Adenosina Trifosfatases/metabolismo , Animais , Transporte Axonal/fisiologia , Caseína Quinase II/metabolismo , Células Cultivadas , Decapodiformes , Modelos Animais de Doenças , Fibroblastos , Humanos , Microtúbulos/metabolismo , Neurônios Motores/metabolismo , Proteínas Mutantes/metabolismo , Mutação , Isoformas de Proteínas/genética , Transporte Proteico/fisiologia , Ratos , Paraplegia Espástica Hereditária/genética , Paraplegia Espástica Hereditária/metabolismo , Espastina
7.
Proc Natl Acad Sci U S A ; 113(49): 14133-14138, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27911765

RESUMO

Magnetic fields from neuronal action potentials (APs) pass largely unperturbed through biological tissue, allowing magnetic measurements of AP dynamics to be performed extracellularly or even outside intact organisms. To date, however, magnetic techniques for sensing neuronal activity have either operated at the macroscale with coarse spatial and/or temporal resolution-e.g., magnetic resonance imaging methods and magnetoencephalography-or been restricted to biophysics studies of excised neurons probed with cryogenic or bulky detectors that do not provide single-neuron spatial resolution and are not scalable to functional networks or intact organisms. Here, we show that AP magnetic sensing can be realized with both single-neuron sensitivity and intact organism applicability using optically probed nitrogen-vacancy (NV) quantum defects in diamond, operated under ambient conditions and with the NV diamond sensor in close proximity (∼10 µm) to the biological sample. We demonstrate this method for excised single neurons from marine worm and squid, and then exterior to intact, optically opaque marine worms for extended periods and with no observed adverse effect on the animal. NV diamond magnetometry is noninvasive and label-free and does not cause photodamage. The method provides precise measurement of AP waveforms from individual neurons, as well as magnetic field correlates of the AP conduction velocity, and directly determines the AP propagation direction through the inherent sensitivity of NVs to the associated AP magnetic field vector.

8.
Proc Natl Acad Sci U S A ; 110(14): 5428-33, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23509252

RESUMO

Mutant human Cu/Zn superoxide dismutase 1 (SOD1) is associated with motor neuron toxicity and death in an inherited form of amyotrophic lateral sclerosis (ALS; Lou Gehrig disease). One aspect of toxicity in motor neurons involves diminished fast axonal transport, observed both in transgenic mice and, more recently, in axoplasm isolated from squid giant axons. The latter effect appears to be directly mediated by misfolded SOD1, whose addition activates phosphorylation of p38 MAPK and phosphorylation of kinesin. Here, we observe that several different oligomeric states of a fusion protein, comprising ALS-associated human G85R SOD1 joined with yellow fluorescent protein (G85R SOD1YFP), which produces ALS in transgenic mice, inhibited anterograde transport when added to squid axoplasm. Inhibition was blocked both by an apoptosis signal-regulating kinase 1 (ASK1; MAPKKK) inhibitor and by a p38 inhibitor, indicating the transport defect is mediated through the MAPK cascade. In further incubations, we observed that addition of the mammalian molecular chaperone Hsc70, abundantly associated with G85R SOD1YFP in spinal cord of transgenic mice, exerted partial correction of the transport defect, associated with diminished phosphorylation of p38. Most striking, the addition of the molecular chaperone Hsp110, in a concentration substoichiometric to the mutant SOD1 protein, completely rescued both the transport defect and the phosphorylation of p38. Hsp110 has been demonstrated to act as a nucleotide exchange factor for Hsc70 and, more recently, to be able to cooperate with it to mediate protein disaggregation. We speculate that it can cooperate with endogenous squid Hsp(c)70 to mediate binding and/or disaggregation of mutant SOD1 protein, abrogating toxicity.


Assuntos
Transporte Axonal/fisiologia , Proteínas de Choque Térmico HSP110/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Superóxido Dismutase/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Decapodiformes , Perfilação da Expressão Gênica , Proteínas de Choque Térmico HSP110/metabolismo , Humanos , Proteínas Luminescentes/metabolismo , MAP Quinase Quinase Quinase 5/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto/genética , Fosforilação/efeitos dos fármacos , Dobramento de Proteína , Proteômica , Medula Espinal/citologia , Medula Espinal/metabolismo , Superóxido Dismutase/química , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Vesículas Transportadoras/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
9.
J Neurosci ; 33(24): 10048-56, 2013 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23761900

RESUMO

Loss of function of galactosylceramidase lysosomal activity causes demyelination and vulnerability of various neuronal populations in Krabbe disease. Psychosine, a lipid-raft-associated sphingolipid that accumulates in this disease, is thought to trigger these abnormalities. Myelin-free in vitro analyses showed that psychosine inhibited fast axonal transport through the activation of axonal PP1 and GSK3ß in the axon. Abnormal levels of activated GSK3ß and abnormally phosphorylated kinesin light chains were found in nerve samples from a mouse model of Krabbe disease. Administration of GSK3ß inhibitors significantly ameliorated transport defects in vitro and in vivo in peripheral axons of the mutant mouse. This study identifies psychosine as a pathogenic sphingolipid able to block fast axonal transport and is the first to provide a molecular mechanism underlying dying-back degeneration in this genetic leukodystrophy.


Assuntos
Transporte Axonal/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Leucodistrofia de Células Globoides/patologia , Proteínas Motores Moleculares/metabolismo , Neurônios/patologia , Psicosina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicogênio Sintase Quinase 3 beta , Leucodistrofia de Células Globoides/tratamento farmacológico , Leucodistrofia de Células Globoides/genética , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Nervo Isquiático/patologia , Fatores de Tempo
10.
Hum Mol Genet ; 21(26): 5484-99, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23001566

RESUMO

Microtubules are essential components of axon guidance machinery. Among ß-tubulin mutations, only those in TUBB3 have been shown to cause primary errors in axon guidance. All identified mutations in TUBB2B result in polymicrogyria, but it remains unclear whether TUBB2B mutations can cause axon dysinnervation as a primary phenotype. We have identified a novel inherited heterozygous missense mutation in TUBB2B that results in an E421K amino acid substitution in a family who segregates congenital fibrosis of the extraocular muscles (CFEOM) with polymicrogyria. Diffusion tensor imaging of brains of affected family members reveals aberrations in the trajectories of commissural projection neurons, implying a paucity of homotopic connections. These observations led us to ask whether axon dysinnervation is a primary phenotype, and why the E421K, but not other, TUBB2B substitutions cause CFEOM. Expression of exogenous Tubb2b-E421K in developing callosal projection neurons is sufficient to perturb homotopic connectivity, without affecting neuronal production or migration. Using in vitro biochemical assays and yeast genetics, we find that TUBB2B-E421K αß-heterodimers are incorporated into the microtubule network where they alter microtubule dynamics and can reduce kinesin localization. These data provide evidence that TUBB2B mutations can cause primary axon dysinnervation. Interestingly, by incorporating into microtubules and altering their dynamic properties, the E421K substitution behaves differently than previously identified TUBB2B substitutions, providing mechanistic insight into the divergence between resulting phenotypes. Together with previous studies, these findings highlight that ß-tubulin isotypes function in both conserved and divergent ways to support proper human nervous system development.


Assuntos
Cinesinas/metabolismo , Malformações do Desenvolvimento Cortical/genética , Músculos Oculomotores/patologia , Tubulina (Proteína)/genética , Alelos , Substituição de Aminoácidos/genética , Axônios/metabolismo , Encéfalo/anormalidades , Encéfalo/metabolismo , Feminino , Fibrose , Heterozigoto , Humanos , Cinesinas/genética , Masculino , Malformações do Desenvolvimento Cortical/patologia , Microtúbulos/genética , Microtúbulos/metabolismo , Mutação de Sentido Incorreto , Neurogênese , Neurônios/metabolismo , Neurônios/fisiologia , Linhagem , Fenótipo , Ligação Proteica , Tubulina (Proteína)/metabolismo
11.
Brain Struct Funct ; 229(4): 909-918, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38483581

RESUMO

Humans display automatic action tendencies toward emotional stimuli, showing faster automatic behavior (i.e., approaching a positive stimulus and avoiding a negative stimulus) than regulated behavior (i.e., avoiding a positive stimulus and approaching a negative stimulus). Previous studies have shown that the primary motor cortex is involved in the processing of automatic actions, with higher motor evoked potential amplitudes during automatic behavior elicited by single-pulse transcranial magnetic stimulation. However, it is unknown how intracortical circuits are involved with automatic action tendencies. Here, we measured short-interval intracortical inhibition and intracortical facilitation within the primary motor cortex by using paired-pulse transcranial magnetic stimulation protocols during a manikin task, which has been widely used to explore approaching and avoiding behavior. Results showed that intracortical facilitation was stronger during automatic behavior than during regulated behavior. Moreover, there was a significant negative correlation between reaction times and intracortical facilitation effect during automatic behavior: individuals with short reaction times had stronger faciliatory activity, as shown by higher intracortical facilitation. By contrast, no significant difference was found for short-interval intracortical inhibition between automatic behavior and regulated behavior. The results indicated that the intracortical facilitation circuit, mediated by excitatory glutamatergic neurons, in the primary motor cortex, plays an important role in mediating automatic action tendencies. This finding further supports the link between emotional perception and the action system.


Assuntos
Córtex Motor , Humanos , Córtex Motor/fisiologia , Potencial Evocado Motor/fisiologia , Tempo de Reação/fisiologia , Estimulação Magnética Transcraniana/métodos , Neurônios , Inibição Neural/fisiologia , Eletromiografia/métodos
12.
Addiction ; 118(10): 1895-1907, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37400937

RESUMO

AIMS: By performing three transcranial magnetic stimulation (TMS) experiments, we measured the motor-specific modulatory mechanisms in the primary motor cortex (M1) at both the intercortical and intracortical levels when smokers actively approach or avoid smoking-related cues. DESIGN, SETTING AND PARTICIPANTS: For all experiments, the design was group (smokers versus non-smokers) × action (approach versus avoidance) × image type (neutral versus smoking-related). The study was conducted at the Shanghai University of Sport, CHN, TMS Laboratory. For experiment 1, 30 non-smokers and 30 smokers; for experiment 2, 16 non-smokers and 16 smokers; for experiment 3, 16 non-smokers and 16 smokers. MEASUREMENTS: For all experiments, the reaction times were measured using the smoking stimulus-response compatibility task. While performing the task, single-pulse TMS was applied to the M1 in experiment 1 to measure the excitability of the corticospinal pathways, and paired-pulse TMS was applied to the M1 in experiments 2 and 3 to measure the activity of intracortical facilitation (ICF) and short-interval intracortical inhibition (SICI) circuits, respectively. FINDINGS: Smokers had faster responses when approaching smoking-related cues (F1,58 = 36.660, P < 0.001, η p 2 = 0.387), accompanied by higher excitability of the corticospinal pathways (F1,58 = 10.980, P = 0.002, η p 2 = 0.159) and ICF circuits (F1,30 = 22.187, P < 0.001, η p 2 = 0.425), while stronger SICI effects were observed when they avoided these cues (F1,30 = 10.672, P = 0.003, η p 2 = 0.262). CONCLUSIONS: Smokers appear to have shorter reaction times, higher motor-evoked potentials and stronger intracortical facilitation effects when performing approach responses to smoking-related cues and longer reaction times, a lower primary motor cortex descending pathway excitability and a stronger short-interval intracortical inhibition effect when avoiding them.


Assuntos
Aprendizagem da Esquiva , Córtex Motor , Humanos , Sinais (Psicologia) , Córtex Motor/fisiologia , China , Fumar
13.
bioRxiv ; 2023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37808871

RESUMO

Expansion of a hexanucleotide repeat in a noncoding region of the C9ORF72 gene is responsible for a significant fraction of Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) cases, but identifying specific toxic gene products and mechanisms has been difficult. Pathogenesis was proposed to involve the production of toxic RNA species and/or accumulation of toxic dipeptide repeats (DPRs), but distinguishing between these mechanisms has been challenging. In this study, we first use complementary model systems for analyzing pathogenesis in adult-onset neurodegenerative diseases to characterize the pathogenicity of DPRs produced by Repeat Associated Non-ATG (RAN) translation of C9ORF72 in specific cellular compartments: isolated axoplasm and giant synapse from the squid. Results showed selective axonal and presynaptic toxicity of GP-DPRs, independent of associated RNA. These effects involved downstream ASK1 signaling pathways that affect fast axonal transport and synaptic function, a pathogenic mechanism shared with other mutant proteins associated with familial ALS, like SOD1 and FUS. These pathways are sufficient to produce the "dying-back" axonopathy seen in ALS. However, other mutant genes (e.g., SOD1) that activate this mechanism rarely produce FTD. When parallel studies in primary motor neurons from rats were conducted, an additional pathogenic mechanism was revealed. The GR- and PR-DPRs, which had no effect on axonal transport or synaptic transmission, were found to disrupt the nuclei of transfected neurons, leading to "dying-forward" neuropathy. All C9-DRP-mediated toxic effects observed here are independent of whether the corresponding mRNAs contained hexanucleotide repeats or alternative codons. These studies establish the divergent toxicity of C9-DPRs that cause neurodegeneration in ALS and FTD, suggesting that these two independent pathogenic mechanisms may contribute to disease heterogeneity and/or synergize on disease progression in C9ORF72 patients with both ALS and FTD symptoms.

14.
bioRxiv ; 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37398461

RESUMO

Selective breakdown of proteins and aggregates is crucial for maintaining normal cellular activities and is involved in the pathogenesis of diverse diseases. How the cell recognizes and tags these targets in different structural states for degradation by the proteasome and autophagy pathways has not been well understood. Here, we discovered that a HECT-family ubiquitin ligase HUWE1 is broadly required for the efficient degradation of soluble factors and for the clearance of protein aggregates/condensates. Underlying this capacity of HUWE1 is a novel Ubiquitin-Directed ubiquitin Ligase (UDL) activity which recognizes both soluble substrates and aggregates that carry a high density of ubiquitin chains and rapidly expand the ubiquitin modifications on these targets. Ubiquitin signal amplification by HUWE1 recruits the ubiquitin-dependent segregase p97/VCP to process these targets for subsequent degradation or clearance. HUWE1 controls the cytotoxicity of protein aggregates, mediates Targeted Protein Degradation and regulates cell-cycle transitions with its UDL activity.

15.
Cells ; 12(19)2023 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-37830617

RESUMO

The amyloid precursor protein (APP) is a key molecular component of Alzheimer's disease (AD) pathogenesis. Proteolytic APP processing generates various cleavage products, including extracellular amyloid beta (Aß) and the cytoplasmic APP intracellular domain (AICD). Although the role of AICD in the activation of kinase signaling pathways is well established in the context of full-length APP, little is known about intracellular effects of the AICD fragment, particularly within discrete neuronal compartments. Deficits in fast axonal transport (FAT) and axonopathy documented in AD-affected neurons prompted us to evaluate potential axon-autonomous effects of the AICD fragment for the first time. Vesicle motility assays using the isolated squid axoplasm preparation revealed inhibition of FAT by AICD. Biochemical experiments linked this effect to aberrant activation of selected axonal kinases and heightened phosphorylation of the anterograde motor protein conventional kinesin, consistent with precedents showing phosphorylation-dependent regulation of motors proteins powering FAT. Pharmacological inhibitors of these kinases alleviated the AICD inhibitory effect on FAT. Deletion experiments indicated this effect requires a sequence encompassing the NPTY motif in AICD and interacting axonal proteins containing a phosphotyrosine-binding domain. Collectively, these results provide a proof of principle for axon-specific effects of AICD, further suggesting a potential mechanistic framework linking alterations in APP processing, FAT deficits, and axonal pathology in AD.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transporte Axonal , Doença de Alzheimer/metabolismo , Axônios/metabolismo
16.
J Neurosci ; 31(27): 9858-68, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21734277

RESUMO

Aggregated filamentous forms of hyperphosphorylated tau (a microtubule-associated protein) represent pathological hallmarks of Alzheimer's disease (AD) and other tauopathies. While axonal transport dysfunction is thought to represent a primary pathogenic factor in AD and other neurodegenerative diseases, the direct molecular link between pathogenic forms of tau and deficits in axonal transport remain unclear. Recently, we demonstrated that filamentous, but not soluble, forms of wild-type tau inhibit anterograde, kinesin-based fast axonal transport (FAT) by activating axonal protein phosphatase 1 (PP1) and glycogen synthase kinase 3 (GSK3), independent of microtubule binding. Here, we demonstrate that amino acids 2-18 of tau, comprising a phosphatase-activating domain (PAD), are necessary and sufficient for activation of this pathway in axoplasms isolated from squid giant axons. Various pathogenic forms of tau displaying increased exposure of PAD inhibited anterograde FAT in squid axoplasm. Importantly, immunohistochemical studies using a novel PAD-specific monoclonal antibody in human postmortem tissue indicated that increased PAD exposure represents an early pathogenic event in AD that closely associates in time with AT8 immunoreactivity, an early marker of pathological tau. We propose a model of pathogenesis in which disease-associated changes in tau conformation lead to increased exposure of PAD, activation of PP1-GSK3, and inhibition of FAT. Results from these studies reveal a novel role for tau in modulating axonal phosphotransferases and provide a molecular basis for a toxic gain-of-function associated with pathogenic forms of tau.


Assuntos
Transporte Axonal/genética , Axônios/patologia , Encéfalo/patologia , Cinesinas/metabolismo , Fosfotransferases/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Análise de Variância , Animais , Transporte Axonal/efeitos dos fármacos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Decapodiformes , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Técnicas In Vitro , Cinesinas/genética , Modelos Biológicos , Mutagênese/genética , Fragmentos de Peptídeos/metabolismo , Isótopos de Fósforo/farmacocinética , Fosfotransferases/genética , Proteínas Proto-Oncogênicas c-jun/farmacocinética , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais/genética , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
17.
J Neurosci ; 29(41): 12776-86, 2009 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-19828789

RESUMO

Adult-onset neurodegenerative diseases (AONDs) comprise a heterogeneous group of neurological disorders characterized by a progressive, age-dependent decline in neuronal function and loss of selected neuronal populations. Alterations in synaptic function and axonal connectivity represent early and critical pathogenic events in AONDs, but molecular mechanisms underlying these defects remain elusive. The large size and complex subcellular architecture of neurons render them uniquely vulnerable to alterations in axonal transport (AT). Accordingly, deficits in AT have been documented in most AONDs, suggesting a common defect acquired through different pathogenic pathways. These observations suggest that many AONDs can be categorized as dysferopathies, diseases in which alterations in AT represent a critical component in pathogenesis. Topics here address various molecular mechanisms underlying alterations in AT in several AONDs. Illumination of such mechanisms provides a framework for the development of novel therapeutic strategies aimed to prevent axonal and synaptic dysfunction in several major AONDs.


Assuntos
Transporte Axonal/fisiologia , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Doenças Neurodegenerativas/genética , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Proteínas tau/metabolismo
18.
Virol J ; 7: 129, 2010 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-20546601

RESUMO

Recombination of Hepatitis E Virus (HEV) has rarely been reported. In the present study, phylogenetic and recombination analyses were performed on 134 complete HEV genomes. Three potentially significant recombination events, including both intra-genotype and one inter-genotype, were identified by recombination detection analysis. Recombination events I and II occurred intra-genotype and inter-genotype, respectively, among three isolates, including the lineage represented by CHN-XJ-SW13 (GU119961, swine isolate), E067-SIJ05C (AB369690, human isolate), and JJT-Kan (AB091394, human isolate), and lead to the recombinant swine isolate swCH31 (DQ450072). Recombination event III occurred between the lineage represented by the NA1 (M73218) and K52-87 (L25595), which resulted in the recombinant Xingjiang-1 (D11092). Our analyses proved that that recombination could occur between human and swine HEV strains, double recombination events existed in HEV, and recombination event could happen within ORF2 region of HEV. These results will provide valuable hints for future research on HEV diversity.


Assuntos
Vírus da Hepatite E/genética , Hepatite E/veterinária , Hepatite E/virologia , Recombinação Genética , Doenças dos Suínos/virologia , Animais , Genoma Viral , Vírus da Hepatite E/classificação , Vírus da Hepatite E/isolamento & purificação , Humanos , Dados de Sequência Molecular , Filogenia , Suínos
19.
eNeuro ; 7(2)2020.
Artigo em Inglês | MEDLINE | ID: mdl-32188708

RESUMO

Altered synaptic function is thought to play a role in many neurodegenerative diseases, but little is known about the underlying mechanisms for synaptic dysfunction. The squid giant synapse (SGS) is a classical model for studying synaptic electrophysiology and ultrastructure, as well as molecular mechanisms of neurotransmission. Here, we conduct a multidisciplinary study of synaptic actions of misfolded human G85R-SOD1 causing familial amyotrophic lateral sclerosis (ALS). G85R-SOD1, but not WT-SOD1, inhibited synaptic transmission, altered presynaptic ultrastructure, and reduced both the size of the readily releasable pool (RRP) of synaptic vesicles and mobility from the reserved pool (RP) to the RRP. Unexpectedly, intermittent high-frequency stimulation (iHFS) blocked inhibitory effects of G85R-SOD1 on synaptic transmission, suggesting aberrant Ca2+ signaling may underlie G85R-SOD1 toxicity. Ratiometric Ca2+ imaging showed significantly increased presynaptic Ca2+ induced by G85R-SOD1 that preceded synaptic dysfunction. Chelating Ca2+ using EGTA prevented synaptic inhibition by G85R-SOD1, confirming the role of aberrant Ca2+ in mediating G85R-SOD1 toxicity. These results extended earlier findings in mammalian motor neurons and advanced our understanding by providing possible molecular mechanisms and therapeutic targets for synaptic dysfunctions in ALS as well as a unique model for further studies.


Assuntos
Esclerose Lateral Amiotrófica , Esclerose Lateral Amiotrófica/genética , Animais , Decapodiformes , Humanos , Camundongos , Camundongos Transgênicos , Superóxido Dismutase/genética , Superóxido Dismutase-1/genética , Sinapses
20.
Sci Data ; 6: 190016, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30778261

RESUMO

The immortalized human ReNcell VM cell line represents a reproducible and easy-to-propagate cell culture system for studying the differentiation of neural progenitors. To better characterize the starting line and its subsequent differentiation, we assessed protein and phospho-protein levels and cell morphology over a 15-day period during which ReNcell progenitors differentiated into neurons, astrocytes and oligodendrocytes. Five of the resulting datasets measured protein levels or states of phosphorylation based on tandem-mass-tag (TMT) mass spectrometry and four datasets characterized cellular phenotypes using high-content microscopy. Proteomic analysis revealed reproducible changes in pathways responsible for cytoskeletal rearrangement, cell phase transitions, neuronal migration, glial differentiation, neurotrophic signalling and extracellular matrix regulation. Proteomic and imaging data revealed accelerated differentiation in cells treated with the poly-selective CDK and GSK3 inhibitor kenpaullone or the HMG-CoA reductase inhibitor mevastatin, both of which have previously been reported to promote neural differentiation. These data provide in-depth information on the ReNcell progenitor state and on neural differentiation in the presence and absence of drugs, setting the stage for functional studies.


Assuntos
Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Proteoma/análise , Linhagem Celular , Movimento Celular , Humanos , Neurogênese/fisiologia , Neurônios/citologia , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA