Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 180: 106075, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36914075

RESUMEN

Prion diseases are pathogenically linked to the normal cellular prion protein (PrPC) misfolding into abnormal conformers (PrPSc), with PrPSc accumulation underpinning both transmission and neurotoxicity. Despite achieving this canonical understanding, however fundamental questions remain incompletely resolved, including the level of pathophysiological overlap between neurotoxic and transmitting species of PrPSc and the temporal profiles of their propagation. To further investigate the likely time of occurrence of significant levels of neurotoxic species during prion disease development, the well characterised in vivo M1000 murine model was employed. Following intracerebral inoculation, detailed serial cognitive and ethological testing at specified time points suggested subtle transition to early symptomatic disease from ∼50% of the overall disease course. In addition to observing a chronological order for impaired behaviours, different behavioural tests also showed distinctive profiles of evolving cognitive impairments with the Barnes maze demonstrating a relatively simple linear worsening of spatial learning and memory over an extended period while in contrast a conditioned fear memory paradigm previously untested in murine prion disease demonstrated more complex alterations during disease progression. These observations support the likely production of neurotoxic PrPSc from at least just prior to the mid-point of murine M1000 prion disease and illustrate the likely need to tailor the types of behavioural testing across the time course of disease progression for optimal detection of cognitive deficits.


Asunto(s)
Disfunción Cognitiva , Enfermedades por Prión , Animales , Ratones , Escala de Evaluación de la Conducta , Enfermedades por Prión/metabolismo , Progresión de la Enfermedad , Cognición
2.
Eur J Neurosci ; 56(9): 5342-5367, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35768153

RESUMEN

Repetitive mild traumatic brain injury, commonly experienced following sports injuries, results in various secondary injury processes and is increasingly recognised as a risk factor for the development of neurodegenerative conditions such as chronic traumatic encephalopathy, which is characterised by tau pathology. We aimed to characterise the underlying pathological mechanisms that might contribute to the onset of neurodegeneration and behavioural changes in the less-explored subacute (1-month) period following single or repetitive controlled cortical impact injury (five impacts, 48 h apart) in 12-week-old male and female C57Bl6 mice. We conducted motor and cognitive testing, extensively characterised the status of tau and its regulatory proteins via western blot and quantified neuronal populations using stereology. We report that r-mTBI resulted in neurobehavioural deficits, gait impairments and anxiety-like behaviour at 1 month post-injury, effects not seen following a single injury. R-mTBI caused a significant increase in amyloid precursor protein, an increased trend towards tau phosphorylation and significant changes in kinase/phosphatase proteins that may promote a downstream increase in tau phosphorylation, but no changes in synaptic or neuroinflammatory markers. Lastly, we report neuronal loss in various brain regions following both single and repeat injuries. We demonstrate herein that repeated impacts are required to promote the initiation of a cascade of biochemical events that are consistent with the onset of neurodegeneration subacutely post-injury. Identifying the timeframe in which these changes occur and the pathological mechanisms involved will be crucial for the development of future therapeutics to prevent the onset or mitigate the progression of neurodegeneration following r-mTBI.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Animales , Masculino , Femenino , Ratones , Proteínas tau/metabolismo , Precursor de Proteína beta-Amiloide , Conmoción Encefálica/complicaciones , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Trastornos de la Memoria , Factores de Transcripción , Lesiones Traumáticas del Encéfalo/complicaciones
3.
J Biol Chem ; 295(51): 17497-17513, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33453994

RESUMEN

Neurodegeneration in Parkinson's disease (PD) can be recapitulated in animals by administration of α-synuclein preformed fibrils (PFFs) into the brain. However, the mechanism by which these PFFs induce toxicity is unknown. Iron is implicated in PD pathophysiology, so we investigated whether α-synuclein PFFs induce ferroptosis, an iron-dependent cell death pathway. A range of ferroptosis inhibitors were added to a striatal neuron-derived cell line (STHdhQ7/7 cells), a dopaminergic neuron-derived cell line (SN4741 cells), and WT primary cortical neurons, all of which had been intoxicated with α-synuclein PFFs. Viability was not recovered by these inhibitors except for liproxstatin-1, a best-in-class ferroptosis inhibitor, when used at high doses. High-dose liproxstatin-1 visibly enlarged the area of a cell that contained acidic vesicles and elevated the expression of several proteins associated with the autophagy-lysosomal pathway similarly to the known lysosomal inhibitors, chloroquine and bafilomycin A1. Consistent with high-dose liproxstatin-1 protecting via a lysosomal mechanism, we further de-monstrated that loss of viability induced by α-synuclein PFFs was attenuated by chloroquine and bafilomycin A1 as well as the lysosomal cysteine protease inhibitors, leupeptin, E-64D, and Ca-074-Me, but not other autophagy or lysosomal enzyme inhibitors. We confirmed using immunofluorescence microscopy that heparin prevented uptake of α-synuclein PFFs into cells but that chloroquine did not stop α-synuclein uptake into lysosomes despite impairing lysosomal function and inhibiting α-synuclein toxicity. Together, these data suggested that α-synuclein PFFs are toxic in functional lysosomes in vitro. Therapeutic strategies that prevent α-synuclein fibril uptake into lysosomes may be of benefit in PD.


Asunto(s)
Lisosomas/metabolismo , alfa-Sinucleína/toxicidad , Animales , Células Cultivadas , Neuronas Dopaminérgicas/metabolismo , Endosomas/metabolismo , Ferroptosis/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
4.
Mol Psychiatry ; 25(11): 2728-2741, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32518388

RESUMEN

Aging and female sex are the major risk factors for Alzheimer's disease and its associated brain amyloid-ß (Aß) neuropathology, but the mechanisms mediating these risk factors remain uncertain. Evidence indicates that Aß aggregation by Zn2+ released from glutamatergic neurons contributes to amyloid neuropathology, so we tested whether aging and sex adversely influences this neurophysiology. Using acute hippocampal slices, we found that extracellular Zn2+-elevation induced by high K+ stimulation was significantly greater with older (65 weeks vs 10 weeks old) rats, and was exaggerated in females. This was driven by slower reuptake of extracellular Zn2+, which could be recapitulated by mitochondrial intoxication. Zn2+:Aß aggregates were toxic to the slices, but Aß alone was not. Accordingly, high K+ caused synthetic human Aß added to the slices to form soluble oligomers as detected by bis-ANS, attaching to neurons and inducing toxicity, with older slices being more vulnerable. Age-dependent energy failure impairing Zn2+ reuptake, and a higher maximal capacity for Zn2+ release by females, could contribute to age and sex being major risk factors for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Espacio Extracelular/metabolismo , Hipocampo/metabolismo , Agregación Patológica de Proteínas/metabolismo , Zinc/metabolismo , Animales , Femenino , Masculino , Ratas , Ratas Wistar , Factores de Riesgo
5.
Biophys J ; 119(1): 128-141, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32562618

RESUMEN

Prion diseases are neurodegenerative disorders pathogenically linked to cellular prion protein (PrPC) misfolding into abnormal conformers (PrPSc), with PrPSc underpinning both transmission and synaptotoxicity. Although the biophysical features of PrPSc required to induce acute synaptic dysfunction remain incompletely defined, we recently reported that acutely synaptotoxic PrPSc appeared to be oligomeric. We herein provide further insights into the kinetic and requisite biophysical characteristics of acutely synaptotoxic ex vivo PrPSc derived from the brains of mice dying from M1000 prion disease. Pooled fractions of M1000 PrPSc located within the molecular weight range approximating monomeric PrP (mM1000) generated through size exclusion chromatography were found to harbor acute synaptotoxicity equivalent to preformed oligomeric fractions (oM1000). Subsequent investigation showed mM1000 corresponded to PrPSc rapidly concatenating in physiological buffer to exist as predominantly, closely associated, small oligomers. The oligomerization of PrP in mM1000 could be substantially mitigated by treatment with the antiaggregation compound epigallocatechin gallate, thereby maintaining the PrPSc as primarily nonoligomeric with completely abrogated acute synaptotoxicity; moreover, despite epigallocatechin gallate treatment, pooled oM1000 remained oligomeric and acutely synaptotoxic. A similar tendency to rapid formation of oligomers was observed for PrPC when monomeric fractions derived from size exclusion chromatography of normal brain homogenates (mNBH) were pooled, but neither mNBH nor preformed higher-order NBH complexes (oNBH) were acutely synaptotoxic. Oligomers formed from mNBH could be reduced to mainly monomers (<100 kDa) after enzymatic digestion of nucleic acids, whereas higher-order PrP assemblies derived from pooled mM1000, oM1000, and oNBH resisted such treatment. Collectively, these findings support that oligomerization of PrPSc into small multimeric assemblies appears to be a critical biophysical feature for engendering inherent acute synaptotoxicity, with preformed oligomers found in oM1000 appearing to be stable, tightly self-associated ensembles that coexist in dynamic equilibrium with mM1000, with the latter appearing capable of rapid aggregation, albeit initially forming smaller, weakly self-associated, acutely synaptotoxic oligomers.


Asunto(s)
Proteínas PrPC , Enfermedades por Prión , Priones , Animales , Encéfalo/metabolismo , Ratones
6.
Subcell Biochem ; 91: 107-122, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30888651

RESUMEN

With an increasingly ageing population that is expected to double by 2050 in the U.S., it is paramount that we further understand the neurological changes that occur during ageing. This is relevant not only in the context of "pathological" ageing, where the development of many neurodegenerative disorders is typically a feature of only the older population (and indeed, age is the primary risk factor for many conditions such as Alzheimer's disease), but also for what is considered to be "normal" or "healthy" ageing. Specifically, a significant proportion of the older population are affected by "age-related cognitive decline" (ARCD), which is both independent of dementia and has an incidence 70% higher than dementia alone. However, whilst it is reported that there are pathogenic and phenotypic overlaps between healthy and pathological ageing, it is clear that there is a need to identify the pathways and understand the mechanisms that contribute to this loss of cognitive function with normal ageing, particularly in light of the increasing life expectancy of the global population. Importantly, there is an increasing body of evidence implicating zinc homeostasis as a key player in learning and memory and also potentially ARCD. Further research will ultimately contribute to the development of targeted therapeutics that will promote successful brain ageing. In this chapter we will explore the notion of ARCD, with a perspective on potential key neurochemical pathways that can be targeted for future intervention.


Asunto(s)
Envejecimiento/fisiología , Envejecimiento/psicología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/psicología , Cognición/fisiología , Envejecimiento/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Enfermedad de Alzheimer/terapia , Trastornos del Conocimiento/terapia , Humanos , Zinc/metabolismo
7.
Int J Mol Sci ; 21(3)2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-32012946

RESUMEN

Metals are critical cellular elements that are involved in a variety of cellular processes, with recent literature demonstrating that zinc, and the synaptic zinc transporter (ZnT3), are specifically involved in learning and memory and may also be key players in age-related neurodegenerative disorders such as Alzheimer's disease. Whilst the cellular content and location of metals is critical, recent data has demonstrated that the metalation state of proteins is a determinant of protein function and potential toxicity. As we have previously reported that ZnT3 knockout (KO) mice have deficits in total zinc levels at both 3 and 6 months of age, we were interested in whether there might be changes in the metalloproteomic profile in these animals. To do this, we utilised size exclusion chromatography-inductively coupled plasma mass spectrometry (SEC-ICP-MS) and examined hippocampal homogenates from ZnT3 KO and age-matched wild-type mice at 3, 6 and 18 months of age. Our data suggest that there are alterations in specific metal binding proteins, for zinc, copper and iron all being modulated in the ZnT3 KO mice compared to wild-type (WT). These data suggest that ZnT3 KO mice may have impairments in the levels or localisation of multiple transition metals, and that copper- and iron-dependent cellular pathways may also be impacted in these mice.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Transporte de Catión/genética , Metaloproteínas/metabolismo , Proteómica/métodos , Envejecimiento/genética , Animales , Cromatografía en Gel , Cobre/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Hipocampo/metabolismo , Hierro/metabolismo , Masculino , Espectrometría de Masas , Ratones , Ratones Noqueados , Zinc/metabolismo
8.
J Neurochem ; 150(1): 88-106, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30716176

RESUMEN

Treatment with the dopamine (DA) precursor l-3,4-dihydroxyphenylalanine (l-DOPA) provides symptomatic relief arising from DA denervation in Parkinson's disease. Mounting evidence that DA autooxidation to neurotoxic quinones is involved in Parkinson's disease pathogenesis has raised concern about potentiation of oxidative stress by l-DOPA. The rate of DA quinone formation increases in the presence of excess redox-active iron (Fe), which is a pathological hallmark of Parkinson's disease. Conversely, l-DOPA has pH-dependent Fe-chelating properties, and may act to 'redox silence' Fe and partially allay DA autoxidation. We examined the effects of l-DOPA in three murine models of parkinsonian neurodegeneration: early-life Fe overexposure in wild-type mice, transgenic human (h)A53T mutant α-synuclein (α-syn) over-expression, and a combined 'multi-hit' model of Fe-overload in hA53T mice. We found that l-DOPA was neuroprotective and prevented age-related Fe accumulation in the substantia nigra pars compacta (SNc), similar to the mild-affinity Fe chelator clioquinol. Chronic l-DOPA treatment showed no evidence of increased oxidative stress in wild-type midbrain and normalized motor performance, when excess Fe was present. Similarly, l-DOPA also did not exacerbate protein oxidation levels in hA53T mice, with or without excess nigral Fe, and showed evidence of neuroprotection. The effects of l-DOPA in Fe-fed hA53T mice were somewhat muted, suggesting that Fe-chelation alone is insufficient to attenuate neuron loss in an animal model also recapitulating altered DA metabolism. In summary, we found no evidence in any of our model systems that l-DOPA treatment accentuated neurodegeneration, suggesting DA replacement therapy does not contribute to oxidative stress in the Parkinson's disease brain.


Asunto(s)
Antiparkinsonianos/farmacología , Encéfalo/efectos de los fármacos , Levodopa/farmacología , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Humanos , Hierro/metabolismo , Sobrecarga de Hierro , Ratones , Ratones Transgénicos , Degeneración Nerviosa/patología , alfa-Sinucleína
9.
Biochem Biophys Res Commun ; 514(3): 888-892, 2019 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-31084925

RESUMEN

Human amyloid-ß1-40 (Aß1-40) and rat Aß1-42 have lower affinity for extracellular Zn2+ than human Aß1-42. Here we report extracellular Zn2+-independent attenuation of dentate gyrus long-term potentiation (LTP) by human Aß1-40 and rat Aß1-42. On the basis of the data that dentate gyrus LTP is extracellular Zn2+-dependently attenuated after local injection of human Aß1-42 (25 pmol, 1 µl) into the dentate gyrus, which increases intracellular Zn2+ in the dentate gyrus, the toxicity of human Aß1-40 and rat Aß1-42 was compared in the in vivo system with human Aß1-42. Dentate gyrus LTP was attenuated after injection of human Aß1-40 and rat Aß1-42 (25 pmol, 1 µl) into the dentate gyrus, which did not increase intracellular Zn2+ in the dentate gyrus. The attenuated LTP was not rescued by co-injection of CaEDTA, an extracellular Zn2+ chelator. The present study suggests that human Aß1-40 and rat Aß1-42 affect cognitive activity via extracellular Zn2+-independent mechanism at low micromolar concentration.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Espacio Extracelular/metabolismo , Potenciación a Largo Plazo , Fragmentos de Péptidos/metabolismo , Zinc/farmacología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Animales , Ácido Edético/farmacología , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Fragmentos de Péptidos/química , Ratas Wistar
10.
J Neurosci ; 37(30): 7253-7262, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28652412

RESUMEN

Brain Aß1-42 accumulation is considered an upstream event in pathogenesis of Alzheimer's disease. However, accumulating evidence indicates that other neurochemical changes potentiate the toxicity of this constitutively generated peptide. Here we report that the interaction of Aß1-42 with extracellular Zn2+ is essential for in vivo rapid uptake of Aß1-42 and Zn2+ into dentate granule cells in the normal rat hippocampus. The uptake of both Aß1-42 and Zn2+ was blocked by CaEDTA, an extracellular Zn2+ chelator, and by Cd2+, a metal that displaces Zn2+ for Aß1-42 binding. In vivo perforant pathway LTP was unaffected by perfusion with 1000 nm Aß1-42 in ACSF without Zn2+ However, LTP was attenuated under preperfusion with 5 nm Aß1-42 in ACSF containing 10 nm Zn2+, recapitulating the concentration of extracellular Zn2+, but not with 5 nm Aß1-40 in ACSF containing 10 nm Zn2+ Aß1-40 and Zn2+ were not taken up into dentate granule cells under these conditions, consistent with lower affinity of Aß1-40 for Zn2+ than Aß1-42 Aß1-42-induced attenuation of LTP was rescued by both CaEDTA and CdCl2, and was observed even with 500 pm Aß1-42 Aß1-42 injected into the dentate granule cell layer of rats induced a rapid memory disturbance that was also rescued by coinjection of CdCl2 The present study supports blocking the formation of Zn-Aß1-42 in the extracellular compartment as an effective preventive strategy for Alzheimer's disease.SIGNIFICANCE STATEMENT Short-term memory loss occurs in normal elderly and increases in the predementia stage of Alzheimer's disease (AD). Amyloid-ß1-42 (Aß1-42), a possible causing peptide in AD, is bound to Zn2+ in the extracellular compartment in the hippocampus induced short-term memory loss in the normal rat brain, suggesting that extracellular Zn2+ is essential for Aß1-42-induced short-term memory loss. The evidence is important to find an effective preventive strategy for AD, which is blocking the formation of Zn-Aß1-42 in the extracellular compartment.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/metabolismo , Zinc/metabolismo , Animales , Disfunción Cognitiva/complicaciones , Progresión de la Enfermedad , Líquido Extracelular/metabolismo , Masculino , Trastornos de la Memoria/complicaciones , Unión Proteica , Ratas , Ratas Wistar
11.
Epilepsia ; 58(9): e136-e141, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28710841

RESUMEN

The biologic processes underlying epileptogenesis following a brain insult are not fully understood, but several lines of evidence suggest that hyperphosphorylation of tau may be an important factor in these processes. To provide further insight into the causal relationship between tau and epileptogenesis, this study applied amygdala kindling to rTg4510 mice that, concurrent with other pathologies, overexpress phosphorylated tau, tau knockout mice, or their respective wild-type controls. Mice were electrically stimulated twice daily, 5 days per week for 3 weeks. Electroencephalography was recorded to measure the primary afterdischarge duration, and the behavioral progression of kindling-induced seizures was assessed. rTg4510 mice (n = 10) had increased primary afterdischarge durations (p < 0.001), and significantly more rapid progression of kindling (p < 0.001), compared with wild-type mice (n = 10). Tau knockout mice (n = 7), however, did not differ from their wild-type counterparts (n = 8) on any of the seizure outcomes. These results suggest that Tg4510 mice are more vulnerable to epileptogenesis, but that the presence of tau itself is not necessary for kindling epileptogenesis to occur.


Asunto(s)
Excitación Neurológica/metabolismo , Proteínas tau/fisiología , Amígdala del Cerebelo/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Proteínas tau/metabolismo
12.
Int J Mol Sci ; 18(12)2017 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-29168792

RESUMEN

The divalent cation zinc is an integral requirement for optimal cellular processes, whereby it contributes to the function of over 300 enzymes, regulates intracellular signal transduction, and contributes to efficient synaptic transmission in the central nervous system. Given the critical role of zinc in a breadth of cellular processes, its cellular distribution and local tissue level concentrations remain tightly regulated via a series of proteins, primarily including zinc transporter and zinc import proteins. A loss of function of these regulatory pathways, or dietary alterations that result in a change in zinc homeostasis in the brain, can all lead to a myriad of pathological conditions with both acute and chronic effects on function. This review aims to highlight the role of zinc signaling in the central nervous system, where it may precipitate or potentiate diverse issues such as age-related cognitive decline, depression, Alzheimer's disease or negative outcomes following brain injury.


Asunto(s)
Encefalopatías/etiología , Encefalopatías/metabolismo , Transducción de Señal , Zinc/metabolismo , Animales , Transporte Biológico , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Encefalopatías/fisiopatología , Proteínas Portadoras/metabolismo , Homeostasis , Humanos , Hierro/metabolismo , Metalotioneína/metabolismo
13.
J Neurosci ; 35(8): 3591-7, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716857

RESUMEN

Elevation of both neuronal iron and nitric oxide (NO) in the substantia nigra are associated with Parkinson's disease (PD) pathogenesis. We reported previously that the Alzheimer-associated ß-amyloid precursor protein (APP) facilitates neuronal iron export. Here we report markedly decreased APP expression in dopaminergic neurons of human PD nigra and that APP(-/-) mice develop iron-dependent nigral cell loss. Conversely, APP-overexpressing mice are protected in the MPTP PD model. NO suppresses APP translation in mouse MPTP models, explaining how elevated NO causes iron-dependent neurodegeneration in PD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Hierro/metabolismo , Óxido Nítrico/metabolismo , Enfermedad de Parkinson/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Tumoral , Neuronas Dopaminérgicas/metabolismo , Femenino , Humanos , Intoxicación por MPTP/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Sustancia Negra/metabolismo , Sustancia Negra/patología
14.
J Neurosci ; 35(7): 2871-84, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25698727

RESUMEN

The extracellular accumulation of amyloid ß (Aß) peptides is characteristic of Alzheimer's disease (AD). However, formation of diffusible, oligomeric forms of Aß, both on and off pathways to amyloid fibrils, is thought to include neurotoxic species responsible for synaptic loss and neurodegeneration, rather than polymeric amyloid aggregates. The 8-hydroxyquinolines (8-HQ) clioquinol (CQ) and PBT2 were developed for their ability to inhibit metal-mediated generation of reactive oxygen species from Aß:Cu complexes and have both undergone preclinical and Phase II clinical development for the treatment of AD. Their respective modes of action are not fully understood and may include both inhibition of Aß fibrillar polymerization and direct depolymerization of existing Aß fibrils. In the present study, we find that CQ and PBT2 can interact directly with Aß and affect its propensity to aggregate. Using a combination of biophysical techniques, we demonstrate that, in the presence of these 8-HQs and in the absence of metal ions, Aß associates with two 8-HQ molecules and forms a dimer. Furthermore, 8-HQ bind Aß with an affinity of 1-10 µm and suppress the formation of large (>30 kDa) oligomers. The stabilized low molecular weight species are nontoxic. Treatment with 8-HQs also reduces the levels of in vivo soluble oligomers in a Caenorhabditis elegans model of Aß toxicity. We propose that 8-HQs possess an additional mechanism of action that neutralizes neurotoxic Aß oligomer formation through stabilization of small (dimeric) nontoxic Aß conformers.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Hidroxiquinolinas/metabolismo , Fragmentos de Péptidos/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/ultraestructura , Animales , Benzotiazoles , Biofisica , Caenorhabditis elegans , Células Cultivadas , Corteza Cerebral/citología , Cromatografía en Gel , Clioquinol/análogos & derivados , Clioquinol/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Microscopía Electrónica , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/ultraestructura , Unión Proteica/efectos de los fármacos , Dispersión del Ángulo Pequeño , Tiazoles/metabolismo
15.
Biochim Biophys Acta ; 1840(12): 3299-310, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25152357

RESUMEN

BACKGROUND: Iron oxidation is thought to be predominantly handled enzymatically in the body, to minimize spontaneous combustion with oxygen and to facilitate cellular iron export by loading transferrin. This process may be impaired in disease, and requires more accurate analytical assays to interrogate enzymatic- and auto-oxidation within a physiologically relevant environment. METHOD: A new triplex ferroxidase activity assay has been developed that overcomes the previous assay limitations of measuring iron oxidation at a physiologically relevant pH and salinity. RESULTS: Revised enzymatic kinetics for ceruloplasmin (Vmax≈35µMFe(3+)/min/µM; Km≈15µM) are provided under physiological conditions, and inhibition by sodium azide (Ki for Ferric Gain 78.3µM, Ki for transferrin loading 8.1×10(4)µM) is quantified. We also used this assay to characterize the non-enzymatic oxidation of iron that proceeded linearly under physiological conditions. CONCLUSIONS AND GENERAL SIGNIFICANCE: These findings indicate that the requirement of an enzyme to oxidize iron may only be necessary under conditions of adverse pH or anionic strength, for example from hypoxia. In a normal physiological environment, Fe(3+) incorporation into transferrin would be sufficiently enabled by the biological polyanions that are prevalent within extracellular fluids.

16.
Neurobiol Dis ; 81: 168-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25796563

RESUMEN

Iron accumulation and tau protein deposition are pathological features of Alzheimer's (AD) and Parkinson's diseases (PD). Soluble tau protein is lower in affected regions of these diseases, and we previously reported that tau knockout mice display motor and cognitive behavioral abnormities, brain atrophy, neuronal death in substantia nigra, and iron accumulation in the brain that all emerged between 6 and 12 months of age. This argues for a loss of tau function in AD and PD. We also showed that treatment with the moderate iron chelator, clioquinol (CQ) restored iron levels and prevented neuronal atrophy and attendant behavioral decline in 12-month old tau KO mice when commenced prior to the onset of deterioration (6 months). However, therapies for AD and PD will need to treat the disease once it is already manifest. So, in the current study, we tested whether CQ could also rescue the phenotype of mice with a developed phenotype. We found that 5-month treatment of symptomatic (13 months old) tau KO mice with CQ increased nigral tyrosine hydroxylase phosphorylation (which induces activity) and reversed the motor deficits. Treatment also reversed cognitive deficits and raised BDNF levels in the hippocampus, which was accompanied by attenuated brain atrophy, and reduced iron content in the brain. These data raise the possibility that lowering brain iron levels in symptomatic patients could reverse neuronal atrophy and improve brain function, possibly by elevating neurotrophins.


Asunto(s)
Clioquinol/uso terapéutico , Demencia/tratamiento farmacológico , Demencia/genética , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/genética , Proteínas tau/deficiencia , Ácido 3,4-Dihidroxifenilacético/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Demencia/patología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Metales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Parkinsonianos/patología , Prueba de Desempeño de Rotación con Aceleración Constante , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/metabolismo , Proteínas tau/genética
17.
Neurobiol Dis ; 81: 38-48, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26117305

RESUMEN

The pathological role of zinc in Alzheimer's disease (AD) is not yet fully elucidated, but there is strong evidence that zinc homeostasis is impaired in the AD brain and that this contributes to disease pathogenesis. In this study we examined the effects of zinc on the proteolysis of synthetic Apolipoprotein E (ApoE), a protein whose allelic variants differentially contribute to the onset/progression of disease. We have demonstrated that zinc promotes the proteolysis (using plasma kallikrein, thrombin and chymotrypsin) of synthetic ApoE in an isoform-specific way (E4>E2 and E3), resulting in more ApoE fragments, particularly for ApoE4. In the absence of exogenous proteases there was no effect of metal modulation on either lipidated or non-lipidated ApoE isoforms. Thus, increased zinc in the complex milieu of the ageing and AD brain could reduce the level of normal full-length ApoE and increase other forms that are involved in neurodegeneration. We further examined human plasma samples from people with different ApoE genotypes. Consistent with previous studies, plasma ApoE levels varied according to different genotypes, with ApoE2 carriers showing the highest total ApoE levels and ApoE4 carriers the lowest. The levels of plasma ApoE were not affected by either the addition of exogenous metals (copper, zinc or iron) or by chelation. Taken together, our study reveals that zinc may contribute to the pathogenesis of AD by affecting the proteolysis of ApoE, which to some extent explains why APOE4 carriers are more susceptible to AD.


Asunto(s)
Apolipoproteínas E/metabolismo , Plasma/efectos de los fármacos , Oligoelementos/farmacología , Zinc/farmacología , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Quelantes/farmacología , Relación Dosis-Respuesta a Droga , Ácido Edético/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Serina Proteasas/metabolismo , Trombina/farmacología , Oligoelementos/metabolismo , Zinc/metabolismo
18.
Neurobiol Dis ; 81: 196-202, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25549871

RESUMEN

Zinc transporter-3 (ZnT3) protein is responsible for loading zinc into presynaptic vesicles and consequently controls the availability of zinc at the glutamatergic synapse. ZnT3 has been shown to decline with age and in Alzheimer's disease (AD) and is crucially involved in learning and memory. In this study, we utilised whole animal behavioural analyses in the ZnT3 KO mouse line, together with electrophysiological analysis of long-term potentiation in brain slices from ZnT3 KO mice, to show that metal chaperones (clioquinol, 30 mg/kg/day for 6weeks) can prevent the age-dependent cognitive phenotype that characterises these animals. This likely occurs as a result of a homeostatic restoration of synaptic protein expression, as clioquinol significantly restored levels of various pre- and postsynaptic proteins that are critical for normal cognition, including PSD-95; AMPAR and NMDAR2b. We hypothesised that this clioquinol-mediated restoration of synaptic health resulted from a selective increase in synaptic zinc content within the hippocampus. While we demonstrated a small regional increase in hippocampal zinc content using synchrotron x-ray fluorescence microscopy, further sub-region analyses are required to determine whether this effect is seen in other regions of the hippocampal formation that are more closely linked to the synaptic plasticity effects observed in this study. These data support our recent report on the use of a different metal chaperone (PBT2) to prevent normal age-related cognitive decline and demonstrate that metal chaperones are efficacious in preventing the zinc-mediated cognitive decline that characterises ageing and disease.


Asunto(s)
Clioquinol/análogos & derivados , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Zinc/metabolismo , Análisis de Varianza , Animales , Proteínas Portadoras/genética , Proteínas de Transporte de Catión , Clioquinol/administración & dosificación , Clioquinol/uso terapéutico , Trastornos del Conocimiento/genética , Modelos Animales de Enfermedad , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/fisiología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/citología , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp
19.
Ann Neurol ; 73(4): 554-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23424051

RESUMEN

Ceruloplasmin is an iron-export ferroxidase that is abundant in plasma and also expressed in glia. We found a ∼80% loss of ceruloplasmin ferroxidase activity in the substantia nigra of idiopathic Parkinson disease (PD) cases, which could contribute to the pro-oxidant iron accumulation that characterizes the pathology. Consistent with a role for ceruloplasmin in PD etiopathogenesis, ceruloplasmin knockout mice developed parkinsonism that was rescued by iron chelation. Additionally, peripheral infusion of ceruloplasmin attenuated neurodegeneration and nigral iron elevation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model for PD. These findings show, in principle, that intravenous ceruloplasmin may have therapeutic potential in PD.


Asunto(s)
Ceruloplasmina/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Anciano , Animales , Estudios de Casos y Controles , Ceruloplasmina/deficiencia , Ceruloplasmina/uso terapéutico , Deferiprona , Modelos Animales de Enfermedad , Dopaminérgicos , Femenino , Humanos , Quelantes del Hierro/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Piridonas/uso terapéutico , Sustancia Negra/metabolismo
20.
Cell Mol Life Sci ; 69(21): 3665-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22766972

RESUMEN

Alzheimer's disease (AD) is by far the most commonly diagnosed dementia, and despite multiple efforts, there are still no effective drugs available for its treatment. One strategy that deserves to be pursued is to alter the expression and/or physiological action of endogenous proteins instead of administering exogenous factors. In this study, we intend to characterize the roles of the antioxidant, anti-inflammatory, and heavy-metal binding proteins, metallothionein-1 + 2 (MT1 + 2), in a mouse model of Alzheimer's disease, Tg2576 mice. Contrary to expectations, MT1 + 2-deficiency rescued partially the human amyloid precursor protein-induced changes in mortality and body weight in a gender-dependent manner. On the other hand, amyloid plaque burden was decreased in the cortex and hippocampus in both sexes, while the amyloid cascade, neuroinflammation, and behavior were affected in the absence of MT1 + 2 in a complex manner. These results highlight that the control of the endogenous production and/or action of MT1 + 2 could represent a powerful therapeutic target in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Metalotioneína/metabolismo , Enfermedad de Alzheimer/mortalidad , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Conducta Animal , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Metalotioneína/deficiencia , Metalotioneína/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA