Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 291(12): 6134-45, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26697885

RESUMEN

Pyroglutamate-modified amyloid-ß (pE-Aß) is a highly neurotoxic amyloid-ß (Aß) isoform and is enriched in the brains of individuals with Alzheimer disease compared with healthy aged controls. Pyroglutamate formation increases the rate of Aß oligomerization and alters the interactions of Aß with Cu(2+) and lipids; however, a link between these properties and the toxicity of pE-Aß peptides has not been established. We report here that Aß3pE-42 has an enhanced capacity to cause lipid peroxidation in primary cortical mouse neurons compared with the full-length isoform (Aß(1-42)). In contrast, Aß(1-42) caused a significant elevation in cytosolic reactive oxygen species, whereas Aß3pE-42 did not. We also report that Aß3pE-42 preferentially associates with neuronal membranes and triggers Ca(2+) influx that can be partially blocked by the N-methyl-d-aspartate receptor antagonist MK-801. Aß3pE-42 further caused a loss of plasma membrane integrity and remained bound to neurons at significantly higher levels than Aß(1-42) over extended incubations. Pyroglutamate formation was additionally found to increase the relative efficiency of Aß-dityrosine oligomer formation mediated by copper-redox cycling.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Señalización del Calcio , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Ascórbico/química , Permeabilidad de la Membrana Celular , Células Cultivadas , Cobre/química , Humanos , Peroxidación de Lípido , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Agregado de Proteínas , Ácido Pirrolidona Carboxílico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
2.
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
3.
Neurobiol Dis ; 71: 34-42, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25014023

RESUMEN

Disruption of redox homeostasis is a prominent feature in the pathogenesis of Huntington's disease (HD). Selenium an essential element nutrient that modulates redox pathways and has been reported to provide protection against both acute neurotoxicity (e.g. methamphetamine) and chronic neurodegeneration (e.g. tauopathy) in mice. The objective of our study was to investigate the effect of sodium selenite, an inorganic form of selenium, on behavioral, brain degeneration and biochemical outcomes in the N171-82Q Huntington's disease mouse model. HD mice, which were supplemented with sodium selenite from 6 to 14 weeks of age, demonstrated increased motor endurance, decreased loss of brain weight, decreased mutant huntingtin aggregate burden and decreased brain oxidized glutathione levels. Biochemical studies revealed that selenite treatment reverted HD-associated changes in liver selenium and plasma glutathione in N171-82Q mice and had effects on brain selenoprotein transcript expression. Further, we found decreased brain selenium content in human autopsy brain. Taken together, we demonstrate a decreased selenium phenotype in human and mouse HD and additionally show some protective effects of selenite in N171-82Q HD mice. Modification of selenium metabolism results in beneficial effects in mouse HD and thus may represent a therapeutic strategy.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Ácido Selenioso/uso terapéutico , Selenio/sangre , Expansión de Repetición de Trinucleótido/genética , Adulto , Animales , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Análisis de Supervivencia , Factores de Tiempo
4.
J Biol Chem ; 286(20): 18320-30, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454633

RESUMEN

Huntington disease (HD) is a progressive neurodegenerative disorder caused by expression of polyglutamine-expanded mutant huntingtin protein (mhtt). Most evidence indicates that soluble mhtt species, rather than insoluble aggregates, are the important mediators of HD pathogenesis. However, the differential roles of soluble monomeric and oligomeric mhtt species in HD and the mechanisms of oligomer formation are not yet understood. We have shown previously that copper interacts with and oxidizes the polyglutamine-containing N171 fragment of huntingtin. In this study we report that oxidation-dependent oligomers of huntingtin form spontaneously in cell and mouse HD models. Levels of these species are modulated by copper, hydrogen peroxide, and glutathione. Mutagenesis of all cysteine residues within N171 blocks the formation of these oligomers. In cells, levels of oligomerization-blocked mutant N171 were decreased compared with native N171. We further show that a subset of the oligomerization-blocked form of glutamine-expanded N171 huntingtin is rapidly depleted from the soluble pool compared with "native " mutant N171. Taken together, our data indicate that huntingtin is subject to specific oxidations that are involved in the formation of stable oligomers and that also delay removal from the soluble pool. These findings show that inhibiting formation of oxidation-dependent huntingtin oligomers, or promoting their dissolution, may have protective effects in HD by decreasing the burden of soluble mutant huntingtin.


Asunto(s)
Cisteína/metabolismo , Enfermedad de Huntington/metabolismo , Mutación Missense , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Multimerización de Proteína , Animales , Células COS , Chlorocebus aethiops , Cisteína/genética , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Oxidación-Reducción , Estructura Terciaria de Proteína , Solubilidad
5.
Proc Natl Acad Sci U S A ; 106(2): 381-6, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19122148

RESUMEN

Cognitive decline in Alzheimer's disease (AD) involves pathological accumulation of synaptotoxic amyloid-beta (Abeta) oligomers and hyperphosphorylated tau. Because recent evidence indicates that glycogen synthase kinase 3beta (GSK3beta) activity regulates these neurotoxic pathways, we developed an AD therapeutic strategy to target GSK3beta. The strategy involves the use of copper-bis(thiosemicarbazonoto) complexes to increase intracellular copper bioavailability and inhibit GSK3beta through activation of an Akt signaling pathway. Our lead compound Cu(II)(gtsm) significantly inhibited GSK3beta in the brains of APP/PS1 transgenic AD model mice. Cu(II)(gtsm) also decreased the abundance of Abeta trimers and phosphorylated tau, and restored performance of AD mice in the Y-maze test to levels expected for cognitively normal animals. Improvement in the Y-maze correlated directly with decreased Abeta trimer levels. This study demonstrates that increasing intracellular copper bioavailability can restore cognitive function by inhibiting the accumulation of neurotoxic Abeta trimers and phosphorylated tau.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Cobre/farmacología , Proteínas tau/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Cobre/farmacocinética , Cobre/uso terapéutico , Dimerización , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Glucógeno Sintasa Quinasas/antagonistas & inhibidores , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Compuestos Organometálicos/farmacocinética , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas tau/metabolismo
6.
J Parkinsons Dis ; 12(1): 105-115, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34744051

RESUMEN

BACKGROUND: An elevation in iron levels, together with an accumulation of α-synuclein within the oligodendrocytes, are features of the rare atypical parkinsonian disorder, Multiple System Atrophy (MSA). We have previously tested the novel compound ATH434 (formally called PBT434) in preclinical models of Parkinson's disease and shown that it is brain-penetrant, reduces iron accumulation and iron-mediated redox activity, provides neuroprotection, inhibits alpha synuclein aggregation and lowers the tissue levels of alpha synuclein. The compound was also well-tolerated in a first-in-human oral dosing study in healthy and older volunteers with a favorable, dose-dependent pharmacokinetic profile. OBJECTIVE: To evaluate the efficacy of ATH434 in a mouse MSA model. METHODS: The PLP-α-syn transgenic mouse overexpresses α-synuclein, demonstrates oligodendroglial pathology, and manifests motor and non-motor aspects of MSA. Animals were provided ATH434 (3, 10, or 30 mg/kg/day spiked into their food) or control food for 4 months starting at 12 months of age and were culled at 16 months. Western blot was used to assess oligomeric and urea soluble α-synuclein levels in brain homogenates, whilst stereology was used to quantitate the number of nigral neurons and glial cell inclusions (GCIs) present in the substantia nigra pars compacta. RESULTS: ATH434 reduced oligomeric and urea soluble α-synuclein aggregation, reduced the number of GCIs, and preserved SNpc neurons. In vitro experiments suggest that ATH434 prevents the formation of toxic oligomeric "species of synuclein". CONCLUSION: ATH434 is a promising small molecule drug candidate that has potential to move forward to trial for treating MSA.


Asunto(s)
Atrofia de Múltiples Sistemas , Enfermedad de Parkinson , Animales , Modelos Animales de Enfermedad , Humanos , Hierro/uso terapéutico , Ratones , Ratones Transgénicos , Atrofia de Múltiples Sistemas/tratamiento farmacológico , Atrofia de Múltiples Sistemas/patología , Urea , alfa-Sinucleína
7.
Biochemistry ; 50(13): 2611-22, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21348480

RESUMEN

The development of inhibitors of insulin-regulated aminopeptidase (IRAP), a membrane-bound zinc metallopeptidase, is a promising approach for the discovery of drugs for the treatment of memory loss such as that associated with Alzheimer's disease. There is, however, no consensus in the literature about the mechanism by which inhibition occurs. Sequence alignments, secondary structure predictions, and homology models based on the structures of recently determined related metallopeptidases suggest that the extracellular region consists of four domains. Partial proteolysis and mass spectrometry reported here confirm some of the domain boundaries. We have produced purified recombinant fragments of human IRAP on the basis of these data and examined their kinetic and biochemical properties. Full-length extracellular constructs assemble as dimers with different nonoverlapping fragments dimerizing as well, suggesting an extended dimer interface. Only recombinant fragments containing domains 1 and 2 possess aminopeptidase activity and bind the radiolabeled hexapeptide inhibitor, angiotensin IV (Ang IV). However, fragments lacking domains 3 and 4 possess reduced activity, although they still bind a range of inhibitors with the same affinity as longer fragments. In the presence of Ang IV, IRAP is resistant to proteolysis, suggesting significant conformational changes occur upon binding of the inhibitor. We show that IRAP has a second Zn(2+) binding site, not associated with the catalytic region, which is lost upon binding Ang IV. Modulation of activity caused by domains 3 and 4 is consistent with a conformational change regulating access to the active site of IRAP.


Asunto(s)
Cistinil Aminopeptidasa/antagonistas & inhibidores , Cistinil Aminopeptidasa/química , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Angiotensina II/análogos & derivados , Angiotensina II/metabolismo , Angiotensina II/farmacología , Sitios de Unión , Dominio Catalítico , Cistinil Aminopeptidasa/genética , Cistinil Aminopeptidasa/metabolismo , Bases de Datos de Proteínas , Humanos , Hidrólisis , Cinética , Modelos Moleculares , Terapia Molecular Dirigida , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Zinc/metabolismo
8.
J Neurochem ; 119(1): 220-30, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21797865

RESUMEN

Impaired metal ion homeostasis causes synaptic dysfunction and treatments for Alzheimer's disease (AD) that target metal ions have therefore been developed. The leading compound in this class of therapeutic, PBT2, improved cognition in a clinical trial with AD patients. The aim of the present study was to examine the cellular mechanism of action for PBT2. We show PBT2 induces inhibitory phosphorylation of the α- and ß-isoforms of glycogen synthase kinase 3 and that this activity is dependent on PBT2 translocating extracellular Zn and Cu into cells. This activity is supported when Aß:Zn aggregates are the source of extracellular Zn and adding PBT2 to Aß:Zn preparations promotes Aß degradation by matrix metalloprotease 2. PBT2-induced glycogen synthase kinase 3 phosphorylation appears to involve inhibition of the phosphatase calcineurin. Consistent with this, PBT2 increased phosphorylation of other calcineurin substrates, including cAMP response element binding protein and Ca²âº/calmodulin-dependent protein kinase. These data demonstrate PBT2 can decrease Aß levels by sequestering the Zn that promotes extracellular formation of protease resistant Aß:Zn aggregates, and that subsequent intracellular translocation of the Zn by PBT2 induces cellular responses with synapto-trophic potential. Intracellular translocation of Zn and Cu via the metal chaperone activity of PBT2 may be an important mechanism by which PBT2 improves cognitive function in people with AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Clioquinol/análogos & derivados , Glucógeno Sintasa Quinasa 3/metabolismo , Metales/metabolismo , Chaperonas Moleculares/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Western Blotting , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Caspasa 3/metabolismo , Línea Celular Tumoral , Clioquinol/farmacología , Cobre/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Espectrometría de Masas , Metaloproteinasa 2 de la Matriz/metabolismo , Péptido Hidrolasas/metabolismo , Fosforilación/efectos de los fármacos , Zinc/metabolismo
9.
Proc Natl Acad Sci U S A ; 105(19): 6813-8, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18463291

RESUMEN

Amelyoid-beta peptide (Abeta) is a major causative agent responsible for Alzheimer's disease (AD). Abeta contains a high affinity metal binding site that modulates peptide aggregation and toxicity. Therefore, identifying molecules targeting this site represents a valid therapeutic strategy. To test this hypothesis, a range of L-PtCl(2) (L = 1,10-phenanthroline derivatives) complexes were examined and shown to bind to Abeta, inhibit neurotoxicity and rescue Abeta-induced synaptotoxicity in mouse hippocampal slices. Coordination of the complexes to Abeta altered the chemical properties of the peptide inhibiting amyloid formation and the generation of reactive oxygen species. In comparison, the classic anticancer drug cisplatin did not affect any of the biochemical and cellular effects of Abeta. This implies that the planar aromatic 1,10-phenanthroline ligands L confer some specificity for Abeta onto the platinum complexes. The potent effect of the L-PtCl(2) complexes identifies this class of compounds as therapeutic agents for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Platino (Metal)/uso terapéutico , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Animales , Dicroismo Circular , Peróxido de Hidrógeno/metabolismo , Concentración 50 Inhibidora , Potenciación a Largo Plazo/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neurotoxinas/toxicidad , Oxidación-Reducción/efectos de los fármacos , Platino (Metal)/química , Platino (Metal)/farmacología , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Sincrotrones
10.
J Parkinsons Dis ; 11(4): 1821-1832, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34366375

RESUMEN

BACKGROUND: Gastrointestinal (GI) complications, that severely impact patient quality of life, are a common occurrence in patients with Parkinson's disease (PD). Damage to enteric neurons and the accumulation of alpha-synuclein in the enteric nervous system (ENS) are thought to contribute to this phenotype. Copper or iron chelators, that bind excess or labile metal ions, can prevent aggregation of alpha-synuclein in the brain and alleviate motor-symptoms in preclinical models of PD. OBJECTIVE: We investigated the effect of ATH434 (formally PBT434), a small molecule, orally bioavailable, moderate-affinity iron chelator, on colonic propulsion and whole gut transit in A53T alpha-synuclein transgenic mice. METHODS: Mice were fed ATH434 (30 mg/kg/day) for either 4 months (beginning at ∼15 months of age), after the onset of slowed propulsion ("treatment group"), or for 3 months (beginning at ∼12 months of age), prior to slowed propulsion ("prevention group"). RESULTS: ATH434, given after dysfunction was established, resulted in a reversal of slowed colonic propulsion and gut transit deficits in A53T mice to WT levels. In addition, ATH434 administered from 12 months prevented the slowed bead expulsion at 15 months but did not alter deficits in gut transit time when compared to vehicle-treated A53T mice. The proportion of neurons with nuclear Hu+ translocation, an indicator of neuronal stress in the ENS, was significantly greater in A53T than WT mice, and was reduced in both groups when ATH434 was administered. CONCLUSION: ATH434 can reverse some of the GI deficits and enteric neuropathy that occur in a mouse model of PD, and thus may have potential clinical benefit in alleviating the GI dysfunctions associated with PD.


Asunto(s)
Enfermedades Gastrointestinales , Enfermedad de Parkinson , alfa-Sinucleína , Animales , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/prevención & control , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína/genética
11.
J Biol Chem ; 284(34): 22697-702, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19574211

RESUMEN

Transgenic expression of human amyloid beta (A beta) peptide in body wall muscle cells of Caenorhabditis elegans has been used to better understand aspects of Alzheimer disease (AD). In human aging and AD, A beta undergoes post-translational changes including covalent modifications, truncations, and oligomerization. Amino truncated A beta is increasingly recognized as potentially contributing to AD pathogenesis. Here we describe surface-enhanced laser desorption ionization-time of flight mass spectrometry mass spectrometry of A beta peptide in established transgenic C. elegans lines. Surprisingly, the A beta being expressed is not full-length 1-42 (amino acids) as expected but rather a 3-42 truncation product. In vitro analysis demonstrates that A beta(3-42) self-aggregates like A beta(1-42), but more rapidly, and forms fibrillar structures. Similarly, A beta(3-42) is also the more potent initiator of A beta(1-40) aggregation. Seeded aggregation via A beta(3-42) is further enhanced via co-incubation with the transition metal Cu(II). Although unexpected, the C. elegans model of A beta expression can now be co-opted to study the proteotoxic effects and processing of A beta(3-42).


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Caenorhabditis elegans/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/ultraestructura , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/ultraestructura , Humanos , Immunoblotting , Microscopía Electrónica de Transmisión , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
J Neurochem ; 108(5): 1198-207, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19141082

RESUMEN

Accumulation of neurotoxic amyloid-beta (Abeta) is central to the pathology of Alzheimer's disease (AD). Elucidating the mechanisms of Abeta accumulation will therefore expedite the development of Abeta-targeting AD therapeutics. We examined activity of an Abeta-degrading protease (matrix metalloprotease 2) to investigate whether biochemical factors consistent with conditions in the AD brain contribute to Abeta accumulation by altering Abeta sensitivity to proteolytic degradation. An Abeta amino acid mutation found in familial AD, Abeta interactions with zinc (Zn), and increased Abeta hydrophobicity all strongly prevented Abeta degradation. Consistent to all of these factors is the promotion of specific Abeta aggregates where the protease cleavage site, confirmed by mass spectrometry, is inaccessible within an amyloid structure. These data indicate decreased degradation due to amyloid formation initiates Abeta accumulation by preventing normal protease activity. Zn also prevented Abeta degradation by the proteases neprilysin and insulin degrading enzyme. Treating Zn-induced Abeta amyloid with the metal-protein attenuating compound clioquinol reversed amyloid formation and restored the peptide's sensitivity to degradation by matrix metalloprotease 2. This provides new data indicating that therapeutic compounds designed to modulate Abeta-metal interactions can inhibit Abeta accumulation by restoring the catalytic potential of Abeta-degrading proteases.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Amiloide/efectos de los fármacos , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/genética , Clioquinol/farmacología , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática/métodos , Ácido Glutámico/genética , Glutamina/genética , Humanos , Insulisina/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Microscopía Electrónica de Transmisión/métodos , Mutación , Neprilisina/farmacología , Fragmentos de Péptidos/efectos de los fármacos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Factores de Tiempo , Zinc/farmacología
13.
Proteomics ; 8(9): 1819-31, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18384105

RESUMEN

Neurodegenerative illnesses are characterized by aberrant metabolism of biometals such as copper (Cu), zinc (Zn) and iron (Fe). However, little is known about the metabolic effects associated with altered metal homeostasis. In this study, we used an in vitro model of altered Cu homeostasis to investigate how Cu regulates cellular protein expression. Human fibroblasts containing a natural deletion mutation of the Menkes (MNK) ATP7A Cu transporter (MNK deleted) were compared to fibroblasts overexpressing ATP7A (MNK transfected). Cultures of MNK-transfected (Low-Cu) cells exhibited 95% less intracellular Cu than MNK-deleted (High-Cu) cells. Comparative proteomic analysis of the two cell-lines was performed using antibody microarrays, and significant differential protein expression was observed between Low-Cu and High-Cu cell-lines. Western blot analysis confirmed the altered protein expression of Ku80, nexilin, L-caldesmon, MAP4, Inhibitor 2 and DNA topoisomerase I. The top 50 altered proteins were analysed using the software program Pathway Studio (Ariadne Genomics) and revealed a significant over-representation of proteins involved in DNA repair and maintenance. Further analysis confirmed that expression of the DNA repair protein Ku80 was dependent on cellular Cu homeostasis and that Low-Cu levels in fibroblasts resulted in elevated susceptibility to DNA oxidation.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Cobre/química , Fibroblastos/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Antígenos Nucleares/biosíntesis , Transporte Biológico , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/metabolismo , Biología Computacional/métodos , ATPasas Transportadoras de Cobre , ADN/química , Proteínas de Unión al ADN/biosíntesis , Humanos , Autoantígeno Ku , Enfermedades Neurodegenerativas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/química , Análisis por Matrices de Proteínas , Proteómica/métodos , Programas Informáticos
14.
Neuron ; 37(6): 899-909, 2003 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-12670420

RESUMEN

Studies on postmortem brains from Parkinson's patients reveal elevated iron in the substantia nigra (SN). Selective cell death in this brain region is associated with oxidative stress, which may be exacerbated by the presence of excess iron. Whether iron plays a causative role in cell death, however, is controversial. Here, we explore the effects of iron chelation via either transgenic expression of the iron binding protein ferritin or oral administration of the bioavailable metal chelator clioquinol (CQ) on susceptibility to the Parkinson's-inducing agent 1-methyl-4-phenyl-1,2,3,6-tetrapyridine (MPTP). Reduction in reactive iron by either genetic or pharmacological means was found to be well tolerated in animals in our studies and to result in protection against the toxin, suggesting that iron chelation may be an effective therapy for prevention and treatment of the disease.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Ferritinas/genética , Quelantes del Hierro/uso terapéutico , Hierro/metabolismo , Enfermedad de Parkinson Secundaria/prevención & control , Enfermedad de Parkinson/tratamiento farmacológico , Ácido 3,4-Dihidroxifenilacético/análisis , Animales , Western Blotting , Muerte Celular , Clioquinol/uso terapéutico , Dopamina/análisis , Ferritinas/metabolismo , Expresión Génica , Terapia Genética , Ácido Homovanílico/análisis , Humanos , Inmunohistoquímica , Quelantes del Hierro/metabolismo , Ratones , Ratones Transgénicos , Estrés Oxidativo , Enfermedad de Parkinson/patología , Enfermedad de Parkinson Secundaria/inducido químicamente , Regiones Promotoras Genéticas , Ratas , Sustancia Negra/química , Sustancia Negra/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/genética
15.
J Clin Invest ; 115(9): 2556-63, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16127459

RESUMEN

The abnormal accumulation of amyloid beta-peptide (Abeta) in the form of senile (or amyloid) plaques is one of the main characteristics of Alzheimer disease (AD). Both cholesterol and Cu2+ have been implicated in AD pathogenesis and plaque formation. Abeta binds Cu2+ with very high affinity, forming a redox-active complex that catalyzes H2O2 production from O2 and cholesterol. Here we show that Abeta:Cu2+ complexes oxidize cholesterol selectively at the C-3 hydroxyl group, catalytically producing 4-cholesten-3-one and therefore mimicking the activity of cholesterol oxidase, which is implicated in cardiovascular disease. Abeta toxicity in neuronal cultures correlated with this activity, which was inhibited by Cu2+ chelators including clioquinol. Cell death induced by staurosporine or H2O2 did not elevate 4-cholesten-3-one levels. Brain tissue from AD subjects had 98% more 4-cholesten-3-one than tissue from age-matched control subjects. We observed a similar increase in the brains of Tg2576 transgenic mice compared with nontransgenic littermates; the increase was inhibited by in vivo treatment with clioquinol, which suggests that brain Abeta accumulation elevates 4-cholesten-3-one levels in AD. Cu2+-mediated oxidation of cholesterol may be a pathogenic mechanism common to atherosclerosis and AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Colesterol Oxidasa/metabolismo , Cobre/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Quelantes/metabolismo , Colestenonas/química , Colestenonas/metabolismo , Colesterol/química , Colesterol/metabolismo , Clioquinol/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Estructura Molecular , Neuronas/citología , Neuronas/metabolismo , Oxidación-Reducción
16.
Biochem J ; 407(3): 435-50, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17680773

RESUMEN

Biometals have an important role in AD (Alzheimer's disease) and metal ligands have been investigated as potential therapeutic agents for treatment of AD. In recent studies the 8HQ (8-hydroxyquinoline) derivative CQ (clioquinol) has shown promising results in animal models and small clinical trials; however, the actual mode of action in vivo is still being investigated. We previously reported that CQ-metal complexes up-regulated MMP (matrix metalloprotease) activity in vitro by activating PI3K (phosphoinositide 3-kinase) and JNK (c-jun N-terminal kinase), and that the increased MMP activity resulted in enhanced degradation of secreted Abeta (amyloid beta) peptide. In the present study, we have further investigated the biochemical mechanisms by which metal ligands affect Abeta metabolism. To achieve this, we measured the effects of diverse metal ligands on cellular metal uptake and secreted Abeta levels in cell culture. We report that different classes of metal ligands including 8HQ and phenanthroline derivatives and the sulfur compound PDTC (pyrrolidine dithiocarbamate) elevated cellular metal levels (copper and zinc), and resulted in substantial loss of secreted Abeta. Generally, the ability to inhibit Abeta levels correlated with a higher lipid solubility of the ligands and their capacity to increase metal uptake. However, we also identified several ligands that potently inhibited Abeta levels while only inducing minimal change to cellular metal levels. Metal ligands that inhibited Abeta levels [e.g. CQ, 8HQ, NC (neocuproine), 1,10-phenanthroline and PDTC] induced metal-dependent activation of PI3K and JNK, resulting in JNK-mediated up-regulation of metalloprotease activity and subsequent loss of secreted Abeta. The findings in the present study show that diverse metal ligands with high lipid solubility can elevate cellular metal levels resulting in metalloprotease-dependent inhibition of Abeta. Given that a structurally diverse array of ligands was assessed, the results are consistent with the effects being due to metal transport rather than the chelating ligand interacting directly with a receptor.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Cobre/metabolismo , Péptidos/metabolismo , Zinc/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/genética , Animales , Transporte Biológico Activo/genética , Células CHO , Cricetinae , Cricetulus , Humanos , Ligandos , Oxiquinolina/análogos & derivados , Oxiquinolina/metabolismo , Péptidos/genética , Fenantrolinas/metabolismo
17.
Metallomics ; 10(9): 1339-1347, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30168573

RESUMEN

Tauopathies are characterized by the pathological accumulation of the microtubule associated protein tau within the brain. We demonstrate here that a copper/zinc chaperone (PBT2, Prana Biotechnology) has rapid and profound effects in the rTg(tauP301L)4510 mouse model of tauopathy. This was evidenced by significantly improved cognition, a preservation of neurons, a decrease in tau aggregates and a decrease in other forms of "pathological" tau (including phosphorylated tau and sarkosyl-insoluble tau). Our data demonstrate that one of the primary mechanisms of action of PBT2 in this model may be driven by an interaction on the pathways responsible for the dephosphorylation of tau. Specifically, PBT2 increased protein levels of both the structural and catalytic subunits of protein phosphatase 2A (PP2A), decreased levels of the methyl esterase (PME1) that dampens PP2A activity, and increased levels of the prolyl isomerase (Pin1) that stimulates the dephosphorylation activity of PP2A. None of these effects were observed when the metal binding site of PBT2 was blocked. This highlights the potential utility of targeting metal ions as a novel therapeutic strategy for diseases in which tau pathology is a feature, which includes conditions such as frontotemporal dementia and Alzheimer's disease.


Asunto(s)
Clioquinol/análogos & derivados , Tauopatías/tratamiento farmacológico , Animales , Clioquinol/uso terapéutico , Femenino , Masculino , Memoria/efectos de los fármacos , Ratones , Aprendizaje Espacial/efectos de los fármacos
18.
Aging Cell ; 5(2): 153-65, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16626394

RESUMEN

Transgenic mice carrying mutant Cu/Zn superoxide dismutase (SOD1) recapitulate the motor impairment of human amyotrophic lateral sclerosis (ALS). The amyloid-beta (Abeta) peptide associated with Alzheimer's disease is neurotoxic. To investigate the potential role of Abeta in ALS development, we generated a double transgenic mouse line that overexpresses SOD1(G93A) and amyloid precursor protein (APP)-C100. The transgenic mouse C100.SOD1(G93A) overexpresses Abeta and shows earlier onset of motor impairment but has the same lifespan as the single transgenic SOD1(G93A) mouse. To determine the mechanism associated with this early-onset phenotype, we measured copper and zinc levels in brain and spinal cord and found both significantly elevated in the single and double transgenic mice compared with their littermate control mice. Increased glial fibrillary acidic protein and decreased APP levels in the spinal cord of C100.SOD1(G93A) mice compared with the SOD1(G93A) mice agree with the neuronal damage observed by immunohistochemical analysis. In the spinal cords of C100.SOD1(G93A) double transgenic mice, soluble Abeta was elevated in mice at end-stage disease compared with the pre-symptomatic stage. Buffer-insoluble SOD1 aggregates were significantly elevated in the pre-symptomatic mice of C100.SOD1(G93A) compared with the age-matched SOD1(G93A) mice, correlating with the earlier onset of motor impairment in the C100.SOD1(G93A) mice. This study supports abnormal SOD1 protein aggregation as the pathogenic mechanism in ALS, and implicates a potential role for Abeta in the development of ALS by exacerbating SOD1(G93A) aggregation.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Esclerosis Amiotrófica Lateral/enzimología , Esclerosis Amiotrófica Lateral/fisiopatología , Modelos Animales de Enfermedad , Movimiento/fisiología , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Supervivencia Celular , Cobre/metabolismo , Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Ratones Transgénicos , Estrés Oxidativo , Estructura Cuaternaria de Proteína , Médula Espinal/citología , Médula Espinal/metabolismo , Superóxido Dismutasa/química , Superóxido Dismutasa-1 , Zinc/metabolismo
19.
Aging Cell ; 5(1): 69-79, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16441845

RESUMEN

Neocortical beta-amyloid (Abeta) aggregates in Alzheimer's disease (AD) are enriched in transition metals that mediate assembly. Clioquinol (CQ) targets metal interaction with Abeta and inhibits amyloid pathology in transgenic mice. Here, we investigated the binding properties of radioiodinated CQ ([(125)I]CQ) to different in vitro and in vivo Alzheimer models. We observed saturable binding of [(125)I]CQ to synthetic Abeta precipitated by Zn(2+) (K(d)=0.45 and 1.40 nm for Abeta(1-42) and Abeta(1-40), respectively), which was fully displaced by free Zn(2+), Cu(2+), the chelator DTPA (diethylene triamine pentaacetic acid) and partially by Congo red. Sucrose density gradient of post-mortem AD brain indicated that [(125)I]CQ concentrated in a fraction enriched for both Abeta and Zn, which was modulated by exogenous addition of Zn(2+) or DTPA. APP transgenic (Tg2576) mice injected with [(125)I]CQ exhibited higher brain retention of tracer compared to non-Tg mice. Autoradiography of brain sections of these animals confirmed selective [(125)I]CQ enrichment in the neocortex. Histologically, both thioflavine-S (ThS)-positive and negative structures were labeled by [(125)I]CQ. A pilot SPECT study of [(123)I]CQ showed limited uptake of the tracer into the brain, which did however, appear to be more rapid in AD patients compared to age-matched controls. These data support metallated Abeta species as the neuropharmacological target of CQ and indicate that this drug class may have potential as in vivo imaging agents for Alzheimer neuropathology.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Clioquinol , Zinc/metabolismo , Animales , Biomarcadores/metabolismo , Encéfalo/citología , Encéfalo/patología , Clioquinol/metabolismo , Clioquinol/farmacocinética , Humanos , Radioisótopos de Yodo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proyectos Piloto , Unión Proteica , Tomografía Computarizada de Emisión de Fotón Único
20.
Brain Res ; 1140: 188-94, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-16631136

RESUMEN

Ferritin elevation has been reported by some laboratories to occur within the substantia nigra (SN), the area of the brain affected in Parkinson's disease (PD), but whether such an increase could be causatively involved in neurodegeneration associated with the disorder is unknown. Here, we report that chronic ferritin elevation in midbrain dopamine-containing neurons results in a progressive age-related neurodegeneration of these cells. This provides strong evidence that chronic ferritin overload could be directly involved in age-related neurodegeneration such as occurs in Parkinson's and other related diseases.


Asunto(s)
Envejecimiento , Dopamina/metabolismo , Ferritinas/metabolismo , Degeneración Nerviosa , Neuronas/metabolismo , Sustancia Negra/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Factores de Edad , Animales , Conducta Exploratoria/fisiología , Ferritinas/genética , Fluoresceínas , Expresión Génica/genética , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Transgénicos , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Compuestos Orgánicos/metabolismo , Tinción con Nitrato de Plata , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA