Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurochem ; 147(3): 409-428, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30091236

RESUMEN

The Parkinson's disease (PD)-causative leucine-rich repeat kinase 2 (LRRK2) belongs to the Roco family of G-proteins comprising a Ras-of-complex (Roc) domain followed by a C-terminal of Roc (COR) domain in tandem (called Roc-COR domain). Two prokaryotic Roc-COR domains have been characterized as 'G proteins activated by guanine nucleotide-dependent dimerization' (GADs), which require dimerization for activation of their GTPase activity and bind guanine nucleotides with relatively low affinities. Additionally, LRRK2 Roc domain in isolation binds guanine nucleotides with relatively low affinities. As such, LRRK2 GTPase domain was predicted to be a GAD. Herein, we describe the design and high-level expression of human LRRK2 Roc-COR domain (LRRK2 Roc-COR). Biochemical analyses of LRRK2 Roc-COR reveal that it forms homodimers, with the C-terminal portion of COR mediating its dimerization. Furthermore, it co-purifies and binds Mg2+ GTP/GDP at 1 : 1 stoichiometry, and it hydrolyzes GTP with Km  and kcat  of 22 nM and 4.70 × 10-4  min-1 ,  respectively. Thus, even though LRRK2 Roc-COR forms GAD-like homodimers, it exhibits conventional Ras-like GTPase properties, with high-affinity binding of Mg2+ -GTP/GDP and low intrinsic catalytic activity. The PD-causative Y1699C mutation mapped to the COR domain was previously reported to reduce the GTPase activity of full-length LRRK2. In contrast, this mutation induces no change in the GTPase activity, and only slight perturbations in the secondary structure contents of LRRK2 Roc-COR. As this mutation does not directly affect the GTPase activity of the isolated Roc-COR tandem, it is possible that the effects of this mutation on full-length LRRK2 occur via other functional domains. Open Practices Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.


Asunto(s)
GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Genes ras/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Animales , Dimerización , Escherichia coli , Regulación Enzimológica de la Expresión Génica/genética , Nucleótidos de Guanina/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/química , Magnesio/metabolismo , Ratones , Mutación/genética , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Multimerización de Proteína , Estructura Secundaria de Proteína/genética , Proteínas Recombinantes , Proteína de Unión al GTP rac1/biosíntesis , Proteína de Unión al GTP rac1/genética
2.
Inorg Chem ; 54(2): 470-5, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25409492

RESUMEN

The design of small molecules that can target the aggregation of Aß as potential therapeutic agents for Alzheimer's disease is an area of study that has attracted a lot of attention recently. The novel ligand methyl 1-butyl-2-pyridyl-benzimidazole carboxylate was prepared for the synthesis of a series of new iridium(III), ruthenium(II), and platinum(II) 2-pyridyl-benzimidazole complexes. The crystal structure of the half-sandwich iridium(III) complex was established by X-ray diffraction. An arrangement of two cationic complexes in the unit cell is observed, and it seems to be organized by weak π···π interactions that are taking place between two symmetry-related benzimidazole ring systems. All new compounds inhibited aggregation of Aß1-42 in vitro as shown by both thioflavin T fluorescence assay and transmission electron microscopy. Among them the Ir compound rescued the toxicity of Aß1-42 in primary cortical neurons effectively.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Bencimidazoles/química , Neuronas/efectos de los fármacos , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/farmacología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Multimerización de Proteína/efectos de los fármacos , Animales , Técnicas de Química Sintética , Diseño de Fármacos , Femenino , Iridio/química , Ligandos , Ratones , Modelos Moleculares , Conformación Molecular , Neuronas/citología , Compuestos Organometálicos/química , Platino (Metal)/química , Embarazo , Estructura Secundaria de Proteína , Rutenio/química
3.
Mol Biosyst ; 11(1): 190-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25352253

RESUMEN

The 140 residue intrinsically disordered protein α-synuclein (α-syn) self-associates to form fibrils that are the major constituent of the Lewy body intracellular protein inclusions, and neurotoxic oligomers. Both of these macromolecular structures are associated with a number of neurodegenerative diseases, including Parkinson's disease and dementia with Lewy bodies. Using ensemble optimisation modelling (EOM) and small angle X-ray scattering (SAXS) on a size-exclusion column equipped beamline, we studied how the distribution of structural conformers in α-syn may be influenced by the presence of the familial early-onset mutations A30P, E45K and A53T, by substituting the four methionine residues with alanines and by reaction with copper (Cu2+) or an anti-fibril organic platinum (Pt) complex. We found that the WT had two major conformer groups, representing ensembles of compact and extended structures. The population of the extended group was increased in the more rapidly fibril-forming E45K and A53T mutants, while the compact group was enlarged in the oligomer-forming A30P mutant. Addition of Cu2+ resulted in the formation of an ensemble of compact conformers, while the anti-fibril agent and alanine substitution substantially reduced the population of extended conformers. Since our observations with the mutants suggest that fibrils may be drawn from the extended conformer ensemble, we propose that the compact and extended ensembles represent the beginning of oligomer and fibril formation pathways respectively, both of which have been reported to lead to a toxic gain of function. Manipulating these pathways and monitoring the results by EOM and SAXS may be useful in the development of anti-Parkinson's disease therapies.


Asunto(s)
Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , alfa-Sinucleína/química , Sustitución de Aminoácidos , Mutación , Relación Estructura-Actividad , alfa-Sinucleína/genética
5.
Inorg Chem ; 52(8): 4303-18, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23537393

RESUMEN

8-Hydroxyquinolines (8HQ) have found widespread application in chemistry and biology due to their ability to complex a range of transition metal ions. The family of 2-substituted 8HQs has been proposed for use in the treatment of Alzheimer's disease (AD). Most notably, the therapeutic PBT2 (Prana Biotechnology Ltd.) has been shown to act as an efficient metal chaperone, disaggregate metal-enriched amyloid plaques comprised of the Aß peptide, inhibit Cu/Aß redox chemistry, and reverse the AD phenotype in transgenic animal models. Yet surprisingly little is known about the molecular interactions at play. In this study, we show that the homologous ligand 2-[(dimethylamino)methyl]-8-hydroxyquinoline (HL) forms a CuL complex with a conditional (apparent) dissociation constant of 0.33 nM at pH 6.9 and is capable of forming ternary Cu(2+) complexes with neurotransmitters including histamine (HA), glutamic acid (Glu), and glycine (Gly), with glutathione disulfide (GSSG), and with histidine (His) side chains of proteins and peptides including the Aß peptide. Our findings suggest a molecular basis for the strong metal chaperone activity of PBT2, its ability to attenuate Cu(2+)/Aß interactions, and its potential to promote neuroprotective and neuroregenerative effects.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Cobre/química , Cobre/farmacología , Oxiquinolina/análogos & derivados , Oxiquinolina/farmacología , Enfermedad de Alzheimer/metabolismo , Ácido Glutámico/metabolismo , Disulfuro de Glutatión/metabolismo , Glicina/metabolismo , Histamina/metabolismo , Histidina/metabolismo , Humanos , Ligandos , Modelos Moleculares , Neurotransmisores/metabolismo
7.
Mol Neurodegener ; 7: 57, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23171715

RESUMEN

BACKGROUND: The definitive indicator of Alzheimer's disease (AD) pathology is the profuse accumulation of amyloid-ß (Aß) within the brain. Various in vitro and cell-based models have been proposed for high throughput drug screening for potential therapeutic benefit in diseases of protein misfolding. Caenorhabditis elegans offers a convenient in vivo system for examination of Aß accumulation and toxicity in a complex multicellular organism. Ease of culturing and a short life cycle make this animal model well suited to rapid screening of candidate compounds. RESULTS: We have generated a new transgenic strain of C. elegans that expresses full length Aß1₋42. This strain differs from existing Aß models that predominantly express amino-truncated Aß3₋42. The Aß1₋42 is expressed in body wall muscle cells, where it oligomerizes, aggregates and results in severe, and fully penetrant, age progressive-paralysis. The in vivo accumulation of Aß1₋42 also stains positive for amyloid dyes, consistent with in vivo fibril formation. The utility of this model for identification of potential protective compounds was examined using the investigational Alzheimer's therapeutic PBT2, shown to be neuroprotective in mouse models of AD and significantly improve cognition in AD patients. We observed that treatment with PBT2 provided rapid and significant protection against the Aß-induced toxicity in C. elegans. CONCLUSION: This C. elegans model of full length Aß1₋42 expression can now be adopted for use in screens to rapidly identify and assist in development of potential therapeutics and to study underlying toxic mechanism(s) of Aß.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/toxicidad , Caenorhabditis elegans , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Animales , Animales Modificados Genéticamente , Clioquinol/análogos & derivados , Clioquinol/farmacología , Evaluación Preclínica de Medicamentos/métodos , Humanos
8.
Br J Pharmacol ; 163(2): 211-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21232050

RESUMEN

Alzheimer's disease (AD) is the most common age related neurodegenerative disease. Currently, there are no disease modifying drugs, existing therapies only offer short-term symptomatic relief. Two of the pathognomonic indicators of AD are the presence of extracellular protein aggregates consisting primarily of the Aß peptide and oxidative stress. Both of these phenomena can potentially be explained by the interactions of Aß with metal ions. In addition, metal ions play a pivotal role in synaptic function and their homeostasis is tightly regulated. A breakdown in this metal homeostasis and the generation of toxic Aß oligomers are likely to be responsible for the synaptic dysfunction associated with AD. Therefore, approaches that are designed to prevent Aß metal interactions, inhibiting the formation of toxic Aß species as well as restoring metal homeostasis may have potential as disease modifying strategies for treating AD. This review summarizes the physiological and pathological interactions that metal ions play in synaptic function with particular emphasis placed on interactions with Aß. A variety of therapeutic strategies designed to address these pathological processes are also described. The most advanced of these strategies is the so-called 'metal protein attenuating compound' approach, with the lead molecule PBT2 having successfully completed early phase clinical trials. The success of these various strategies suggests that manipulating metal ion interactions offers multiple opportunities to develop disease modifying therapies for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Cobre/fisiología , Zinc/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/fisiología , Animales , Sitios de Unión , Cationes , Ensayos Clínicos como Asunto , Clioquinol/análogos & derivados , Clioquinol/farmacología , Clioquinol/uso terapéutico , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , Humanos , Estrés Oxidativo , Unión Proteica , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
9.
ACS Chem Neurosci ; 1(11): 720-7, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-22778809

RESUMEN

Apoptotic cell death via activation of the caspase family of cysteine proteases is a common feature of many neurodegenerative diseases including Creutzfeldt-Jakob disease. Molecular imaging of cysteine protease activities at the preclinical stage may provide valuable mechanistic information about pathophysiological pathways involved in disease evolution and in response to therapy. In this study, we report synthesis and characterization of a near-infrared (NIR) fluorescent contrast agent capable of noninvasively imaging neuronal apoptosis in vivo, by conjugating a NIR cyanine dye to Val-Ala-Asp-fluoromethylketone (VAD-fmk), a general inhibitor of active caspases. Following intravenous administration of the NIR-VAD-fmk contrast agent, in vivo fluorescence reflectance imaging identified significantly higher levels of active caspases in the brain of mice with advanced but preclinical prion disease, when compared with healthy controls. The contrast agent and related analogues will enable the longitudinal study of disease progression and therapy in animal models of many neurodegenerative conditions.


Asunto(s)
Apoptosis/fisiología , Carbocianinas , Neuroimagen/métodos , Neuronas/patología , Oligopéptidos , Enfermedades por Prión/patología , Animales , Western Blotting , Carbocianinas/síntesis química , Caspasas/metabolismo , Colorantes Fluorescentes , Humanos , Inmunohistoquímica , Rayos Infrarrojos , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , Oligopéptidos/síntesis química
10.
J Mol Biol ; 387(3): 771-85, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19361420

RESUMEN

The deposition of alpha-synuclein (alpha-syn) aggregates in dopaminergic neurons is a key feature of Parkinson's disease. While dopamine (DA) can modulate alpha-syn aggregation, it is unclear which other factors can regulate the actions of DA on alpha-syn. In this study, we investigated the effect of solution conditions (buffer, salt and pH) on the oligomerization of alpha-syn by DA. We show that alpha-syn oligomerization is dependent on the oxidation of DA into reactive intermediates. Under acidic pH conditions, DA is stable, and DA-mediated oligomerization of alpha-syn is inhibited. From pH 7.0 to pH 11.0, DA is unstable and undergoes redox reactions, promoting the formation of SDS-resistant soluble oligomers of alpha-syn. We show that the reactive intermediate 5,6-dihydroxylindole mediates the formation of alpha-syn soluble oligomers under physiological conditions (pH 7.4). In contrast, under acidic conditions (pH 4.0), 5,6-dihydroxylindole promotes the formation of SDS-resistant insoluble oligomers that further associate to form sheet-like fibrils with beta-sheet structure that do not bind the dye thioflavin T. These results suggest that distinct reactive intermediates of DA, and not DA itself, interact with alpha-syn to generate the alpha-syn aggregates implicated in Parkinson's disease.


Asunto(s)
Dopamina , Concentración de Iones de Hidrógeno , Indoles , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Antioxidantes/química , Tampones (Química) , Dopamina/química , Dopamina/metabolismo , Humanos , Indoles/química , Indoles/metabolismo , Estructura Molecular , Oxidación-Reducción , Enfermedad de Parkinson/metabolismo , Pliegue de Proteína , Estructura Secundaria de Proteína , Sales (Química)/química , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X , alfa-Sinucleína/ultraestructura
11.
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
12.
Recent Pat CNS Drug Discov ; 2(3): 188-99, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18221231

RESUMEN

Accumulation of Abeta peptide in the brain results in the formation of amyloid plaques characteristic of Alzheimer's disease (AD) pathology. Abeta soluble oligomers and protofibrils are neurotoxic and these are believed to be a major cause of neurodegeneration in AD. Abeta is derived from a precursor protein by two sequential cleavage steps involving beta- and gamma-secretases, two proteolytic enzymes that represent rational drug targets. beta-secretase was identified as the membrane-anchored aspartyl protease BACE (or BACE1) and found to be elevated in brain cortex of patients with sporadic Alzheimer's disease. In this review, we summarize current approaches towards the development of BACE inhibitors with focus on bioactive compounds and related patents. Recent reports have described drugs that are effective at inhibiting Abeta production in the brain of transgenic mouse models. The beginning of Phase I clinical trials has been approved for one of them and we can expect that in the near future BACE inhibitors will provide novel effective therapeutics to treat AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Etilenos/antagonistas & inhibidores , Etilenos/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...