Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
MAbs ; 14(1): 2080628, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771588

RESUMEN

Approaches for antibody discovery have seen substantial improvement and success in recent years. Yet, advancing antibodies into the clinic remains difficult because therapeutic developability concerns are challenging to predict. We developed a computational model to simplify antibody developability assessment and enable accelerated early-stage screening. To this end, we quantified the ability of hundreds of sequence- and structure-based descriptors to differentiate clinical antibodies that have undergone rigorous screening and characterization for drug-like properties from antibodies in the human repertoire that are not natively paired. This analysis identified 144 descriptors capable of distinguishing clinical from repertoire antibodies. Five descriptors were selected and combined based on performance and orthogonality into a single model referred to as the Therapeutic Antibody Developability Analysis (TA-DA). On a hold-out test set, this tool separated clinical antibodies from repertoire antibodies with an AUC = 0.8, demonstrating the ability to identify developability attributes unique to clinical antibodies. Based on our results, the TA-DA score may serve as an approach for selecting lead antibodies for further development.Abbreviations: Affinity-Capture Self-Interaction Nanoparticle Spectroscopy (AC-SINS), Area Under the Curve (AUC), Complementary-Determining Region (CDR), Clinical-Stage Therapeutics (CST), Framework (FR), Monoclonal Antibodies (mAbs), Observed Antibody Space (OAS), Receiver Operating Characteristic (ROC), Size-Exclusion Chromatography (SEC), Structural Aggregation Propensity (SAP), Therapeutic Antibody Developability Analysis (TA-DA), Therapeutic Antibody Profiler (TAP), Therapeutic Structural Antibody Database (Thera-SAbDab), Variable Heavy (VH), Variable Light (VL).


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Monoclonales/química , Cromatografía en Gel , Humanos
2.
Nature ; 596(7870): 103-108, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34153975

RESUMEN

Rapidly emerging SARS-CoV-2 variants jeopardize antibody-based countermeasures. Although cell culture experiments have demonstrated a loss of potency of several anti-spike neutralizing antibodies against variant strains of SARS-CoV-21-3, the in vivo importance of these results remains uncertain. Here we report the in vitro and in vivo activity of a panel of monoclonal antibodies (mAbs), which correspond to many in advanced clinical development by Vir Biotechnology, AbbVie, AstraZeneca, Regeneron and Lilly, against SARS-CoV-2 variant viruses. Although some individual mAbs showed reduced or abrogated neutralizing activity in cell culture against B.1.351, B.1.1.28, B.1.617.1 and B.1.526 viruses with mutations at residue E484 of the spike protein, low prophylactic doses of mAb combinations protected against infection by many variants in K18-hACE2 transgenic mice, 129S2 immunocompetent mice and hamsters, without the emergence of resistance. Exceptions were LY-CoV555 monotherapy and LY-CoV555 and LY-CoV016 combination therapy, both of which lost all protective activity, and the combination of AbbVie 2B04 and 47D11, which showed a partial loss of activity. When administered after infection, higher doses of several mAb cocktails protected in vivo against viruses with a B.1.351 spike gene. Therefore, many-but not all-of the antibody products with Emergency Use Authorization should retain substantial efficacy against the prevailing variant strains of SARS-CoV-2.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/farmacología , Anticuerpos Antivirales/uso terapéutico , COVID-19/virología , Pruebas de Neutralización , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/inmunología , COVID-19/genética , COVID-19/inmunología , COVID-19/prevención & control , Chlorocebus aethiops , Femenino , Humanos , Masculino , Mesocricetus/inmunología , Mesocricetus/virología , Ratones , Ratones Transgénicos , Profilaxis Posexposición , Profilaxis Pre-Exposición , SARS-CoV-2/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Células Vero
3.
Methods Mol Biol ; 988: 227-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23475724

RESUMEN

Development of a thorough understanding of the solution polydispersity of therapeutic glycoproteins including monoclonal antibodies is an important and challenging undertaking. Degradation pathways involving fragmentation could result in loss of therapeutic efficacy. Protein aggregation on the other hand is frequently considered a critical quality attribute, and concerns exist that protein aggregates could result in undesirable immunological consequences (1). Sedimentation velocity analysis performed in the analytical ultracentrifuge (SV-AUC) provides a uniquely powerful first principal measure of the hydrodynamic size and shape of proteins under conditions that can come very close to the formulated drug product. This technique avoids the potential pitfalls associated with size exclusion chromatography (SEC) including on-column dilution, adsorption or disruption of species by a stationary phase, and the need to use high ionic strength mobile phases to screen unwanted electrostatic interactions (2, 3). Furthermore, not only does SV-AUC provide a quantitative size distribution analysis, but it also provides information about macromolecular conformation. For these reasons, use of SV-AUC for analysis of therapeutic monoclonal antibodies has become widespread throughout the biopharmaceutical industry and is one of the most common orthogonal techniques to SEC for measuring aggregate and fragment levels (4-9). The studies outlined in this chapter describe the basic principles of designing, collecting, and analyzing experimental data using SV-AUC with a focus on methods for therapeutic monoclonal antibodies and other similar biologics. Details are given that facilitate the acquisition of high quality data sets that in turn simplify data analysis resulting in robust and accurate measures of solution polydispersity.


Asunto(s)
Anticuerpos Monoclonales/química , Algoritmos , Centrifugación por Gradiente de Densidad/métodos , Interpretación Estadística de Datos
4.
MAbs ; 4(4): 521-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22647389

RESUMEN

Therapeutic proteins circulating in blood are in a highly crowded, redox environment at high temperatures of ~37°C. These molecules circulate in the presence of enzymes and other serum proteins making it difficult to predict from in vitro studies the stability, aggregation or pharmacokinetics of a therapeutic protein in vivo. Here, we describe use of a high throughput capillary electrophoresis based microfluidic device (LabChip GXII) to obtain pharmacokinetics (PK) of a fluorescently labeled human mAb directly from serum. The non-labeled and labeled mAbs were evaluated in single dose rat PK studies using a traditional ELISA method or LabChip GXII, respectively. The fluorescent dye did not significantly alter clearance of this particular mAb, and PK parameters were comparable for labeled and unlabeled molecules. Further, from the CE profile we concluded that the mAb was resistant to fragmentation or aggregation during circulation. In a follow-up experiment, dimers were generated from the mAb using photo-induced cross-linking of unmodified proteins (PICUP) and labeled with the same fluorophore. The extent of dimerization was incomplete and some monomer and higher molecular weight species were found in the preparation. In rat PK studies, the serum concentration-time profile of the three entities present in the dimer preparation could be followed simultaneously with the GXII technology. While further studies are warranted, we believe this method could be adapted to obtain PK of different forms of antibodies (oxidized, deamidated or various glycosylated species) and other proteins.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Electroforesis Capilar/métodos , Técnicas Analíticas Microfluídicas/métodos , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/química , Colorantes Fluorescentes/química , Humanos , Masculino , Multimerización de Proteína , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo
5.
Mol Neurodegener ; 7: 8, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22423893

RESUMEN

BACKGROUND: The form(s) of amyloid-ß peptide (Aß) associated with the pathology characteristic of Alzheimer's disease (AD) remains unclear. In particular, the neurotoxicity of intraneuronal Aß accumulation is an issue of considerable controversy; even the existence of Aß deposits within neurons has recently been challenged by Winton and co-workers. These authors purport that it is actually intraneuronal APP that is being detected by antibodies thought to be specific for Aß. To further address this issue, an anti-Aß antibody was developed (MOAB-2) that specifically detects Aß, but not APP. This antibody allows for the further evaluation of the early accumulation of intraneuronal Aß in transgenic mice with increased levels of human Aß in 5xFAD and 3xTg mice. RESULTS: MOAB-2 (mouse IgG2b) is a pan-specific, high-titer antibody to Aß residues 1-4 as demonstrated by biochemical and immunohistochemical analyses (IHC), particularly compared to 6E10 (a commonly used commercial antibody to Aß residues 3-8). MOAB-2 did not detect APP or APP-CTFs in cell culture media/lysates (HEK-APPSwe or HEK-APPSwe/BACE1) or in brain homogenates from transgenic mice expressing 5 familial AD (FAD) mutation (5xFAD mice). Using IHC on 5xFAD brain tissue, MOAB-2 immunoreactivity co-localized with C-terminal antibodies specific for Aß40 and Aß42. MOAB-2 did not co-localize with either N- or C-terminal antibodies to APP. In addition, no MOAB-2-immunoreactivity was observed in the brains of 5xFAD/BACE-/- mice, although significant amounts of APP were detected by N- and C-terminal antibodies to APP, as well as by 6E10. In both 5xFAD and 3xTg mouse brain tissue, MOAB-2 co-localized with cathepsin-D, a marker for acidic organelles, further evidence for intraneuronal Aß, distinct from Aß associated with the cell membrane. MOAB-2 demonstrated strong intraneuronal and extra-cellular immunoreactivity in 5xFAD and 3xTg mouse brain tissues. CONCLUSIONS: Both intraneuronal Aß accumulation and extracellular Aß deposition was demonstrated in 5xFAD mice and 3xTg mice with MOAB-2, an antibody that will help differentiate intracellular Aß from APP. However, further investigation is required to determine whether a molecular mechanism links the presence of intraneuronal Aß with neurotoxicity. As well, understanding the relevance of these observations to human AD patients is critical.


Asunto(s)
Péptidos beta-Amiloides/análisis , Precursor de Proteína beta-Amiloide/análisis , Anticuerpos/análisis , Inmunohistoquímica/métodos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/inmunología , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Placa Amiloide/química , Placa Amiloide/patología
6.
Methods Mol Biol ; 670: 13-32, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-20967580

RESUMEN

This chapter outlines protocols that produce homogenous preparations of oligomeric and fibrillar amyloid-ß peptide (Aß). While there are several isoforms of this peptide, the 42 amino acid form is the focus because of its genetic and pathological link to Alzheimer's disease (AD). Past decades of AD research highlight the dependence of Aß42 function on its structural assembly state. Biochemical, cellular and in vivo studies of Aß42 usually begin with purified peptide obtained by chemical synthesis or recombinant expression. The initial steps to solubilize and prepare these purified dry peptide stocks are critical to controlling the structural assembly of Aß. To develop homogenous Aß42 assemblies, we initially monomerize the peptide, erasing any "structural history" that could seed aggregation, by using a strong solvent. It is this starting material that has allowed us to define and optimize conditions that consistently produce homogenous solutions of soluble oligomeric and fibrillar Aß42 assemblies. These preparations have been developed and characterized by using atomic force microscopy (AFM) to identify the structurally discrete species formed by Aß42 under specific solution conditions. These preparations have been used extensively to demonstrate a variety of functional differences between oligomeric and fibrillar Aß42. We also present a protocol for fluorescently labeling oligomeric Aß42 that does not affect structure, as measured by AFM, or function, as measured by a cellular uptake assay. These reagents are critical experimental tools that allow for defining specific structure/function connections.


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/síntesis química , Péptidos beta-Amiloides/toxicidad , Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Ratones , Microscopía de Fuerza Atómica
7.
Neurosci Lett ; 403(1-2): 162-5, 2006 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-16765515

RESUMEN

Amyloid-beta (Abeta) is causally implicated in Alzheimer's disease and neuroplasticity failure has acquired validity as a possible mechanism of early AD pathogenesis. We have previously demonstrated that oligomeric Abeta(1-42) inhibits LTP in the dentate gyrus of rat hippocampal slices. We now show, using whole cell recordings in hippocampal granule cells, that oligomeric Abeta(1-42) decreases neuronal excitability. In particular, Abeta(1-42) application was associated with a decrease in the number of action potentials fired in response to current injection, and with an increase in the amplitude of the afterhyperpolarization. Reduced excitability may underlie the Abeta-mediated impairment in neuroplasticity, and ultimately may contribute to the memory loss in Alzheimer disease.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Giro Dentado/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/fisiología , Potenciales de Acción , Péptidos beta-Amiloides/farmacología , Animales , Biopolímeros , Giro Dentado/efectos de los fármacos , Técnicas In Vitro , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología
8.
Biochemistry ; 45(2): 381-90, 2006 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-16401069

RESUMEN

Recent studies have shown that the lipidation and assembly state of apolipoprotein E (apoE) determine receptor recognition and amyloid-beta peptide (Abeta) binding. We previously demonstrated that apoE secreted by HEK cells stably expressing apoE3 or apoE4 (HEK-apoE) binds Abeta and inhibits Abeta-induced neurotoxicity by an isoform-specific process that requires apoE receptors. Here we characterized the structure of HEK-apoE assemblies and determined their receptor binding specificity. By chromatography, HEK-apoE elutes in high molecular mass fractions and is the size of plasma HDL, consistent with a multiprotein assembly. No lipid was associated with these apoE assemblies. Several methods for analyzing receptor binding indicate that HEK-apoE is a ligand for low-density lipoprotein (LDL) receptor-related protein (LRP) but not the LDL receptor. This suggests that self-assembly of apoE may induce a functional conformation necessary for binding to LRP. Our results indicate that, in addition to lipid content, the assembly state of apoE influences Abeta binding and receptor recognition.


Asunto(s)
Apolipoproteínas E/química , Apolipoproteínas E/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Animales , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/ultraestructura , Línea Celular , Cromatografía en Gel , Fibroblastos/metabolismo , Humanos , Radioisótopos de Yodo , Ligandos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/química , Ratones , Unión Proteica , Receptores de LDL/metabolismo , Ultracentrifugación
9.
Neurol Res ; 27(8): 869-81, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16354549

RESUMEN

Extracellular fibrillar amyloid deposits are prominent and universal Alzheimer's disease (AD) features, but senile plaque abundance does not always correlate directly with the degree of dementia exhibited by AD patients. The mechanism(s) and dynamics of Abeta fibril genesis and deposition remain obscure. Enhanced Abeta synthesis rates coupled with decreased degradative enzyme production and accumulating physical modifications that dampen proteolysis may all enhance amyloid deposit formation. Amyloid accumulation may indirectly exert the greatest pathologic effect on the brain vasculature by destroying smooth muscle cells and creating a cascade of negative impacts on cerebral blood flow. The most visible manifestation of amyloid dis-equilibrium could actually be a defense mechanism employed to avoid serious vascular wall degradation while the major toxic effects to the gray and white matter neurons are mediated by soluble oligomeric Abeta peptides with high beta-sheet content. The recognition that dynamic soluble oligomeric Abeta pools exist in AD and are correlated to disease severity led to neurotoxicity and physical conformation studies. It is now recognized that the most basic soluble Abeta peptides are stable dimers with hydrophobic regions sequestered from the aqueous environment and are capable of higher order aggregations. Time course experiments employing a modified ELISA method able to detect Abeta oligomers revealed dynamic intermolecular interactions and additional experiments physically confirmed the presence of stable amyloid multimers. Amyloid peptides that are rich in beta-sheet structure are capable of creating toxic membrane ion channels and a capacity to self-assemble as annular structures was confirmed in vitro using atomic force microscopy. Biochemical studies have established that soluble Abeta peptides perturb metabolic processes, provoke release of deleterious reactive compounds, reduce blood flow, induce mitochondrial apoptotic toxicity and inhibit angiogenesis. While there is no question that gross amyloid deposition does contribute to AD pathology, the destructive potential now associated with soluble Abeta suggests that treatment strategies that target these molecules may be efficacious in preventing some of the devastating effects of AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/química , Fragmentos de Péptidos/química , 3-Hidroxiacil-CoA Deshidrogenasas/antagonistas & inhibidores , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Amiloide/química , Péptidos beta-Amiloides/aislamiento & purificación , Animales , Apoptosis/fisiología , Biopolímeros , Líquidos Corporales/química , Química Encefálica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Ratones Transgénicos , Microglía/fisiología , Microscopía de Fuerza Atómica , Mitocondrias/fisiología , Peso Molecular , Neovascularización Fisiológica , Fragmentos de Péptidos/aislamiento & purificación , Placa Amiloide/química , Conformación Proteica , Ratas , Solubilidad , Vacunación
10.
Biochim Biophys Acta ; 1741(1-2): 199-205, 2005 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-15882940

RESUMEN

In the AD brain, there are elevated amounts of soluble and insoluble Abeta peptides which enhance the expression of membrane bound and soluble receptor for advanced glycation end products (RAGE). The binding of soluble Abeta to soluble RAGE inhibits further aggregation of Abeta peptides, while membrane bound RAGE-Abeta interactions elicit activation of the NF-kappaB transcription factor promoting sustained chronic neuroinflammation. Atomic force microscopy observations demonstrated that the N-terminal domain of RAGE, by interacting with Abeta, is a powerful inhibitor of Abeta polymerization even at prolonged periods of incubation. Hence, the potential RAGE-Abeta structural interactions were further explored utilizing a series of computational chemistry algorithms. Our modeling suggests that a soluble dimeric RAGE assembly creates a positively charged well into which the negative charges of the N-terminal domain of dimeric Abeta dock.


Asunto(s)
Microscopía de Fuerza Atómica , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Dimerización , Disulfuros/química , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Inmunoglobulina G/inmunología , Modelos Moleculares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Solubilidad
11.
Neurobiol Dis ; 18(1): 75-82, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15649697

RESUMEN

Amyloid-beta1-42 (Abeta1-42) is crucial to Alzheimer disease (AD) pathogenesis but the conformation of the toxic Abeta species remains uncertain. AD risk is increased by apolipoprotein E4 (apoE4) and decreased by apoE2 compared with the apoE3 isoform, but whether inheritance of apoE4 represents a gain of negative or a loss of protective function is also unresolved. Using hippocampal slices from apoE knockout (apoE-KO) and human apoE2, E3, and E4 targeted replacement (apoE-TR) mice, we found that oligomeric Abeta1-42 inhibited long-term potentiation (LTP) with a hierarchy of susceptibility mirroring clinical AD risk (apoE4-TR > apoE3-TR = apoE-KO > apoE2-TR), and that comparable doses of unaggregated Abeta1-42 did not affect LTP. These data provide a novel link among apoE isoform, Abeta1-42, and a functional cellular model of memory. In this model, apoE4 confers a gain of negative function synergistic with Abeta1-42, apoE2 is protective, and the apoE-Abeta interaction is specific to oligomeric Abeta1-42.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Apolipoproteínas E/antagonistas & inhibidores , Hipocampo/metabolismo , Potenciación a Largo Plazo/genética , Fragmentos de Péptidos/farmacología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteína E2 , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Peso Molecular , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Técnicas de Cultivo de Órganos , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
12.
J Mol Neurosci ; 23(3): 235-46, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15181252

RESUMEN

Abnormalities in the processing of amyloid precursor protein to amyloid-beta (Abeta) are causal factors, and the presence of the epsilon4 allele of apolipoprotein E (apoE) is the primary risk factor for Alzheimer's disease (AD). Based, at least in part, on these genetics, the potential structural and functional interactions between these two proteins are the focus of our research. To understand the nature of the physical interactions between apoE and Abeta, we initially utilized gel-shift assays to demonstrate that native apoE2 and E3 (associated with lipid particles) form an SDS-stable complex with Abeta that is more abundant than the apoE4:Abeta complex. We further demonstrated that exogenous apoE3 but not E4 prevents Abeta-induced neurotoxicity by a process that requires apoE receptors. In addition, both exogenous apoE3 and E4 prevent Abeta-induced, glial-mediated inflammation, also via a process that requires apoE receptors. These functional effects all occur at a molar ratio of apoE to Abeta of 1:30. Because the biological activities for both apoE and Abeta are profoundly influenced by their isoform and conformation, respectively, we further investigated the idea that apoE3 and E4 differentially interact with particular aggregation species of Abeta1-42. Our overall hypothesis is that apoE has two general functions in relation to Abeta. First, apoE interacts with oligomeric Abeta via an apoE receptor-mediated process to inhibit neurotoxicity and neuroinflammation (apoE3 > apoE4) a process possibly related to binding and clearance of apoE3:oligomer complexes. Second, apoE facilitates the deposition of Abeta as amyloid (apoE4 > apoE3). We will continue to investigate the effect of apoE isoform and Abeta conformation on the structural and functional interactions between these two proteins in relation to the pathogenesis of AD.


Asunto(s)
Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/epidemiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Animales , Humanos , Modelos Animales , Neurofibrillas/patología , Neurofibrillas/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/química , Conformación Proteica , Factores de Riesgo
13.
J Biol Chem ; 278(13): 11612-22, 2003 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-12499373

RESUMEN

Extensive research causally links amyloid-beta peptide (A beta) to Alzheimer's disease, although the pathologically relevant A beta conformation remains unclear. A beta spontaneously aggregates into the fibrils that deposit in senile plaques. However, recent in vivo and in vitro reports describe a potent biological activity for oligomeric assemblies of A beta. To consistently prepare in vitro oligomeric and fibrillar forms of A beta 1-42, a detailed knowledge of how solution parameters influence structure is required. This manuscript represents the first study using a single chemically and structurally homogeneous unaggregated starting material to demonstrate that the formation of oligomers, fibrils, and fibrillar aggregates is determined by time, concentration, temperature, pH, ionic strength, and A beta species. We recently reported that oligomers inhibit neuronal viability 10-fold more than fibrils and approximately 40-fold more than unaggregated peptide, with oligomeric A beta 1-42-induced neurotoxicity significant at 10 nm. In addition, we were able to differentiate by structure and neurotoxic activity wild-type A beta1-42 from isoforms containing familial mutations (Dahlgren, K. N., Manelli, A. M., Stine, W. B., Jr., Baker, L. K., Krafft, G. A., and LaDu, M. J. (2002) J. Biol. Chem. 277, 32046-32053). Understanding the biological role of specific A beta conformations may define the link between A beta and Alzheimer's disease, re-focusing therapeutic approaches by identifying the pernicious species of A beta ultimately responsible for the cognitive dysfunction that defines the disease.


Asunto(s)
Péptidos beta-Amiloides/química , Biopolímeros/química , Fragmentos de Péptidos/química , Western Blotting , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Solubilidad
14.
J Biol Chem ; 277(35): 32046-53, 2002 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-12058030

RESUMEN

Genetic evidence predicts a causative role for amyloid-beta (A beta) in Alzheimer's disease. Recent debate has focused on whether fibrils (amyloid) or soluble oligomers of A beta are the active species that contribute to neurodegeneration and dementia. We developed two aggregation protocols for the consistent production of stable oligomeric or fibrillar preparations of A beta-(1-42). Here we report that oligomers inhibit neuronal viability 10-fold more than fibrils and approximately 40-fold more than unaggregated peptide, with oligomeric A beta-(1-42)-induced inhibition significant at 10 nm. Under A beta-(1-42) oligomer- and fibril-forming conditions, A beta-(1-40) remains predominantly as unassembled monomer and had significantly less effect on neuronal viability than preparations of A beta-(1-42). We applied the aggregation protocols developed for wild type A beta-(1-42) to A beta-(1-42) with the Dutch (E22Q) or Arctic (E22G) mutations. Oligomeric preparations of the mutations exhibited extensive protofibril and fibril formation, respectively, but were not consistently different from wild type A beta-(1-42) in terms of inhibition of neuronal viability. However, fibrillar preparations of the mutants appeared larger and induced significantly more inhibition of neuronal viability than wild type A beta-(1-42) fibril preparations. These data demonstrate that protocols developed to produce oligomeric and fibrillar A beta-(1-42) are useful in distinguishing the structural and functional differences between A beta-(1-42) and A beta-(1-40) and genetic mutations of A beta-(1-42).


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/fisiología , Supervivencia Celular/fisiología , Neuronas/citología , Fragmentos de Péptidos/química , Enfermedad de Alzheimer , Humanos , Microscopía de Fuerza Atómica , Neuroblastoma , Células Tumorales Cultivadas
15.
Brain Res ; 924(2): 133-40, 2002 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-11750898

RESUMEN

The dementia in Alzheimer disease (AD) is usually attributed to widespread neuronal loss in conjunction with the pathologic hallmarks of intracellular neurofibrillary tangles and extracellular plaques containing amyloid (A beta) in fibrillar form. Recently it has been demonstrated that non-fibrillar assemblies of A beta possess electrophysiologic activity, with the corollary that they may produce dementia by disrupting neuronal signaling prior to cell death. We therefore examined the effects of soluble oligomers of A beta(1-42) on long-term potentiation (LTP) and long-term depression (LTD), two cellular models of memory, in the dentate gyrus of rat hippocampal slices. Compared with vehicle controls, slices pre-incubated 60 min in the presence of A beta-derived diffusible ligands (ADDLs) showed no differences in threshold intensity to evoke a synaptic response, slope of field excitatory post-synaptic potentials (EPSPs), or the input/output function. Tetanus-induced LTP and reversal of LTD were strongly inhibited in ADDLs-treated slices whereas LTD was unaffected. These data suggest that soluble non-fibrillar amyloid may contribute to the pathogenesis of AD both by impairing LTP/memory formation at the cellular level and by creating 'neuroplasticity imbalance' manifested by unopposed LTD in the setting of impaired capacity for neural repair via reversal of LTD or LTP.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Giro Dentado/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Animales , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Masculino , Plasticidad Neuronal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Solubilidad , Sinapsis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...