Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Acta Neuropathol Commun ; 11(1): 181, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37964332

RESUMEN

Tau seed amplification assays (SAAs) directly measure the seeding activity of tau and would therefore be ideal biomarkers for clinical trials targeting seeding-competent tau in Alzheimer's disease (AD). However, the precise relationship between tau seeding measured by SAA and the levels of pathological forms of tau in the AD brain remains unknown. We developed a new tau SAA based on full-length 0N3R tau with sensitivity in the low fg/ml range and used it to characterize 103 brain samples from three independent cohorts. Tau seeding clearly discriminated between AD and control brain samples. Interestingly, seeding was absent in Progressive Supranuclear Palsy (PSP) putamen, suggesting that our tau SAA did not amplify 4R tau aggregates from PSP brain. The specificity of our tau SAA for AD brain was further supported by analysis of matched hippocampus and cerebellum samples. While seeding was detected in hippocampus from Braak stages I-II, no seeding was present in AD cerebellum that is devoid of tau inclusions. Analysis of 40 middle frontal gyrus samples encompassing all Braak stages showed that tau SAA seeding activity gradually increased with Braak stage. This relationship between seeding activity and the presence of tau inclusions in AD brain was further supported by robust correlations between tau SAA results and the levels of phosphorylated tau212/214, phosphorylated tau181, aggregated tau, and sarkosyl-insoluble tau. Strikingly, we detected tau seeding in the middle frontal gyrus already at Braak stage II-III, suggesting that tau SAA can detect tau pathology earlier than conventional immunohistochemical staining. In conclusion, our data suggest a quantitative relationship between tau seeding activity and pathological forms of tau in the human brain and provides an important basis for further development of tau SAA for accessible human samples.


Asunto(s)
Enfermedad de Alzheimer , Parálisis Supranuclear Progresiva , Humanos , Enfermedad de Alzheimer/patología , Proteínas tau/metabolismo , Encéfalo/patología , Parálisis Supranuclear Progresiva/patología , Cerebelo/patología
3.
Transl Stroke Res ; 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37326791

RESUMEN

Repulsive guidance molecule A (RGMa) is an inhibitor of neuronal growth and survival which is upregulated in the damaged central nervous system following acute spinal cord injury (SCI), traumatic brain injury, acute ischemic stroke (AIS), and other neuropathological conditions. Neutralization of RGMa is neuroprotective and promotes neuroplasticity in several preclinical models of neurodegeneration and injury including multiple sclerosis, AIS, and SCI. Given the limitations of current treatments for AIS due to narrow time windows to intervention (TTI), and restrictive patient selection criteria, there is significant unmet need for therapeutic agents that enable tissue survival and repair following acute ischemic damage for a broader population of stroke patients. In this preclinical study, we evaluated whether elezanumab, a human anti-RGMa monoclonal antibody, could improve neuromotor function and modulate neuroinflammatory cell activation following AIS with delayed intervention times up to 24 h using a rabbit embolic permanent middle cerebral artery occlusion model (pMCAO). In two replicate 28-day pMCAO studies, weekly intravenous infusions of elezanumab, over a range of doses and TTIs of 6 and 24 h after stroke, significantly improved neuromotor function in both pMCAO studies when first administered 6 h after stroke. All elezanumab treatment groups, including the 24 h TTI group, had significantly less neuroinflammation as assessed by microglial and astrocyte activation. The novel mechanism of action and potential for expanding TTI in human AIS make elezanumab distinct from current acute reperfusion therapies, and support evaluation in clinical trials of acute CNS damage to determine optimal dose and TTI in humans. A: Ramified/resting astrocytes and microglia in a normal, uninjured rabbit brain. B: Rabbit pMCAO brain illustrating lesion on right side of brain (red), surrounded by penumbra (pink) during acute phase post stroke, with minimal injury to left brain hemisphere. Penumbra characterized by activated astrocytes and microglia (region in crosshair within circle), with upregulation of free and bound RGMa. C: Elezanumab binds to both free and bound RGMa, preventing full activation of astrocytes and microglia. D: Elezanumab is efficacious in rabbit pMCAO with a 4 × larger TTI window vs. tPA (6 vs. 1.5 h, respectively). In human AIS, tPA is approved for a TTI of 3-4.5 h. Elezanumab is currently being evaluated in a clinical Ph2 study of AIS to determine the optimal dose and TTI (NCT04309474).

4.
MAbs ; 13(1): 1887628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33596779

RESUMEN

ABT-736 is a humanized monoclonal antibody generated to target a specific conformation of the amyloid-beta (Aß) protein oligomer. Development of ABT-736 for Alzheimer's disease was discontinued due to severe adverse effects (AEs) observed in cynomolgus monkey toxicity studies. The acute nature of AEs observed only at the highest doses suggested potential binding of ABT-736 to an abundant plasma protein. Follow-up investigations indicated polyspecificity of ABT-736, including unintended high-affinity binding to monkey and human plasma protein platelet factor 4 (PF-4), known to be involved in heparin-induced thrombocytopenia (HIT) in humans. The chronic AEs observed at the lower doses after repeat administration in monkeys were consistent with HIT pathology. Screening for a backup antibody revealed that ABT-736 possessed additional unintended binding characteristics to other, unknown factors. A subsequently implemented screening funnel focused on nonspecific binding led to the identification of h4D10, a high-affinity Aß oligomer binding antibody that did not bind PF-4 or other unintended targets and had no AEs in vivo. This strengthened the hypothesis that ABT-736 toxicity was not Aß target-related, but instead was the consequence of polyspecificity including PF-4 binding, which likely mediated the acute and chronic AEs and the HIT-like pathology. In conclusion, thorough screening of antibody candidates for nonspecific interactions with unrelated molecules at early stages of discovery can eliminate candidates with polyspecificity and reduce potential for toxicity caused by off-target binding.


Asunto(s)
Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/toxicidad , Plaquetas/efectos de los fármacos , Inmunidad Heteróloga , Factor Plaquetario 4/antagonistas & inhibidores , Púrpura Trombocitopénica Idiopática/inducido químicamente , Vacunas contra el Alzheimer/farmacocinética , Vacunas contra el Alzheimer/toxicidad , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacocinética , Especificidad de Anticuerpos , Plaquetas/inmunología , Plaquetas/metabolismo , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Nivel sin Efectos Adversos Observados , Activación Plaquetaria/efectos de los fármacos , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/inmunología , Medición de Riesgo , Factores de Tiempo , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Crónica
5.
Acta Neuropathol Commun ; 7(1): 177, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31722749

RESUMEN

Intraneuronal insoluble inclusions made of Tau protein are neuropathological hallmarks of Alzheimer Disease (AD). Cleavage of Tau by legumain (LGMN) has been proposed to be crucial for aggregation of Tau into fibrils. However, it remains unclear if LGMN-cleaved Tau fragments accumulate in AD Tau inclusions.Using an in vitro enzymatic assay and non-targeted mass spectrometry, we identified four putative LGMN cleavage sites at Tau residues N167-, N255-, N296- and N368. Cleavage at N368 generates variously sized N368-Tau fragments that are aggregation prone in the Thioflavin T assay in vitro. N368-cleaved Tau is not detected in the brain of legumain knockout mice, indicating that LGMN is required for Tau cleavage in the mouse brain in vivo. Using a targeted mass spectrometry method in combination with tissue fractionation and biochemical analysis, we investigated whether N368-cleaved Tau is differentially produced and aggregated in brain of AD patients and control subjects. In brain soluble extracts, despite reduced uncleaved Tau in AD, levels of N368-cleaved Tau are comparable in AD and control hippocampus, suggesting that LGMN-mediated cleavage of Tau is not altered in AD. Consistently, levels of activated, cleaved LGMN are also similar in AD and control brain extracts. To assess the potential accumulation of N368-cleaved Tau in insoluble Tau aggregates, we analyzed sarkosyl-insoluble extracts from AD and control hippocampus. Both N368-cleaved Tau and uncleaved Tau were significantly increased in AD as a consequence of pathological Tau inclusions accumulation. However, the amount of N368-cleaved Tau represented only a very minor component (< 0.1%) of insoluble Tau.Our data indicate that LGMN physiologically cleaves Tau in the mouse and human brain generating N368-cleaved Tau fragments, which remain largely soluble and are present only in low proportion in Tau insoluble aggregates compared to uncleaved Tau. This suggests that LGMN-cleaved Tau has limited role in the progressive accumulation of Tau inclusions in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Cisteína Endopeptidasas/metabolismo , Agregado de Proteínas/fisiología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Cisteína Endopeptidasas/deficiencia , Cisteína Endopeptidasas/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Proteínas tau/genética
6.
Cell Rep ; 10(11): 1887-98, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25801027

RESUMEN

Repulsive guidance molecule A (RGMa) is a potent inhibitor of neuronal regeneration and a regulator of cell death, and it plays a role in multiple sclerosis (MS). In autopsy material from progressive MS patients, RGMa was found in active and chronic lesions, as well as in normal-appearing gray and white matter, and was expressed by cellular meningeal infiltrates. Levels of soluble RGMa in the cerebrospinal fluid were decreased in progressive MS patients successfully treated with intrathecal corticosteroid triamcinolone acetonide (TCA), showing functional improvements. In vitro, RGMa monoclonal antibodies (mAbs) reversed RGMa-mediated neurite outgrowth inhibition and chemorepulsion. In animal models of CNS damage and MS, RGMa antibody stimulated regeneration and remyelination of damaged nerve fibers, accelerated functional recovery, and protected the retinal nerve fiber layer as measured by clinically relevant optic coherence tomography. These data suggest that targeting RGMa is a promising strategy to improve functional recovery in MS patients.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Regeneración Nerviosa , Proteínas del Tejido Nervioso/metabolismo , Adulto , Anciano , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Femenino , Proteínas Ligadas a GPI , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Esclerosis Múltiple/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/fisiología , Ratas , Ratas Wistar
7.
J Neural Transm (Vienna) ; 122(6): 841-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25209051

RESUMEN

Repeated intrathecal application of the sustained release steroid triamcinolone acetonide is beneficial in progressive multiple sclerosis patients. Its putative regenerative effect may involve regulation of the repulsive guidance molecule A synthesis. This protein inhibits axonal regeneration and functional recovery. Objectives were to demonstrate the efficacy of four triamcinolone applications every other day in association with repulsive guidance molecule A levels in cerebrospinal fluid. Clinical evaluation was performed at baseline and on each day after a triamcinolone administration in 25 progressive multiple sclerosis patients. Repulsive guidance molecule A concentrations were determined before each triamcinolone application by western blot analysis with quantification. Clinical scores for multiple sclerosis improved, and the maximum walking distance and speed ameliorated in 17 patients. Repulsive guidance molecule A levels declined in these responders. The remaining patients showed no prompt clinical benefit and no decrease of repulsive guidance molecule A concentrations. Decline of repulsive guidance molecule A may reflect regeneration and functional recovery by triamcinolone in progressive multiple sclerosis patients.


Asunto(s)
Inmunosupresores/uso terapéutico , Esclerosis Múltiple Crónica Progresiva/líquido cefalorraquídeo , Esclerosis Múltiple Crónica Progresiva/tratamiento farmacológico , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Triamcinolona Acetonida/uso terapéutico , Western Blotting , Prueba de Esfuerzo , Femenino , Proteínas Ligadas a GPI/líquido cefalorraquídeo , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Caminata
8.
Brain ; 137(Pt 12): 3319-26, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25281869

RESUMEN

Cognitive decline in Alzheimer's disease is attributed to loss of functional synapses, most likely caused by synaptotoxic, oligomeric forms of amyloid-ß. Many treatment options aim at reducing amyloid-ß levels in the brain, either by decreasing its production or by increasing its clearance. We quantified the effects of immunotherapy directed against oligomeric amyloid-ß in Tg2576 mice, a mouse model of familial Alzheimer's disease. Treatment of 12-month-old mice with oligomer-specific (A-887755) or conformation-unspecific (6G1) antibodies for 8 weeks did not affect fibrillar plaque density or growth. We also quantified densities of DLG4 (previously known as PSD95) expressing post-synapses and synapsin expressing presynapses immunohistochemically. We found that both pre- and post-synapses were strongly reduced in the vicinity of plaques, whereas distant from plaques, in the cortex and hippocampal CA1 field, only post-synapses were reduced. Immunotherapy alleviated this synapse loss. Synapse loss was completely abolished distant from plaques, whereas it was only attenuated in the vicinity of plaques. These results suggest that fibrillar plaques may act as reservoirs for synaptotoxic, oligomeric amyloid-ß and that sequestering oligomers suffices to counteract synaptic pathology. Therefore, cognitive function may be improved by immunotherapy even when the load of fibrillar amyloid remains unchanged.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Inmunoterapia , Placa Amiloide/patología , Sinapsis/patología , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/terapia , Animales , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Sinapsis/metabolismo
9.
MAbs ; 6(3): 577-618, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24589717

RESUMEN

The 24th Antibody Engineering & Therapeutics meeting brought together a broad range of participants who were updated on the latest advances in antibody research and development. Organized by IBC Life Sciences, the gathering is the annual meeting of The Antibody Society, which serves as the scientific sponsor. Preconference workshops on 3D modeling and delineation of clonal lineages were featured, and the conference included sessions on a wide variety of topics relevant to researchers, including systems biology; antibody deep sequencing and repertoires; the effects of antibody gene variation and usage on antibody response; directed evolution; knowledge-based design; antibodies in a complex environment; polyreactive antibodies and polyspecificity; the interface between antibody therapy and cellular immunity in cancer; antibodies in cardiometabolic medicine; antibody pharmacokinetics, distribution and off-target toxicity; optimizing antibody formats for immunotherapy; polyclonals, oligoclonals and bispecifics; antibody discovery platforms; and antibody-drug conjugates.


Asunto(s)
Anticuerpos/química , Anticuerpos/uso terapéutico , Animales , Anticuerpos/genética , Evolución Molecular Dirigida , Diseño de Fármacos , Humanos , Inmunoterapia , Neoplasias/terapia , Ingeniería de Proteínas , Sociedades Científicas , Biología de Sistemas
10.
J Neurosci ; 30(31): 10369-79, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20685980

RESUMEN

Oligomers of the beta-amyloid (Abeta) peptide have been indicated in early neuropathologic changes in Alzheimer's disease. Here, we present a synthetic Abeta(20-42) oligomer (named globulomer) with a different conformation to monomeric and fibrillar Abeta peptide, enabling the generation of highly Abeta oligomer-specific monoclonal antibodies. The globulomer-derived antibodies specifically detect oligomeric but not monomeric or fibrillar Abeta in various Abeta preparations. The globulomer-specific antibody A-887755 was able to prevent Abeta oligomer binding and dynamin cleavage in primary hippocampal neurons and to reverse globulomer-induced reduced synaptic transmission. In amyloid precursor protein (APP) transgenic mice, vaccination with Abeta globulomer and treatment with A-887755 improved novel object recognition. The cognitive improvement is likely attributable to reversing a deficit in hippocampal synaptic spine density in APP transgenic mice as observed after treatment with A-887755. Our findings demonstrate that selective reduction of Abeta oligomers by immunotherapy is sufficient to normalize cognitive behavior and synaptic deficits in APP transgenic mice.


Asunto(s)
Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Anticuerpos Monoclonales/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/inmunología , Análisis de Varianza , Animales , Anticuerpos Monoclonales/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hipocampo/citología , Hipocampo/inmunología , Inmunoprecipitación , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/inmunología , Ratas , Ratas Wistar , Reconocimiento en Psicología
11.
Biochemistry ; 48(9): 1870-7, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19216516

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder that is linked to the presence of amyloid beta-peptides that can form insoluble fibrils or soluble oligomeric assemblies. Soluble forms are present in the brains and tissues of Alzheimer's patients, and their presence correlates with disease progression. Long-lived soluble forms can be generated in vitro by using small amounts of aliphatic hydrocarbon chains of detergents or fatty acids in preparations of amyloid beta-peptides. Using NMR, we have characterized soluble oligomers of Abeta preglobulomer and globulomer that are stable and alter synaptic activity. The NMR data indicate that these soluble forms have a mixed parallel and antiparallel beta-sheet structure that is different from fibrils which contain only parallel beta-sheets. Using the structural data, we engineered a disulfide bond into the soluble Abeta globulomer to give a "new" soluble antigen that is stable, homogeneous, and binds with the same affinity to selective antibodies as the parent wt globulomer.


Asunto(s)
Péptidos beta-Amiloides/química , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Secuencia de Aminoácidos , Amiloide/química , Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Microscopía de Fuerza Atómica , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Multimerización de Proteína , Solubilidad
12.
Neurobiol Dis ; 30(2): 212-20, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18353662

RESUMEN

Soluble A beta-oligomers are currently discussed as the major causative species for the development of Alzheimer's disease (AD). Consequently, the beta-amyloid cascade hypothesis was extended by A beta-oligomers and their central neuropathogenic role in AD. However, the molecular structure of A beta-oligomers and their relation to amyloid fibril formation remains elusive. Previously we demonstrated that incubation of A beta(1-42) with SDS or fatty acids induces the formation of a homogeneous globular A beta-oligomer termed A beta-globulomer. In this study we investigated the role of A beta-globulomers in the aggregation pathway of A beta-peptide. We used in vitro assays such as thioflavin-T binding and aggregation inhibitors like Congo red to reveal that A beta-peptide in its A beta-globulomer conformation is a structural entity which is independent from amyloid fibril formation. In addition, cellular Alzheimer's-like plaque forming assays show the resistance of A beta-globulomers to deposition as amyloid plaques. We hypothesize that a conformational switch of A beta is decisive for either fibril formation or alternatively and independently A beta-globulomer formation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Transducción de Señal/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/química , Amiloide/fisiología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/fisiología , Animales , Astrocitos/química , Astrocitos/metabolismo , Astrocitos/patología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ovillos Neurofibrilares/química , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/fisiología , Placa Amiloide/química , Conformación Proteica
13.
J Neurosci ; 28(4): 788-97, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18216187

RESUMEN

Abnormal accumulation of soluble oligomers of amyloid beta (Abeta) is believed to cause malfunctioning of neurons in Alzheimer's disease. It has been shown that Abeta oligomers impair synaptic plasticity, thereby altering the ability of the neuron to store information. We examined the underlying cellular mechanism of Abeta oligomer-induced synaptic modifications by using a recently described stable oligomeric Abeta preparation called "Abeta(1-42) globulomer." Synthetically prepared Abeta(1-42) globulomer has been shown to localize to neurons and impairs long-term potentiation (Barghorn et al., 2005). Here, we demonstrate that Abeta(1-42) globulomer does not affect intrinsic neuronal properties, as assessed by measuring input resistance and discharge characteristics, excluding an unspecific alteration of membrane properties. We provide evidence that Abeta(1-42) globulomer, at concentrations as low as 8 nM, specifically suppresses spontaneous synaptic activity resulting from a reduction of vesicular release at terminals of both GABAergic and glutamatergic synapses. EPSCs and IPSCs were primarily unaffected. A detailed search for the precise molecular target of Abeta(1-42) globulomer revealed a specific inhibition of presynaptic P/Q calcium currents, whereas other voltage-activated calcium currents remained unaltered. Because intact P/Q calcium currents are needed for synaptic plasticity, the disruption of such currents by Abeta(1-42) globulomer may cause deficits in cellular mechanisms of information storage in brains of Alzheimer's disease patients. The inhibitory effect of Abeta(1-42) globulomer on synaptic vesicle release could be reversed by roscovitine, a specific enhancer of P/Q currents. Selective enhancement of the P/Q calcium current may provide a promising strategy in the treatment of Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/química , Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo P/metabolismo , Canales de Calcio Tipo Q/metabolismo , Inhibición Neural/fisiología , Fragmentos de Péptidos/química , Transmisión Sináptica/fisiología , Péptidos beta-Amiloides/fisiología , Animales , Células Cultivadas , Ácido Glutámico/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Fragmentos de Péptidos/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
14.
Biochemistry ; 45(20): 6446-57, 2006 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-16700555

RESUMEN

In Alzheimer's disease and frontotemporal dementias the microtubule-associated protein tau forms intracellular paired helical filaments (PHFs). The filaments formed in vivo consist mainly of full-length molecules of the six different isoforms present in adult brain. The substructure of the PHF core is still elusive. Here we applied scanning transmission electron microscopy (STEM) and limited proteolysis to probe the mass distribution of PHFs and their surface exposure. Tau filaments assembled from the three repeat domain have a mass per length (MPL) of approximately 60 kDa/nm and filaments from full-length tau (htau40DeltaK280 mutant) have approximately 160 kDa/nm, compared with approximately 130 kDa/nm for PHFs from Alzheimer's brain. Polyanionic cofactors such as heparin accelerate assembly but are not incorporated into PHFs. Limited proteolysis combined with N-terminal sequencing and mass spectrometry of fragments reveals a protease-sensitive N-terminal half and semiresistant PHF core starting in the first repeat and reaching to the C-terminus of tau. Continued proteolysis leads to a fragment starting at the end of the first repeat and ending in the fourth repeat. PHFs from tau isoforms with four repeats revealed an additional cleavage site within the middle of the second repeat. Probing the PHFs with antibodies detecting epitopes either over longer stretches in the C-terminal half of tau or in the fourth repeat revealed that they grow in a polar manner. These data describe the physical parameters of the PHFs and enabled us to build a model of the molecular arrangement within the filamentous structures.


Asunto(s)
Proteínas tau/química , Proteínas tau/ultraestructura , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Secuencia de Aminoácidos , Encéfalo/citología , Encéfalo/metabolismo , Humanos , Inmunohistoquímica , Microscopía Electrónica de Transmisión de Rastreo , Microtúbulos/química , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Modelos Biológicos , Modelos Genéticos , Datos de Secuencia Molecular , Peso Molecular , Ovillos Neurofibrilares/química , Ovillos Neurofibrilares/metabolismo , Péptido Hidrolasas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas tau/metabolismo
15.
J Neurochem ; 95(3): 834-47, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16135089

RESUMEN

Amyloid beta-peptide (Abeta)(1-42) oligomers have recently been discussed as intermediate toxic species in Alzheimer's disease (AD) pathology. Here we describe a new and highly stable Abeta(1-42) oligomer species which can easily be prepared in vitro and is present in the brains of patients with AD and Abeta(1-42)-overproducing transgenic mice. Physicochemical characterization reveals a pure, highly water-soluble globular 60-kDa oligomer which we named 'Abeta(1-42) globulomer'. Our data indicate that Abeta(1-42) globulomer is a persistent structural entity formed independently of the fibrillar aggregation pathway. It is a potent antigen in mice and rabbits eliciting generation of Abeta(1-42) globulomer-specific antibodies that do not cross-react with amyloid precursor protein, Abeta(1-40) and Abeta(1-42) monomers and Abeta fibrils. Abeta(1-42) globulomer binds specifically to dendritic processes of neurons but not glia in hippocampal cell cultures and completely blocks long-term potentiation in rat hippocampal slices. Our data suggest that Abeta(1-42) globulomer represents a basic pathogenic structural principle also present to a minor extent in previously described oligomer preparations and that its formation is an early pathological event in AD. Selective neutralization of the Abeta globulomer structure epitope is expected to have a high potential for treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos , Especificidad de Anticuerpos , Células Cultivadas , Epítopos/química , Epítopos/inmunología , Epítopos/metabolismo , Ácidos Grasos , Hipocampo/citología , Humanos , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Conformación Proteica , Conejos , Ratas , Ratas Sprague-Dawley , Solubilidad , Agua/metabolismo
16.
Methods Mol Biol ; 299: 35-51, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15980594

RESUMEN

The tau protein is a neuronal microtubule-associated protein. Apart of its physiological function-the binding to and stabilization of microtubules-tau is found in Alzheimer's disease brain as insoluble fibers, the so-called "paired helical filaments" (PHFs). Investigating the fundamentals of tau polymerization is indispensable for identifying inhibitory conditions or compounds preventing PHF formation, which may slow down or even stop the degeneration of neurons in Alzheimer's disease. In this chapter, we describe the methods necessary for studying the characteristics of tau polymerization to PHFs. These include: a purification protocol for recombinantly expressed tau; a general method for the polyanion induced polymerization of tau to PHFs; the quantitation of PHFs by a fluorescence-based assay; the imaging and verification of PHFs by negative stain transmission electron microscopy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Proteínas tau/aislamiento & purificación , Amiloide/análisis , Amiloide/síntesis química , Amiloide/química , Benzotiazoles , Clonación Molecular/métodos , Humanos , Microscopía Electrónica/métodos , Isoformas de Proteínas/genética , Isoformas de Proteínas/aislamiento & purificación , Espectrometría de Fluorescencia/métodos , Tiazoles , Proteínas tau/genética
17.
Biochim Biophys Acta ; 1739(2-3): 158-66, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15615635

RESUMEN

The abnormal aggregation of the microtubule associated protein tau into paired helical filaments (PHFs) is one the hallmarks of Alzheimer's disease. The soluble protein is one of the longest natively unfolded proteins, lacking significant amounts of secondary structure over a sequence of 441 amino acids in the longest isoform. Furthermore, the unfolded character is consistent with some notable features of the protein like stability towards heat and acid treatment. It is still unclear how these characteristics support the physiological function of binding to and stabilization of microtubules. We review here some recent studies on how an unfolded protein such as tau can adopt beta-structure, which then leads to the highly ordered morphology of the PHFs. The core sequence for both microtubule binding and PHF formation is the microtubule binding domain containing three or four repeats. This region alone is sufficient for PHF formation and mostly unfolded in the soluble state. A search for sequence motifs within this region crucial for PHF building revealed two hexapeptides in the second and the third repeat. Some of the genetically linked cases of FTDP-17 show missense mutations in or adjacent to these hexapeptide motifs. Proteins containing the P301L and the DeltaK280 mutations exhibit accelerated aggregation. The importance of the two hexapeptides stems from their capacity to undergo a conformational change from a random coil to a beta sheet structure. The increase of beta sheet structure is a typical feature of an amyloidogenic protein and is the basis of other characteristics like a decreased sensitivity towards proteolytic degradation and Congo red binding. PHFs aggregated in vitro and in vivo contain beta-sheet structure, as judged by circular dichroism (CD) spectroscopy, Fourier transform infrared (FTIR) spectroscopy and X-ray diffraction.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas tau/química , Secuencia de Aminoácidos , Química Encefálica , Dicroismo Circular , Humanos , Modelos Químicos , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Espectroscopía Infrarroja por Transformada de Fourier , Difracción de Rayos X
18.
Biochemistry ; 43(6): 1694-703, 2004 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-14769047

RESUMEN

Tau protein, a neuronal microtubule-associated protein, forms insoluble fibers ("paired helical filaments") in Alzheimer's disease and other tauopathies. Conflicting views on the structure of the fibers have been proposed recently, ranging from mainly alpha-helical structure to mainly beta-sheet, or a mixture of mostly random coil and beta-sheet. We have addressed this issue by studying tau fibers immunopurified from Alzheimer brain tissue by a conformation-specific antibody and comparing them with fibers reassembled from recombinant tau or tau constructs in vitro, using a combination of electron microscopy and spectroscopic methods. Brain-derived fibers and reassembled fibers both exhibit a typical twisted appearance when examined by electron microscopy. The soluble tau protein is a natively unfolded protein dominated by random coil structure, whereas Alzheimer PHFs and reassembled fibers show a shift toward an increase in the level of beta-structure. The results support a model in which the repeat domain of tau (which lies within the core of PHFs) adopts an increasing level of beta-structure during aggregation, whereas the N- and C-terminal domains projecting away from the PHF core are mostly random coil.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Química Encefálica , Proteínas de Neurofilamentos/química , Proteínas Recombinantes/química , Proteínas tau/química , Enfermedad de Alzheimer/patología , Amiloide/química , Western Blotting , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Humanos , Ovillos Neurofibrilares/química , Ovillos Neurofibrilares/ultraestructura , Proteínas de Neurofilamentos/ultraestructura , Isoformas de Proteínas/química , Isoformas de Proteínas/aislamiento & purificación , Isoformas de Proteínas/ultraestructura , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/ultraestructura , Espectroscopía Infrarroja por Transformada de Fourier , Proteínas tau/aislamiento & purificación , Proteínas tau/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA