Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 10868, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740836

ABSTRACT

Therapeutic antibodies have been developed to target amyloid-beta (Aß), and some of these slow the progression of Alzheimer's disease (AD). However, they can also cause adverse events known as amyloid-related imaging abnormalities with edema (ARIA-E). We investigated therapeutic Aß antibody binding to cerebral amyloid angiopathy (CAA) fibrils isolated from human leptomeningeal tissue to study whether this related to the ARIA-E frequencies previously reported by clinical trials. The binding of Aß antibodies to CAA Aß fibrils was evaluated in vitro using immunoprecipitation, surface plasmon resonance, and direct binding assay. Marked differences in Aß antibody binding to CAA fibrils were observed. Solanezumab and crenezumab showed negligible CAA fibril binding and these antibodies have no reported ARIA-E cases. Lecanemab showed a low binding to CAA fibrils, consistent with its relatively low ARIA-E frequency of 12.6%, while aducanumab, bapineuzumab, and gantenerumab all showed higher binding to CAA fibrils and substantially higher ARIA-E frequencies (25-35%). An ARIA-E frequency of 24% was reported for donanemab, and its binding to CAA fibrils correlated with the amount of pyroglutamate-modified Aß present. The findings of this study support the proposal that Aß antibody-CAA interactions may relate to the ARIA-E frequency observed in patients treated with Aß-based immunotherapies.


Subject(s)
Amyloid beta-Peptides , Cerebral Amyloid Angiopathy , Humans , Cerebral Amyloid Angiopathy/immunology , Cerebral Amyloid Angiopathy/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Protein Binding , Amyloid/metabolism , Amyloid/immunology , Surface Plasmon Resonance
2.
J Fish Biol ; 104(3): 807-824, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37823583

ABSTRACT

Thiamin is an essential water-soluble B vitamin known for its wide range of metabolic functions and antioxidant properties. Over the past decades, reproductive failures induced by thiamin deficiency have been observed in several salmonid species worldwide, but it is unclear why this micronutrient deficiency arises. Few studies have compared thiamin concentrations in systems of salmonid populations with or without documented thiamin deficiency. Moreover, it is not well known whether and how thiamin concentration changes during the marine feeding phase and the spawning migration. Therefore, samples of Atlantic salmon (Salmo salar) were collected when actively feeding in the open Baltic Sea, after the sea migration to natal rivers, after river migration, and during the spawning period. To compare populations of Baltic salmon with systems without documented thiamin deficiency, a population of landlocked salmon located in Lake Vänern (Sweden) was sampled as well as salmon from Norwegian rivers draining into the North Atlantic Ocean. Results showed the highest mean thiamin concentrations in Lake Vänern salmon, followed by North Atlantic, and the lowest in Baltic populations. Therefore, salmon in the Baltic Sea seem to be consistently more constrained by thiamin than those in other systems. Condition factor and body length had little to no effect on thiamin concentrations in all systems, suggesting that there is no relation between the body condition of salmon and thiamin deficiency. In our large spatiotemporal comparison of salmon populations, thiamin concentrations declined toward spawning in all studied systems, suggesting that the reduction in thiamin concentration arises as a natural consequence of starvation rather than to be related to thiamin deficiency in the system. These results suggest that factors affecting accumulation during the marine feeding phase are key for understanding the thiamin deficiency in salmonids.


Subject(s)
Salmo salar , Thiamine , Animals , Thiamine/metabolism , Salmo salar/metabolism , Life Cycle Stages , Oceans and Seas , Atlantic Ocean , Rivers
3.
Cell Biosci ; 13(1): 142, 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37542303

ABSTRACT

BACKGROUND: Standard neuropathologic analysis of Alzheimer's brain relies on traditional fluorescence microscopy, which suffers from limited spatial resolution due to light diffraction. As a result, it fails to reveal intricate details of amyloid plaques. While electron microscopy (EM) offers higher resolution, its extensive sample preparation, involving fixation, dehydration, embedding, and sectioning, can introduce artifacts and distortions in the complex brain tissue. Moreover, EM lacks molecular specificity and has limited field of view and imaging depth. RESULTS: In our study, we employed super-resolution Stimulated Emission Depletion (STED) microscopy in conjunction with the anti-human APP recombinant antibody 1C3 fluorescently labelled with DyLightTM633 (1C3-DyLight633). This combination allowed us to visualize amyloidogenic aggregates in vitro and in brain sections from a 17-month-old 3×Tg-AD mouse with sub-diffraction limited spatial resolution. Remarkably, we achieved a spatial resolution of 29 nm in vitro and 62 nm in brain tissue sections, surpassing the capabilities of conventional confocal microscopy by 5-10 times. Consequently, we could discern individual fibrils within plaques, an achievement previously only possible with EM. CONCLUSIONS: The utilization of STED microscopy represents a groundbreaking advancement in the field, enabling researchers to delve into the characterization of local mechanisms that underlie Amyloid (Aß) deposition into plaques and their subsequent clearance. This unprecedented level of detail is especially crucial for comprehending the etiology of Alzheimer's disease and developing the next generation of anti-amyloid treatments. By facilitating the evaluation of drug candidates and non-pharmacological interventions aiming to reduce amyloid burden, STED microscopy emerges as an indispensable tool for driving scientific progress in Alzheimer's research.

4.
Neurotherapeutics ; 20(1): 195-206, 2023 01.
Article in English | MEDLINE | ID: mdl-36253511

ABSTRACT

Immunotherapy against amyloid-beta (Aß) is a promising option for the treatment of Alzheimer's disease (AD). Aß exists as various species, including monomers, oligomers, protofibrils, and insoluble fibrils in plaques. Oligomers and protofibrils have been shown to be toxic, and removal of these aggregates might represent an effective treatment for AD. We have characterized the binding properties of lecanemab, aducanumab, and gantenerumab to different Aß species with inhibition ELISA, immunodepletion, and surface plasmon resonance. All three antibodies bound monomers with low affinity. However, lecanemab and aducanumab had very weak binding to monomers, and gantenerumab somewhat stronger binding. Lecanemab was distinctive as it had tenfold stronger binding to protofibrils compared to fibrils. Aducanumab and gantenerumab preferred binding to fibrils over protofibrils. Our results show different binding profiles of lecanemab, aducanumab, and gantenerumab that may explain clinical results observed for these antibodies regarding both efficacy and side effects.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism
5.
Neuropharmacology ; 208: 108985, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35149134

ABSTRACT

The protein alpha-synuclein (αSYN) plays a central role in synucleinopathies such as Parkinsons's disease (PD) and multiple system atrophy (MSA). Presently, there are no selective αSYN positron emission tomography (PET) radioligands that do not also show affinity to amyloid-beta (Aß). We have previously shown that radiolabeled antibodies, engineered to enter the brain via the transferrin receptor (TfR), is a promising approach for PET imaging of intrabrain targets. In this study, we used this strategy to visualize αSYN in the living mouse brain. Five bispecific antibodies, binding to both the murine TfR and αSYN were generated and radiolabeled with iodine-125 or iodine-124. All bispecific antibodies bound to αSYN and mTfR before and after radiolabelling in an ELISA assay, and bound to brain sections prepared from αSYN overexpressing mice as well as human PD- and MSA subjects, but not control tissues in autoradiography. Brain concentrations of the bispecific antibodies were between 26 and 63 times higher than the unmodified IgG format 2 h post-injection, corresponding to about 1.5% of the injected dose per gram brain tissue. Additionally, intrastriatal αSYN fibrils were visualized with PET in an αSYN deposition mouse model with one of the bispecific antibodies, [124I]RmAbSynO2-scFv8D3. However, PET images acquired in αSYN transgenic mice with verified brain pathology injected with [124I]RmAbSynO2-scFv8D3 and [124I]RmAb48-scFv8D3 showed no increase in antibody retention compared to WT mice. Despite successful imaging of deposited extracellular αSYN using a brain-penetrating antibody-based radioligand with no cross-specificity towards Aß, this proof-of-concept study demonstrates challenges in imaging intracellular αSYN inclusions present in synucleinopathies.


Subject(s)
Antibodies, Bispecific , Multiple System Atrophy , Parkinson Disease , Synucleinopathies , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Bispecific/metabolism , Brain/metabolism , Humans , Mice , Multiple System Atrophy/metabolism , Parkinson Disease/metabolism , Positron-Emission Tomography/methods , alpha-Synuclein/metabolism
6.
Neurobiol Dis ; 161: 105543, 2021 12.
Article in English | MEDLINE | ID: mdl-34737044

ABSTRACT

A growing body of evidence suggests that aggregated α-synuclein, the major constituent of Lewy bodies, plays a key role in the pathogenesis of Parkinson's disease and related α-synucleinopathies. Immunotherapies, both active and passive, against α-synuclein have been developed and are promising novel treatment strategies for such disorders. Here, we report on the humanization and pharmacological characteristics of ABBV-0805, a monoclonal antibody that exhibits a high selectivity for human aggregated α-synuclein and very low affinity for monomers. ABBV-0805 binds to a broad spectrum of soluble aggregated α-synuclein, including small and large aggregates of different conformations. Binding of ABBV-0805 to pathological α-synuclein was demonstrated in Lewy body-positive post mortem brains of Parkinson's disease patients. The functional potency of ABBV-0805 was demonstrated in several cellular assays, including Fcγ-receptor mediated uptake of soluble aggregated α-synuclein in microglia and inhibition of neurotoxicity in primary neurons. In vivo, the murine version of ABBV-0805 (mAb47) displayed significant dose-dependent decrease of α-synuclein aggregates in brain in several mouse models, both in prophylactic and therapeutic settings. In addition, mAb47 treatment of α-synuclein transgenic mice resulted in a significantly prolonged survival. ABBV-0805 selectively targets soluble toxic α-synuclein aggregates with a picomolar affinity and demonstrates excellent in vivo efficacy. Based on the strong preclinical findings described herein, ABBV-0805 has been progressed into clinical development as a potential disease-modifying treatment for Parkinson's disease.


Subject(s)
Antibodies, Monoclonal , Parkinson Disease , Synucleinopathies , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Longevity , Mice , Mice, Transgenic , Parkinson Disease/metabolism , Parkinson Disease/therapy , Synucleinopathies/therapy , alpha-Synuclein/metabolism
7.
Transl Neurodegener ; 10(1): 38, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34579778

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) immunotherapy is a promising therapeutic strategy in the fight against Alzheimer's disease (AD). A number of monoclonal antibodies have entered clinical trials for AD. Some of them have failed due to the lack of efficacy or side-effects, two antibodies are currently in phase 3, and one has been approved by FDA. The soluble intermediate aggregated species of Aß, termed oligomers and protofibrils, are believed to be key pathogenic forms, responsible for synaptic and neuronal degeneration in AD. Therefore, antibodies that can strongly and selectively bind to these soluble intermediate aggregates are of great diagnostic and therapeutic interest. METHODS: We designed and recombinantly produced a hexavalent antibody based on mAb158, an Aß protofibril-selective antibody. The humanized version of mAb158, lecanemab (BAN2401), is currently in phase 3 clinical trials for the treatment of AD. The new designs involved recombinantly fusing single-chain fragment variables to the N-terminal ends of mAb158 antibody. Real-time interaction analysis with LigandTracer and surface plasmon resonance were used to evaluate the kinetic binding properties of the generated antibodies to Aß protofibrils. Different ELISA setups were applied to demonstrate the binding strength of the hexavalent antibody to Aß aggregates of different sizes. Finally, the ability of the antibodies to protect cells from Aß-induced effects was evaluated by MTT assay. RESULTS: Using real-time interaction analysis with LigandTracer, the hexavalent design promoted a 40-times enhanced binding with avidity to protofibrils, and most of the added binding strength was attributed to the reduced rate of dissociation. Furthermore, ELISA experiments demonstrated that the hexavalent design also had strong binding to small oligomers, while retaining weak and intermediate binding to monomers and insoluble fibrils. The hexavalent antibody also reduced cell death induced by a mixture of soluble Aß aggregates. CONCLUSION: We provide a new antibody design with increased valency to promote binding avidity to an enhanced range of sizes of Aß aggregates. This approach should be general and work for any aggregated protein or repetitive target.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal , Mice , Mice, Transgenic
8.
Sci Transl Med ; 13(606)2021 08 11.
Article in English | MEDLINE | ID: mdl-34380771

ABSTRACT

Point mutations in the amyloid precursor protein gene (APP) cause familial Alzheimer's disease (AD) by increasing generation or altering conformation of amyloid ß (Aß). Here, we describe the Uppsala APP mutation (Δ690-695), the first reported deletion causing autosomal dominant AD. Affected individuals have an age at symptom onset in their early forties and suffer from a rapidly progressing disease course. Symptoms and biomarkers are typical of AD, with the exception of normal cerebrospinal fluid (CSF) Aß42 and only slightly pathological amyloid-positron emission tomography signals. Mass spectrometry and Western blot analyses of patient CSF and media from experimental cell cultures indicate that the Uppsala APP mutation alters APP processing by increasing ß-secretase cleavage and affecting α-secretase cleavage. Furthermore, in vitro aggregation studies and analyses of patient brain tissue samples indicate that the longer form of mutated Aß, AßUpp1-42Δ19-24, accelerates the formation of fibrils with unique polymorphs and their deposition into amyloid plaques in the affected brain.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Humans
9.
Mol Cell Neurosci ; 114: 103641, 2021 07.
Article in English | MEDLINE | ID: mdl-34091073

ABSTRACT

Down syndrome (DS) is caused by trisomy of chromosome 21, which leads to a propensity to develop amyloid ß (Aß) brain pathology in early adulthood followed later by cognitive and behavioral deterioration. Characterization of the Aß pathology is important to better understand the clinical deterioration of DS individuals and to identify interventive strategies. Brain samples from people with DS and Alzheimer's disease (AD), as well as non-demented controls (NDC), were analyzed with respect to different Aß species. Immunohistochemical staining using antibodies towards Aß was also performed. Elevated levels of soluble Aß protofibrils and insoluble Aßx-40 and Aßx-42 in formic acid brain extracts, and elevated immunohistochemical staining of Aß deposits were demonstrated with the antibody BAN2401 (lecanemab) in DS and AD compared with NDC. These data and the promising data in a large phase 2 CE clinical trial with lecanemab suggest that lecanemab may have the potential to preserve cognitive capacity in DS. Lecanemab is currently in a phase 3 CE clinical trial.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Brain/metabolism , Down Syndrome/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Brain/pathology , Down Syndrome/pathology , Female , Humans , Male , Middle Aged
10.
Neurosci Lett ; 754: 135894, 2021 05 29.
Article in English | MEDLINE | ID: mdl-33848613

ABSTRACT

The major characteristics of Alzheimer's disease (AD) are amyloid plaques, consisting of aggregated beta amyloid (Aß) peptides, together with tau pathology (tangles, neuropil treads and dystrophic neurites surrounding the plaques), in the brain. Down's syndrome (DS) individuals are at increased risk to develop AD-type pathology; most DS individuals have developed substantial pathology already at the age of 40. DS individuals have an extra copy of chromosome 21, harbouring the amyloid precursor protein gene (APP). Our aim was to investigate the Aß peptide pattern in DS and AD brains to investigate differences in their amyloid deposition and aggregation, respectively. Cortical tissue from patients with DS (with amyloid pathology), sporadic AD and controls were homogenized and fractionated into TBS (water soluble) and formic acid (water insoluble) fractions. Immunoprecipitation (IP) was performed using a variety of antibodies targeting different Aß species including oligomeric Aß. Mass spectrometry was then used to evaluate the presence of Aß species in the different patient groups. A large number of Aß peptides were identified including Aß1-X, 2-X, 3-X, 4-X, 5-X, 11-X, and Aß peptides extended N terminally of the BACE1 cleavage site and ending at amino 15 in the Aß sequence APP/Aß(-X to 15), as well as peptides post-translationally modified by pyroglutamate formation. Most Aß peptides had higher abundance in AD and DS compared to controls, except the APP/Aß(-X to 15) peptides which were most abundant in DS followed by controls and AD. Furthermore, the abundancies of AßX-40 and AßX-34 were increased in DS compared with AD. Aß1-40, Aß1-42, and Aß4-42 were identified as the main constitutes of protofibrils (IP'd using mAb158) and higher relative Aß1-42 signals were obtained compared with samples IP'd with 6E10 + 4G8, indicating that the protofibrils/oligomers were enriched with peptides ending at amino acid 42. All Aß peptides found in AD were also present in DS indicating similar pathways of Aß peptide production, degradation and accumulation, except for APP/Aß(-X to 15). Likewise, the Aß peptides forming protofibrils/oligomers in both AD and DS were similar, implying the possibility that treatment with clinical benefit in sporadic AD might also be beneficial for subjects with DS.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Down Syndrome/pathology , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/analysis , Aspartic Acid Endopeptidases/metabolism , Case-Control Studies , Female , Humans , Male , Mass Spectrometry , Middle Aged , Peptide Fragments/analysis , Protein Aggregates
11.
J Parkinsons Dis ; 10(4): 1429-1442, 2020.
Article in English | MEDLINE | ID: mdl-33016895

ABSTRACT

BACKGROUND: Currently, there is no established biomarker for Parkinson's disease (PD) and easily accessible biomarkers are crucial for developing disease-modifying treatments. OBJECTIVE: To develop a novel method to quantify cerebrospinal fluid (CSF) levels of α-synuclein protofibrils (α-syn PF) and apply it to clinical cohorts of patients with PD and atypical parkinsonian disorders. METHODS: A cohort composed of 49 patients with PD, 12 with corticobasal degeneration (CBD), 22 with progressive supranuclear palsy, and 33 controls, that visited the memory clinic but had no biomarker signs of Alzheimer's disease (AD, tau<350 pg/mL, amyloid-beta 42 (Aß42)>530 pg/mL, and phosphorylated tau (p-tau)<60 pg/mL) was used in this study. The CSF samples were analyzed with the Single molecule array (Simoa) technology. Total α-synuclein (α-syn) levels were analyzed with a commercial ELISA-kit. RESULTS: The assay is specific to α-syn PF, with no cross-reactivity to monomeric α-syn, or the ß- and γ-synuclein variants. CSF α-syn PF levels were increased in PD compared with controls (62.1 and 40.4 pg/mL, respectively, p = 0.03), and CBD (62.1 and 34.2 pg/mL, respectively, p = 0.02). The accuracy of predicting PD using α-syn PF is significantly different from controls (area under the curve 0.68, p = 0.0097) with a sensitivity of 62.8% and specificity of 67.7%. Levels of total α-syn were significantly different between the PD and CBD groups (p = 0.04). CONCLUSION: The developed method specifically quantifies α-syn PF in human CSF with increased concentrations in PD, but with an overlap with asymptomatic elderly controls.


Subject(s)
Parkinson Disease/cerebrospinal fluid , Parkinsonian Disorders/cerebrospinal fluid , alpha-Synuclein/cerebrospinal fluid , Aged , Biomarkers/cerebrospinal fluid , Female , Humans , Immunoassay , Male , Middle Aged , Supranuclear Palsy, Progressive/cerebrospinal fluid
12.
Alzheimers Res Ther ; 10(1): 49, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29793530

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) immunotherapy is one of the most promising disease-modifying strategies for Alzheimer's disease (AD). Despite recent progress targeting aggregated forms of Aß, low antibody brain penetrance remains a challenge. In the present study, we used transferrin receptor (TfR)-mediated transcytosis to facilitate brain uptake of our previously developed Aß protofibril-selective mAb158, with the aim of increasing the efficacy of immunotherapy directed toward soluble Aß protofibrils. METHODS: Aß protein precursor (AßPP)-transgenic mice (tg-ArcSwe) were given a single dose of mAb158, modified for TfR-mediated transcytosis (RmAb158-scFv8D3), in comparison with an equimolar dose or a tenfold higher dose of unmodified recombinant mAb158 (RmAb158). Soluble Aß protofibrils and total Aß in the brain were measured by enzyme-linked immunosorbent assay (ELISA). Brain distribution of radiolabeled antibodies was visualized by positron emission tomography (PET) and ex vivo autoradiography. RESULTS: ELISA analysis of Tris-buffered saline brain extracts demonstrated a 40% reduction of soluble Aß protofibrils in both RmAb158-scFv8D3- and high-dose RmAb158-treated mice, whereas there was no Aß protofibril reduction in mice treated with a low dose of RmAb158. Further, ex vivo autoradiography and PET imaging revealed different brain distribution patterns of RmAb158-scFv8D3 and RmAb158, suggesting that these antibodies may affect Aß levels by different mechanisms. CONCLUSIONS: With a combination of biochemical and imaging analyses, this study demonstrates that antibodies engineered to be transported across the blood-brain barrier can be used to increase the efficacy of Aß immunotherapy. This strategy may allow for decreased antibody doses and thereby reduced side effects and treatment costs.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/therapeutic use , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Autoradiography , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Iodine Isotopes/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Positron-Emission Tomography , Protein Binding/drug effects , Protein Binding/genetics , Tomography, X-Ray Computed
13.
J Neuroinflammation ; 15(1): 98, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29592816

ABSTRACT

BACKGROUND: Currently, several amyloid beta (Aß) antibodies, including the protofibril selective antibody BAN2401, are in clinical trials. The murine version of BAN2401, mAb158, has previously been shown to lower the levels of pathogenic Aß and prevent Aß deposition in animal models of Alzheimer's disease (AD). However, the cellular mechanisms of the antibody's action remain unknown. We have recently shown that astrocytes effectively engulf Aß42 protofibrils, but store rather than degrade the ingested Aß aggregates. In a co-culture set-up, the incomplete degradation of Aß42 protofibrils by astrocytes results in increased neuronal cell death, due to the release of extracellular vesicles, containing N-truncated, neurotoxic Aß. METHODS: The aim of the present study was to investigate if the accumulation of Aß in astrocytes can be affected by the Aß protofibril selective antibody mAb158. Co-cultures of astrocytes, neurons, and oligodendrocytes, derived from embryonic mouse cortex, were exposed to Aß42 protofibrils in the presence or absence of mAb158. RESULTS: Our results demonstrate that the presence of mAb158 almost abolished Aß accumulation in astrocytes. Consequently, mAb158 treatment rescued neurons from Aß-induced cell death. CONCLUSION: Based on these findings, we conclude that astrocytes may play a central mechanistic role in anti-Aß immunotherapy.


Subject(s)
Amyloidogenic Proteins , Antibodies, Monoclonal/pharmacology , Cell Death/drug effects , Neuroglia/drug effects , Neurons/drug effects , Amyloidogenic Proteins/immunology , Amyloidogenic Proteins/metabolism , Amyloidogenic Proteins/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL
14.
Brain Pathol ; 28(4): 451-462, 2018 07.
Article in English | MEDLINE | ID: mdl-28557010

ABSTRACT

Deposition of amyloid-ß (Aß) is central to Alzheimer's disease (AD) pathogenesis and associated with progressive neurodegeneration in traumatic brain injury (TBI). We analyzed predisposing factors for Aß deposition including monomeric Aß40, Aß42 and Aß oligomers/protofibrils, Aß species with pronounced neurotoxic properties, following human TBI. Highly selective ELISAs were used to analyze N-terminally intact and truncated Aß40 and Aß42, as well as Aß oligomers/protofibrils, in human brain tissue, surgically resected from severe TBI patients (n = 12; mean age 49.5 ± 19 years) due to life-threatening brain swelling/hemorrhage within one week post-injury. The TBI tissues were compared to post-mortem AD brains (n = 5), to post-mortem tissue of neurologically intact (NI) subjects (n = 4) and to cortical biopsies obtained at surgery for idiopathic normal pressure hydrocephalus patients (iNPH; n = 4). The levels of Aß40 and Aß42 were not elevated by TBI. The levels of Aß oligomers/protofibrils in TBI were similar to those in the significantly older AD patients and increased compared to NI and iNPH controls (P < 0.05). Moreover, TBI patients carrying the AD risk genotype Apolipoprotein E epsilon3/4 (APOE ε3/4; n = 4) had increased levels of Aß oligomers/protofibrils (P < 0.05) and of both N-terminally intact and truncated Aß42 (P < 0.05) compared to APOE ε3/4-negative TBI patients (n = 8). Neuropathological analysis showed insoluble Aß aggregates (commonly referred to as Aß plaques) in three TBI patients, all of whom were APOE ε3/4 carriers. We conclude that soluble intermediary Aß aggregates form rapidly after TBI, especially among APOE ε3/4 carriers. Further research is needed to determine whether these aggregates aggravate the clinical short- and long-term outcome in TBI.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain/metabolism , Brain/pathology , Adult , Aged , Aged, 80 and over , Amyloid/metabolism , Apolipoproteins E/genetics , Female , Humans , Male , Middle Aged , Young Adult
15.
Mol Neurodegener ; 11(1): 38, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27176225

ABSTRACT

BACKGROUND: Despite the clear physical association between activated astrocytes and amyloid-ß (Aß) plaques, the importance of astrocytes and their therapeutic potential in Alzheimer's disease remain elusive. Soluble Aß aggregates, such as protofibrils, have been suggested to be responsible for the widespread neuronal cell death in Alzheimer's disease, but the mechanisms behind this remain unclear. Moreover, ineffective degradation is of great interest when it comes to the development and progression of neurodegeneration. Based on our previous results that astrocytes are extremely slow in degrading phagocytosed material, we hypothesized that astrocytes may be an important player in these processes. Hence, the aim of this study was to clarify the role of astrocytes in clearance, spreading and neuronal toxicity of Aß. RESULTS: To examine the role of astrocytes in Aß pathology, we added Aß protofibrils to a co-culture system of primary neurons and glia. Our data demonstrates that astrocytes rapidly engulf large amounts of Aß protofibrils, but then store, rather than degrade the ingested material. The incomplete digestion results in a high intracellular load of toxic, partly N-terminally truncated Aß and severe lysosomal dysfunction. Moreover, secretion of microvesicles containing N-terminally truncated Aß, induce apoptosis of cortical neurons. CONCLUSIONS: Taken together, our results suggest that astrocytes play a central role in the progression of Alzheimer's disease, by accumulating and spreading toxic Aß species.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis/physiology , Astrocytes/metabolism , Cell-Derived Microparticles/metabolism , Endosomes/metabolism , Neurons/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Blotting, Western , Cells, Cultured , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neurons/metabolism , Time-Lapse Imaging
16.
J Alzheimers Dis ; 43(2): 575-88, 2015.
Article in English | MEDLINE | ID: mdl-25096615

ABSTRACT

Amyloid-ß (Aß) immunotherapy for Alzheimer's disease (AD) has good preclinical support from transgenic mouse models and clinical data suggesting that a long-term treatment effect is possible. Soluble Aß protofibrils have been shown to exhibit neurotoxicity in vitro and in vivo, and constitute an attractive target for immunotherapy. Here, we demonstrate that the humanized antibody BAN2401 and its murine version mAb158 exhibit a strong binding preference for Aß protofibrils over Aß monomers. Further, we confirm the presence of the target by showing that both antibodies efficiently immunoprecipitate soluble Aß aggregates in human AD brain extracts. mAb158 reached the brain and reduced the brain protofibril levels by 42% in an exposure-dependent manner both after long-term and short-term treatment in tg-ArcSwe mice. Notably, a 53% reduction of protofibrils/oligomers in cerebrospinal fluid (CSF) that correlated with reduced brain protofibril levels was observed after long-term treatment, suggesting that CSF protofibrils/oligomers could be used as a potential biomarker. No change in native monomeric Aß42 could be observed in brain TBS extracts after mAb158-treatment in tg-ArcSwe mice. By confirming the specific ability of mAb158 to selectively bind and reduce soluble Aß protofibrils, with minimal binding to Aß monomers, we provide further support in favor of its position as an attractive new candidate for AD immunotherapy. BAN2401 has undergone full phase 1 development, and available data indicate a favorable safety profile in AD patients.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal/therapeutic use , Brain/metabolism , Immunologic Factors/therapeutic use , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Immunologic Factors/pharmacology , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid , Presenilin-1/genetics , Protein Binding/drug effects , Protein Binding/genetics
17.
J Alzheimers Dis ; 37(1): 29-40, 2013.
Article in English | MEDLINE | ID: mdl-23780660

ABSTRACT

Evidence suggests that amyloid-ß (Aß) protofibrils/oligomers are pathogenic agents in Alzheimer's disease (AD). Unfortunately, techniques enabling quantitative estimates of these species in patients or patient samples are still rather limited. Here we describe the in vitro and ex vivo characteristics of a new antibody-based radioactive ligand, [125I]mAb158, which binds to Aß protofibrils with high affinity. [125I]mAb158 was specifically taken up in brain of transgenic mice expressing amyloid-ß protein precursor (AßPP) as shown ex vivo. This was in contrast to [125I]mAb-Ly128 which does not bind to Aß. The uptake of intraperitoneally-administered [125I]mAb158 into the brain was age- and time-dependent, and saturable in AßPP transgenic mice with modest Aß deposition. Brain uptake was also found in young AßPP transgenic mice that were devoid of Aß deposits, suggesting that [125I]mAb158 targets soluble Aß protofibrils. The radioligand was diffusely located in the parenchyma, sometimes around senile plaques and only occasionally colocalized with cerebral amyloid angiopathy. A refined iodine-124-labeled version of mAb158 with much improved blood-brain barrier passage and a shorter plasma half-life might be useful for PET imaging of Aß protofibrils.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Antibodies, Monoclonal/metabolism , Brain/metabolism , Iodine Radioisotopes/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid/immunology , Amyloid beta-Protein Precursor/immunology , Animals , Brain/immunology , Brain/pathology , Humans , Mice , Mice, Transgenic , Protein Binding/immunology
18.
Article in English | MEDLINE | ID: mdl-21888546

ABSTRACT

31P NMR spectroscopy was used to investigate a stereochemical course of a nitrite-promoted conversion of phosphoramidate diesters into the corresponding phosphotriesters. It was found that this reaction occurred with almost complete epimerization at the phosphorus center and at the C1 atom in the amine moiety. On the basis of the 31P NMR data, a plausible mechanism for the reaction was proposed. The density functional theory calculation of the key step of the reaction, i.e., breaking of the P-N bond and formation of the P-O bond, suggested a one-step S(N)2(P) process with retention of configuration at the phosphorus center.


Subject(s)
Amides/chemistry , Magnetic Resonance Spectroscopy , Nitrites/chemistry , Phosphoric Acids/chemistry , Esters/chemistry , Models, Molecular , Molecular Conformation , Phosphorus Isotopes/chemistry , Stereoisomerism
19.
Neurobiol Dis ; 36(3): 425-34, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19703562

ABSTRACT

Human genetics link Alzheimer's disease pathogenesis to excessive accumulation of amyloid-beta (Abeta) in brain, but the symptoms do not correlate with senile plaque burden. Since soluble Abeta aggregates can cause synaptic dysfunctions and memory deficits, these species could contribute to neuronal dysfunction and dementia. Here we explored selective targeting of large soluble aggregates, Abeta protofibrils, as a new immunotherapeutic strategy. The highly protofibril-selective monoclonal antibody mAb158 inhibited in vitro fibril formation and protected cells from Abeta protofibril-induced toxicity. When the mAb158 antibody was administered for 4 months to plaque-bearing transgenic mice with both the Arctic and Swedish mutations (tg-ArcSwe), Abeta protofibril levels were lowered while measures of insoluble Abeta were unaffected. In contrast, when treatment began before the appearance of senile plaques, amyloid deposition was prevented and Abeta protofibril levels diminished. Therapeutic intervention with mAb158 was however not proven functionally beneficial, since place learning depended neither on treatment nor transgenicity. Our findings suggest that Abeta protofibrils can be selectively cleared with immunotherapy in an animal model that display highly insoluble Abeta deposits, similar to those of Alzheimer's disease brain.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Amyloid/immunology , Amyloid/metabolism , Antibodies, Monoclonal/therapeutic use , Aging , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Brain/immunology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Immunization, Passive , Kinetics , Learning , Mice , Mice, Transgenic , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Multimerization , Space Perception
20.
FEBS J ; 276(4): 995-1006, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19215301

ABSTRACT

Oligomeric assemblies of amyloid-beta (Abeta) are suggested to be central in the pathogenesis of Alzheimer's disease because levels of soluble Abeta correlate much better with the extent of cognitive dysfunctions than do senile plaque counts. Moreover, such Abeta species have been shown to be neurotoxic, to interfere with learned behavior and to inhibit the maintenance of hippocampal long-term potentiation. The tg-ArcSwe model (i.e. transgenic mice with the Arctic and Swedish Alzheimer mutations) expresses elevated levels of Abeta protofibrils in the brain, making tg-ArcSwe a highly suitable model for investigating the pathogenic role of these Abeta assemblies. In the present study, we estimated Abeta protofibril levels in the brain and cerebrospinal fluid of tg-ArcSwe mice, and also assessed their role with respect to cognitive functions. Protofibril levels, specifically measured with a sandwich ELISA, were found to be elevated in young tg-ArcSwe mice compared to several transgenic models lacking the Arctic mutation. In aged tg-ArcSwe mice with considerable plaque deposition, Abeta protofibrils were approximately 50% higher than in younger mice, whereas levels of total Abeta were exponentially increased. Young tg-ArcSwe mice showed deficits in spatial learning, and individual performances in the Morris water maze were correlated inversely with levels of Abeta protofibrils, but not with total Abeta levels. We conclude that Abeta protofibrils accumulate in an age-dependent manner in tg-ArcSwe mice, although to a far lesser extent than total Abeta. Our findings suggest that increased levels of Abeta protofibrils could result in spatial learning impairment.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/genetics , Animals , Mice , Mice, Transgenic , Mutation , Plaque, Amyloid/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...