Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Alzheimers Res Ther ; 11(1): 97, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31787113

ABSTRACT

BACKGROUND: Accumulation of amyloid ß (Aß) in the brain is proposed as a cause of Alzheimer's disease (AD), with Aß oligomers hypothesized to be the primary mediators of neurotoxicity. Crenezumab is a humanized immunoglobulin G4 monoclonal antibody that has been shown to bind to synthetic monomeric and aggregated Aß in vitro; however, less is known about the binding characteristic in vivo. In this study, we evaluated the binding patterns of crenezumab to synthetic and native forms of Aß both in vitro and in vivo. METHODS: Crenezumab was used to immunoprecipitate Aß from synthetic Aß preparations or brain homogenates from a PS2APP mouse model of AD to determine the forms of Aß that crenezumab interacts with. Following systemic dosing in PS2APP or nontransgenic control mice, immunohistochemistry was used to localize crenezumab and assess its relative distribution in the brain, compared with amyloid plaques and markers of neuritic dystrophies (BACE1; LAMP1). Pharmacodynamic correlations were performed to investigate the relationship between peripheral and central target engagement. RESULTS: In vitro, crenezumab immunoprecipitated Aß oligomers from both synthetic Aß preparations and endogenous brain homogenates from PS2APP mice. In vivo studies in the PS2APP mouse showed that crenezumab localizes to regions surrounding the periphery of amyloid plaques in addition to the hippocampal mossy fibers. These regions around the plaques are reported to be enriched in oligomeric Aß, actively incorporate soluble Aß, and contribute to Aß-induced neurotoxicity and axonal dystrophy. In addition, crenezumab did not appear to bind to the dense core region of plaques or vascular amyloid. CONCLUSIONS: Crenezumab binds to multiple forms of amyloid ß (Aß), particularly oligomeric forms, and localizes to brain areas rich in Aß oligomers, including the halo around plaques and hippocampal mossy fibers, but not to vascular Aß. These insights highlight a unique mechanism of action for crenezumab of engaging Aß oligomers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Brain/drug effects , Animals , Brain/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism , Protein Binding
2.
Br J Pharmacol ; 174(22): 4173-4185, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28859225

ABSTRACT

BACKGROUND AND PURPOSE: The potential for therapeutic antibody treatment of neurological diseases is limited by poor penetration across the blood-brain barrier. I.c.v. delivery is a promising route to the brain; however, it is unclear how efficiently antibodies delivered i.c.v. penetrate the cerebrospinal spinal fluid (CSF)-brain barrier and distribute throughout the brain parenchyma. EXPERIMENTAL APPROACH: We evaluated the pharmacokinetics and pharmacodynamics of an inhibitory monoclonal antibody against ß-secretase 1 (anti-BACE1) following continuous infusion into the left lateral ventricle of healthy adult cynomolgus monkeys. KEY RESULTS: Animals infused with anti-BACE1 i.c.v. showed a robust and sustained reduction (~70%) of CSF amyloid-ß (Aß) peptides. Antibody distribution was near uniform across the brain parenchyma, ranging from 20 to 40 nM, resulting in a ~50% reduction of Aß in the cortical parenchyma. In contrast, animals administered anti-BACE1 i.v. showed no significant change in CSF or cortical Aß levels and had a low (~0.6 nM) antibody concentration in the brain. CONCLUSION AND IMPLICATIONS: I.c.v. administration of anti-BACE1 resulted in enhanced BACE1 target engagement and inhibition, with a corresponding dramatic reduction in CNS Aß concentrations, due to enhanced brain exposure to antibody.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/pharmacokinetics , Aspartic Acid Endopeptidases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/immunology , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/cerebrospinal fluid , Aspartic Acid Endopeptidases/immunology , Brain/metabolism , Female , Infusions, Intraventricular , Macaca fascicularis
3.
Sci Transl Med ; 9(403)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28814543

ABSTRACT

Hallmarks of chronic neurodegenerative disease include progressive synaptic loss and neuronal cell death, yet the cellular pathways that underlie these processes remain largely undefined. We provide evidence that dual leucine zipper kinase (DLK) is an essential regulator of the progressive neurodegeneration that occurs in amyotrophic lateral sclerosis and Alzheimer's disease. We demonstrate that DLK/c-Jun N-terminal kinase signaling was increased in mouse models and human patients with these disorders and that genetic deletion of DLK protected against axon degeneration, neuronal loss, and functional decline in vivo. Furthermore, pharmacological inhibition of DLK activity was sufficient to attenuate the neuronal stress response and to provide functional benefit even in the presence of ongoing disease. These findings demonstrate that pathological activation of DLK is a conserved mechanism that regulates neurodegeneration and suggest that DLK inhibition may be a potential approach to treat multiple neurodegenerative diseases.


Subject(s)
Leucine Zippers , MAP Kinase Kinase Kinases/metabolism , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Signal Transduction , Alzheimer Disease/enzymology , Alzheimer Disease/pathology , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Gene Deletion , Gene Expression Regulation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Mice, Transgenic , Neuroprotection , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Spinal Cord/enzymology , Spinal Cord/pathology , Superoxide Dismutase/metabolism
4.
Sci Rep ; 7: 44249, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281673

ABSTRACT

Assessing BACE1 (ß-site APP cleaving enzyme 1) knockout mice for general health and neurological function may be useful in predicting risks associated with prolonged pharmacological BACE1 inhibition, a treatment approach currently being developed for Alzheimer's disease. To determine whether BACE1 deletion-associated effects in mice generalize to another species, we developed a novel Bace1-/- rat line using zinc-finger nuclease technology and compared Bace1-/- mice and rats with their Bace1+/+ counterparts. Lack of BACE1 was confirmed in Bace1-/- animals from both species. Removal of BACE1 affected startle magnitude, balance beam performance, pain response, and nerve myelination in both species. While both mice and rats lacking BACE1 have shown increased mortality, the increase was smaller and restricted to early developmental stages for rats. Bace1-/- mice and rats further differed in body weight, spontaneous locomotor activity, and prepulse inhibition of startle. While the effects of species and genetic background on these phenotypes remain difficult to distinguish, our findings suggest that BACE1's role in myelination and some sensorimotor functions is consistent between mice and rats and may be conserved in other species. Other phenotypes differ between these models, suggesting that some effects of BACE1 inhibition vary with the biological context (e.g. species or background strain).


Subject(s)
Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Gene Deletion , Reflex, Startle/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Body Weight/genetics , Body Weight/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Prepulse Inhibition/genetics , Prepulse Inhibition/physiology , Rats , Reflex, Startle/physiology , Species Specificity
5.
Sci Transl Med ; 6(261): 261ra154, 2014 Nov 05.
Article in English | MEDLINE | ID: mdl-25378646

ABSTRACT

Using therapeutic antibodies that need to cross the blood-brain barrier (BBB) to treat neurological disease is a difficult challenge. We have shown that bispecific antibodies with optimized binding to the transferrin receptor (TfR) that target ß-secretase (BACE1) can cross the BBB and reduce brain amyloid-ß (Aß) in mice. Can TfR enhance antibody uptake in the primate brain? We describe two humanized TfR/BACE1 bispecific antibody variants. Using a human TfR knock-in mouse, we observed that anti-TfR/BACE1 antibodies could cross the BBB and reduce brain Aß in a TfR affinity-dependent fashion. Intravenous dosing of monkeys with anti-TfR/BACE1 antibodies also reduced Aß both in cerebral spinal fluid and in brain tissue, and the degree of reduction correlated with the brain concentration of anti-TfR/BACE1 antibody. These results demonstrate that the TfR bispecific antibody platform can robustly and safely deliver therapeutic antibody across the BBB in the primate brain.


Subject(s)
Amyloid Precursor Protein Secretases/immunology , Antibodies, Bispecific/pharmacokinetics , Antigens, CD/immunology , Aspartic Acid Endopeptidases/immunology , Blood-Brain Barrier/metabolism , Capillary Permeability , Receptors, Transferrin/immunology , Administration, Intravenous , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/blood , Antibodies, Bispecific/immunology , Antibody Specificity , Antigens, CD/genetics , Antigens, CD/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Biological Transport , CHO Cells , Cricetulus , Cross Reactions , Down-Regulation , HEK293 Cells , Humans , Macaca fascicularis , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/cerebrospinal fluid , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Transfection
6.
Nat Med ; 20(12): 1452-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25419706

ABSTRACT

We have identified a rare coding mutation, T835M (rs137875858), in the UNC5C netrin receptor gene that segregated with disease in an autosomal dominant pattern in two families enriched for late-onset Alzheimer's disease and that was associated with disease across four large case-control cohorts (odds ratio = 2.15, Pmeta = 0.0095). T835M alters a conserved residue in the hinge region of UNC5C, and in vitro studies demonstrate that this mutation leads to increased cell death in human HEK293T cells and in rodent neurons. Furthermore, neurons expressing T835M UNC5C are more susceptible to cell death from multiple neurotoxic stimuli, including ß-amyloid (Aß), glutamate and staurosporine. On the basis of these data and the enriched hippocampal expression of UNC5C in the adult nervous system, we propose that one possible mechanism in which T835M UNC5C contributes to the risk of Alzheimer's disease is by increasing susceptibility to neuronal cell death, particularly in vulnerable regions of the Alzheimer's disease brain.


Subject(s)
Alzheimer Disease/genetics , Neurons/metabolism , Receptors, Cell Surface/genetics , Receptors, Nerve Growth Factor/genetics , Aged , Aged, 80 and over , Amyloid beta-Peptides , Animals , CA3 Region, Hippocampal/cytology , Cell Death/genetics , Female , Genetic Predisposition to Disease , Glutamic Acid , HEK293 Cells , Humans , Male , Mice , Netrin Receptors , Rats , Staurosporine
7.
J Biol Chem ; 289(45): 30990-1000, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25253696

ABSTRACT

Pathogenic mutations in the amyloid precursor protein (APP) gene have been described as causing early onset familial Alzheimer disease (AD). We recently identified a rare APP variant encoding an alanine-to-threonine substitution at residue 673 (A673T) that confers protection against development of AD (Jonsson, T., Atwal, J. K., Steinberg, S., Snaedal, J., Jonsson, P. V., Bjornsson, S., Stefansson, H., Sulem, P., Gudbjartsson, D., Maloney, J., Hoyte, K., Gustafson, A., Liu, Y., Lu, Y., Bhangale, T., Graham, R. R., Huttenlocher, J., Bjornsdottir, G., Andreassen, O. A., Jönsson, E. G., Palotie, A., Behrens, T. W., Magnusson, O. T., Kong, A., Thorsteinsdottir, U., Watts, R. J., and Stefansson, K. (2012) Nature 488, 96-99). The Ala-673 residue lies within the ß-secretase recognition sequence and is part of the amyloid-ß (Aß) peptide cleavage product (position 2 of Aß). We previously demonstrated that the A673T substitution makes APP a less favorable substrate for cleavage by BACE1. In follow-up studies, we confirm that A673T APP shows reduced cleavage by BACE1 in transfected mouse primary neurons and in isogenic human induced pluripotent stem cell-derived neurons. Using a biochemical approach, we show that the A673T substitution modulates the catalytic turnover rate (V(max)) of APP by the BACE1 enzyme, without affecting the affinity (K(m)) of the APP substrate for BACE1. We also show a reduced level of Aß(1-42) aggregation with A2T Aß peptides, an observation not conserved in Aß(1-40) peptides. When combined in a ratio of 1:9 Aß(1-42)/Aß(1-40) to mimic physiologically relevant mixtures, A2T retains a trend toward slowed aggregation kinetics. Microglial uptake of the mutant Aß(1-42) peptides correlated with their aggregation level. Cytotoxicity of the mutant Aß peptides was not dramatically altered. Taken together, our findings demonstrate that A673T, a protective allele of APP, reproducibly reduces amyloidogenic processing of APP and also mildly decreases Aß aggregation. These effects could together have an additive or even synergistic impact on the risk of developing AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Alleles , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , Catalysis , DNA, Complementary/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Heterozygote , Humans , Inhibitory Concentration 50 , Kinetics , Mice , Mice, Inbred C57BL , Microglia/metabolism , Mutation , Neurons/metabolism , Peptide Fragments/genetics , Protein Binding
8.
J Neurosci ; 32(39): 13439-53, 2012 Sep 26.
Article in English | MEDLINE | ID: mdl-23015435

ABSTRACT

In addition to being a hallmark of neurodegenerative disease, axon degeneration is used during development of the nervous system to prune unwanted connections. In development, axon degeneration is tightly regulated both temporally and spatially. Here, we provide evidence that degeneration cues are transduced through various kinase pathways functioning in spatially distinct compartments to regulate axon degeneration. Intriguingly, glycogen synthase kinase-3 (GSK3) acts centrally, likely modulating gene expression in the cell body to regulate distally restricted axon degeneration. Through a combination of genetic and pharmacological manipulations, including the generation of an analog-sensitive kinase allele mutant mouse for GSK3ß, we show that the ß isoform of GSK3, not the α isoform, is essential for developmental axon pruning in vitro and in vivo. Additionally, we identify the dleu2/mir15a/16-1 cluster, previously characterized as a regulator of B-cell proliferation, and the transcription factor tbx6, as likely downstream effectors of GSK3ß in axon degeneration.


Subject(s)
Axons/metabolism , Glycogen Synthase Kinase 3/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Neurons/pathology , Phosphotransferases/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Cells, Cultured , Electroporation , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/cytology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Genotype , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Degeneration/drug therapy , Nerve Degeneration/prevention & control , Nerve Growth Factor/deficiency , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Organ Culture Techniques , Phosphorylation/physiology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , Transfection , Red Fluorescent Protein
9.
Nature ; 488(7409): 96-9, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22801501

ABSTRACT

The prevalence of dementia in the Western world in people over the age of 60 has been estimated to be greater than 5%, about two-thirds of which are due to Alzheimer's disease. The age-specific prevalence of Alzheimer's disease nearly doubles every 5 years after age 65, leading to a prevalence of greater than 25% in those over the age of 90 (ref. 3). Here, to search for low-frequency variants in the amyloid-ß precursor protein (APP) gene with a significant effect on the risk of Alzheimer's disease, we studied coding variants in APP in a set of whole-genome sequence data from 1,795 Icelanders. We found a coding mutation (A673T) in the APP gene that protects against Alzheimer's disease and cognitive decline in the elderly without Alzheimer's disease. This substitution is adjacent to the aspartyl protease ß-site in APP, and results in an approximately 40% reduction in the formation of amyloidogenic peptides in vitro. The strong protective effect of the A673T substitution against Alzheimer's disease provides proof of principle for the hypothesis that reducing the ß-cleavage of APP may protect against the disease. Furthermore, as the A673T allele also protects against cognitive decline in the elderly without Alzheimer's disease, the two may be mediated through the same or similar mechanisms.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Mutation/genetics , Alleles , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/chemistry , Aspartic Acid Endopeptidases/metabolism , Cognition/physiology , Cognition Disorders/prevention & control , Genetic Predisposition to Disease , HEK293 Cells , Humans , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism
10.
J Neurosci ; 32(28): 9677-89, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22787053

ABSTRACT

Passive immunization against ß-amyloid (Aß) has become an increasingly desirable strategy as a therapeutic treatment for Alzheimer's disease (AD). However, traditional passive immunization approaches carry the risk of Fcγ receptor-mediated overactivation of microglial cells, which may contribute to an inappropriate proinflammatory response leading to vasogenic edema and cerebral microhemorrhage. Here, we describe the generation of a humanized anti-Aß monoclonal antibody of an IgG4 isotype, known as MABT5102A (MABT). An IgG4 subclass was selected to reduce the risk of Fcγ receptor-mediated overactivation of microglia. MABT bound with high affinity to multiple forms of Aß, protected against Aß1-42 oligomer-induced cytotoxicity, and increased uptake of neurotoxic Aß oligomers by microglia. Furthermore, MABT-mediated amyloid plaque removal was demonstrated using in vivo live imaging in hAPP((V717I))/PS1 transgenic mice. When compared with a human IgG1 wild-type subclass, containing the same antigen-binding variable domains and with equal binding to Aß, MABT showed reduced activation of stress-activated p38MAPK (p38 mitogen-activated protein kinase) in microglia and induced less release of the proinflammatory cytokine TNFα. We propose that a humanized IgG4 anti-Aß antibody that takes advantage of a unique Aß binding profile, while also possessing reduced effector function, may provide a safer therapeutic alternative for passive immunotherapy for AD. Data from a phase I clinical trial testing MABT is consistent with this hypothesis, showing no signs of vasogenic edema, even in ApoE4 carriers.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Immunoglobulin G/pharmacology , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , CX3C Chemokine Receptor 1 , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Immunoglobulin G/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/genetics , Statistics, Nonparametric , Time Factors , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
EMBO J ; 26(23): 4902-12, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-17989695

ABSTRACT

Neuropilins (Nrps) are co-receptors for class 3 semaphorins and vascular endothelial growth factors and important for the development of the nervous system and the vasculature. The extracellular portion of Nrp is composed of two domains that are essential for semaphorin binding (a1a2), two domains necessary for VEGF binding (b1b2), and one domain critical for receptor dimerization (c). We report several crystal structures of Nrp1 and Nrp2 fragments alone and in complex with antibodies that selectively block either semaphorin or vascular endothelial growth factor (VEGF) binding. In these structures, Nrps adopt an unexpected domain arrangement in which the a2, b1, and b2 domains form a tightly packed core that is only loosely connected to the a1 domain. The locations of the antibody epitopes together with in vitro experiments indicate that VEGF and semaphorin do not directly compete for Nrp binding. Based upon our structural and functional data, we propose possible models for ligand binding to neuropilins.


Subject(s)
Neuropilins/chemistry , Semaphorin-3A/chemistry , Vascular Endothelial Growth Factor A/chemistry , Amino Acid Sequence , Antibodies/chemistry , Binding Sites , Crystallography, X-Ray/methods , Dimerization , Molecular Conformation , Molecular Sequence Data , Neuropilins/physiology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Semaphorin-3A/metabolism , Semaphorins/metabolism , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...