Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Neurobiol Dis ; 146: 105120, 2020 12.
Article in English | MEDLINE | ID: mdl-32991997

ABSTRACT

Deposition of tau aggregates in the brain is a pathological hallmark of several neurodegenerative diseases, termed tauopathies, such as Alzheimer's disease (AD), corticobasal degeneration, and progressive supranuclear palsy (PSP). As transcellular spread of pathological tau aggregates has been implicated in disease progression, immunotherapy is being considered as a treatment for tauopathies. Here we report a detailed biochemical and biophysical characterization of the tau-binding properties of gosuranemab, a humanized monoclonal antibody directed against N-terminal tau that is currently being investigated as a treatment for AD. Binding experiments showed that gosuranemab exhibited high affinity for tau monomer, tau fibrils, and insoluble tau from different tauopathies. Epitope mapping studies conducted using X-ray crystallography and mutagenesis showed that gosuranemab bound to human tau residues 15-22. Immunodepletion of pathological human brain homogenates and transgenic mouse interstitial fluid (ISF) with gosuranemab resulted in reduced tau aggregation in tau biosensor cells. Preincubation of seed-competent AD-tau with gosuranemab significantly inhibited tau aggregation in mouse primary cortical neurons. Gosuranemab also significantly reduced unbound N-terminal tau in cerebrospinal fluid (CSF) from individuals with PSP and AD, and in ISF and CSF of treated transgenic mice. These results are consistent with the >90% target engagement observed in the CSF of some clinical trial dosing cohorts and support the evaluation of gosuranemab as a potential treatment for AD.


Subject(s)
Alzheimer Disease/metabolism , Antibodies, Monoclonal, Humanized/metabolism , Brain/metabolism , tau Proteins/metabolism , Animals , Basal Ganglia Diseases/metabolism , Mice, Transgenic , Neurons/metabolism , Supranuclear Palsy, Progressive/metabolism , Tauopathies/metabolism , Tauopathies/pathology
2.
ACS Chem Neurosci ; 14(14): 2560-2568, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37377046

ABSTRACT

Imaging O-GlcNAcase OGA by positron emission tomography (PET) could provide information on the pathophysiological pathway of neurodegenerative diseases and important information on drug-target engagement and be helpful in dose selection of therapeutic drugs. Our aim was to develop an efficient synthetic method for labeling BIO-1819578 with carbon-11 using 11CO for evaluation of its potential to measure levels of OGA enzyme in non-human primate (NHP) brain using PET. Radiolabeling was achieved in one-pot via a carbon-11 carbonylation reaction using [11C]CO. The detailed regional brain distribution of [11C]BIO-1819578 binding was evaluated using PET measurements in NHPs. Brain radioactivity was measured for 93 min using a high-resolution PET system, and radiometabolites were measured in monkey plasma using gradient radio HPLC. Radiolabeling of [11C]BIO-1819578 was successfully accomplished, and the product was found to be stable at 1 h after formulation. [11C]BIO-1819578 was characterized in the cynomolgus monkey brain where a high brain uptake was found (7 SUV at 4 min). A pronounced pretreatment effect was found, indicating specific binding to OGA enzyme. Radiolabeling of [11C]BIO-1819578 with [11C]CO was successfully accomplished. [11C]BIO-1819578 binds specifically to OGA enzyme. The results suggest that [11C]BIO-1819578 is a potential radioligand for imaging and for measuring target engagement of OGA in the human brain.


Subject(s)
Brain , Positron-Emission Tomography , Animals , Macaca fascicularis/metabolism , Positron-Emission Tomography/methods , Carbon Radioisotopes/metabolism , Brain/diagnostic imaging , Brain/metabolism , Radiopharmaceuticals/metabolism
3.
Nat Aging ; 3(12): 1591-1601, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38012285

ABSTRACT

In Alzheimer's disease, the spread of aberrantly phosphorylated tau is an important criterion in the Braak staging of disease severity and correlates with disease symptomatology. Here, we report the results of TANGO ( NCT03352557 ), a randomized, double-blind, placebo-controlled, parallel-group and multiple-dose long-term trial of gosuranemab-a monoclonal antibody to N-terminal tau-in patients with early Alzheimer's disease. The primary objective was to assess the safety and tolerability of gosuranemab compared to placebo. The secondary objectives were to assess the efficacy of multiple doses of gosuranemab in slowing cognitive and functional impairment (using the Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) scores at week 78) and evaluate the immunogenicity of gosuranemab (using the incidence of anti-gosuranemab antibody responses). Participants were randomized (n = 654); received (n = 650) low-dose (125 mg once every 4 weeks (q4w), n = 58; 375 mg q12w, n = 58), intermediate-dose (600 mg q4w, n = 106) or high-dose (2,000 mg q4w, n = 214) gosuranemab or placebo (q4w, n = 214) intravenously for 78 weeks; and assigned to cerebrospinal fluid (n = 327) and/or tau positron emission tomography (n = 357) biomarker substudies. Gosuranemab had an acceptable safety profile and was generally well tolerated (incidence of serious adverse events: placebo, 12.1%; low dose, 10.3%; intermediate dose, 12.3%; high dose, 11.7%). The incidence of treatment-emergent gosuranemab antibody responses was low at all time points. No significant effects were identified in cognitive and functional tests as no dose resulted in a favorable change from the baseline CDR-SB score at week 78 compared to placebo control (adjusted mean change: placebo, 1.85; low dose, 2.20; intermediate dose, 2.24; high dose, 1.85). At week 76, all doses caused significant (P < 0.0001) reductions in the cerebrospinal fluid levels of unbound N-terminal tau compared to placebo.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Tomography, X-Ray Computed , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/adverse effects
4.
J Nucl Med ; 64(10): 1588-1593, 2023 10.
Article in English | MEDLINE | ID: mdl-37934021

ABSTRACT

O-GlcNAcylation is thought to play a role in the development of tau pathology in Alzheimer's disease because of its ability to modulate tau's aggregation propensity. O-GlcNAcylation is regulated by 2 enzymes: O-GlcNAc transferase and O-GlcNAcase (OGA). Development of a PET tracer would therefore be an essential tool for developing therapeutic small-molecule inhibitors of OGA, enabling clinical testing of target engagement and dose selection. Methods: A collection of small-molecule compounds was screened for inhibitory activity and high-affinity binding to OGA, as well as favorable PET tracer attributes (multidrug resistance protein 1 efflux, central nervous system PET multiparameter optimization, etc.). Two lead compounds with high affinity and selectivity for OGA were selected for further profiling, including OGA binding to tissue homogenate using a radioligand competition binding assay. In vivo pharmacokinetics were established using a microdosing approach with unlabeled compounds in rats. In vivo imaging studies were performed in rodents and nonhuman primates (NHPs) with 11C-labeled compounds. Results: Two selected candidates, BIO-735 and BIO-578, displayed promising attributes in vitro. After radiolabeling with tritium, [3H]BIO-735 and [3H]BIO-578 binding in rodent brain homogenates demonstrated dissociation constants of 0.6 and 2.3 nM, respectively. Binding was inhibited, concentration-dependently, by homologous compounds and thiamet G, a well-characterized and structurally diverse OGA inhibitor. Imaging studies in rats and NHPs showed both tracers had high uptake in the brain and inhibition of binding to OGA in the presence of a nonradioactive compound. However, only BIO-578 demonstrated reversible binding kinetics within the time frame of a PET study with a 11C-labeled molecule to enable quantification using kinetic modeling. Specificity of tracer uptake was confirmed with a 10 mg/kg blocking dose of thiamet G. Conclusion: We describe the development and testing of 2 11C PET tracers targeting the protein OGA. The lead compound BIO-578 demonstrated high affinity and selectivity for OGA in rodent and human postmortem brain tissue, leading to its further testing in NHPs. NHP PET imaging studies showed that the tracer had excellent brain kinetics, with full inhibition of specific binding by thiamet G. These results suggest that the tracer [11C]BIO-578 is well suited for further characterization in humans.


Subject(s)
Brain , beta-N-Acetylhexosaminidases , Humans , Rats , Animals , Pyrans
5.
Am J Pathol ; 178(1): 284-95, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224065

ABSTRACT

Mutations resulting in progranulin haploinsufficiency cause disease in patients with a subset of frontotemporal lobar degeneration; however, the biological functions of progranulin in the brain remain unknown. To address this subject, the present study initially assessed changes in gene expression and cytokine secretion in rat primary cortical neurons treated with progranulin. Molecular pathways enriched in the progranulin gene set included cell adhesion and cell motility pathways and pathways involved in growth and development. Secretion of cytokines and several chemokines linked to chemoattraction but not inflammation were also increased from progranulin-treated primary neurons. Therefore, whether progranulin is involved in recruitment of immune cells in the brain was investigated. Localized lentiviral expression of progranulin in C57BL/6 mice resulted in an increase of Iba1-positive microglia around the injection site. Moreover, progranulin alone was sufficient to promote migration of primary mouse microglia in vitro. Primary microglia and C4B8 cells demonstrated more endocytosis of amyloid ß1-42 when treated with progranulin. These data demonstrate that progranulin acts as a chemoattractant in the brain to recruit or activate microglia and can increase endocytosis of extracellular peptides such as amyloid ß.


Subject(s)
Brain/physiology , Chemotactic Factors/physiology , Endocytosis , Intercellular Signaling Peptides and Proteins/physiology , Microglia/physiology , Animals , Brain/drug effects , Brain/metabolism , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement , Chemotactic Factors/genetics , Chemotactic Factors/pharmacology , Cytokines/metabolism , Endocytosis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Mutant Strains , Microfilament Proteins , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Progranulins , Rats
6.
Front Neurol ; 12: 624051, 2021.
Article in English | MEDLINE | ID: mdl-34262517

ABSTRACT

Acetylation of tau protein is dysregulated in Alzheimer's Disease (AD). It has been proposed that acetylation of specific sites in the KXGS motif of tau can regulate phosphorylation of nearby residues and reduce the propensity of tau to aggregate. Histone deacetylase 6 (HDAC6) is a cytoplasmic enzyme involved in deacetylation of multiple targets, including tau, and it has been suggested that inhibition of HDAC6 would increase tau acetylation at the KXGS motifs and thus may present a viable therapeutic approach to treat AD. To directly test the contribution of HDAC6 to tau pathology, we intracerebroventricularly injected an antisense oligonucleotide (ASO) directed against HDAC6 mRNA into brains of P301S tau mice (PS19 model), which resulted in a 70% knockdown of HDAC6 protein in the brain. Despite a robust decrease in levels of HDAC6, no increase in tau acetylation was observed. Additionally, no change of tau phosphorylation or tau aggregation was detected upon the knockdown of HDAC6. We conclude that HDAC6 does not impact tau pathology in PS19 mice.

7.
Neuropharmacology ; 108: 229-37, 2016 09.
Article in English | MEDLINE | ID: mdl-27039042

ABSTRACT

Neurosteroids such as progesterone and allopregnanolone have been shown to exert neuroprotective effects under a variety of pathological or insult conditions, and there is evidence that the neurosteroid system is perturbed in Multiple Sclerosis (MS) patients. Neurosteroids are synthesized in the central nervous system (CNS) through a series of metabolic transformations, beginning with a rate-limiting step of cholesterol transport through the outer mitochondrial membrane via the transporter translocator protein (TSPO). We examined the effects of etifoxine and XBD-173, two different brain penetrant TSPO agonists, for their ability to ameliorate clinical signs in two different experimental autoimmune encephalitis (EAE) models. Etifoxine, as previously reported, was efficacious in EAE, while XBD-173 was not. Surprisingly, XBD-173, but not etifoxine elevated relevant neurosteroids in brain of female rats and differed in its ability to exert anti-inflammatory and direct neuroprotective effects in vitro as compared to etifoxine. We conclude that the neurosteroid elevations produced in brain by XBD-173 are not sufficient to ameliorate EAE and suggest that etifoxine may have additional mechanisms of action that provide therapeutic benefit in this model system.


Subject(s)
Disease Models, Animal , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Oxazines/metabolism , Purines/metabolism , Receptors, GABA/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Ligands , Mice , Mice, Inbred C57BL , Oxazines/therapeutic use , Purines/therapeutic use , Rats , Treatment Outcome
8.
J Neurosci ; 23(37): 11759-69, 2003 Dec 17.
Article in English | MEDLINE | ID: mdl-14684878

ABSTRACT

The number and shape of dendritic spines are influenced by activity and regulated by molecules that organize the actin cytoskeleton of spines. Cortactin is an F-actin binding protein and activator of the Arp2/3 actin nucleation machinery that also interacts with the postsynaptic density (PSD) protein Shank. Cortactin is concentrated in dendritic spines of cultured hippocampal neurons, and the N-terminal half of the protein containing the Arp2/3 and F-actin binding domains is necessary and sufficient for spine targeting. Knockdown of cortactin protein by short-interfering RNA (siRNA) results in depletion of dendritic spines in hippocampal neurons, whereas overexpression of cortactin causes elongation of spines. In response to synaptic stimulation and NMDA receptor activation, cortactin redistributes rapidly from spines to dendritic shaft, correlating with remodeling of the actin cytoskeleton, implicating cortactin in the activity-dependent regulation of spine morphogenesis.


Subject(s)
Dendrites/chemistry , Dendrites/ultrastructure , Hippocampus/cytology , Microfilament Proteins/analysis , Microfilament Proteins/physiology , Animals , Cells, Cultured , Cortactin , Dendrites/metabolism , Glutamic Acid/pharmacology , Microfilament Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Protein Structure, Tertiary , Protein Transport , RNA Interference , Rats , Receptors, N-Methyl-D-Aspartate/metabolism
9.
J Neurosci ; 23(8): 3262-71, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12716933

ABSTRACT

Cholesterol/sphingolipid microdomains (lipid rafts) in the membrane are involved in protein trafficking, formation of signaling complexes, and regulation of actin cytoskeleton. Here, we show that lipid rafts exist abundantly in dendrites of cultured hippocampal neurons, in which they are associated with several postsynaptic proteins including surface AMPA receptors. Depletion of cholesterol/sphingolipid leads to instability of surface AMPA receptors and gradual loss of synapses (both inhibitory and excitatory) and dendritic spines. The remaining synapses and spines in raft-depleted neurons become greatly enlarged. The importance of lipid rafts for normal synapse density and morphology could explain why cholesterol promotes synapse maturation in retinal ganglion cells (Mauch et al., 2001) and offers a potential link between disordered cholesterol metabolism and the synapse loss seen in neurodegenerative disease.


Subject(s)
Dendrites/chemistry , Lovastatin/analogs & derivatives , Membrane Microdomains/chemistry , Neurons/chemistry , Receptors, AMPA/chemistry , Synapses/chemistry , Animals , Brain Chemistry , Carrier Proteins/analysis , Carrier Proteins/metabolism , Cell Membrane/chemistry , Cells, Cultured , Cholesterol/analysis , Dendrites/metabolism , Dendrites/ultrastructure , Detergents/chemistry , Enzyme Inhibitors/pharmacology , Filipin/chemistry , Filipin/metabolism , Fumonisins/pharmacology , Intracellular Signaling Peptides and Proteins , Lovastatin/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/analysis , Sphingolipids/analysis , Subcellular Fractions/chemistry , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure
10.
FEBS Lett ; 521(1-3): 185-9, 2002 Jun 19.
Article in English | MEDLINE | ID: mdl-12067714

ABSTRACT

In Drosophila, the frizzled gene plays a critical role in the establishment of tissue polarity, but the function of the Frizzled family of proteins in mammals is largely unknown. Recent evidence suggested that Frizzleds are receptors for the Wnt family of secreted glycoproteins which are involved in cell fate determination. However, it is unclear how Frizzled receptors transduce Wnt signals to intracellular signaling components. Here we show that the mouse Frizzled-1, -2, -4 and -7 can bind to proteins of the PSD-95 family, which are implicated in the assembly and localization of multiprotein signaling complexes in the brain. Moreover, PSD-95 can form a ternary complex with Frizzled-2 and the adenomatous polyposis coli protein, a negative regulator of Wnt signaling, suggesting that members of the PSD-95 family may serve to recruit intracellular signaling molecules of the Wnt/Frizzled pathway into the vicinity of the receptor.


Subject(s)
Nerve Tissue Proteins/metabolism , Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Disks Large Homolog 4 Protein , Frizzled Receptors , Guanylate Kinases , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary , Proteins/genetics , Receptors, Cell Surface/genetics , Receptors, Neurotransmitter/genetics
11.
Neuropharmacology ; 79: 307-13, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24326295

ABSTRACT

Neurofibrillary tangles (NFT), mainly consisting of fibrillar aggregates of hyperphosphorylated tau, are a defining pathological feature of Alzheimer's Disease and other tauopathies. Progressive accumulation of tau into NFT is considered to be a toxic cellular event causing neurodegeneration. Tau is subject to O-linked N-acetylglucosamine (O-GlcNAc) modification and O-GlcNAcylation of tau has been suggested to regulate tau phosphorylation. We tested if an increase in tau O-GlcNAcylation affected tau phosphorylation and aggregation in the rTg4510 tau transgenic mouse model. Acute treatment of rTg4510 mice with an O-GlcNAcase inhibitor transiently reduced tau phosphorylation at epitopes implicated in tau pathology. More importantly, long-term inhibitor treatment strongly increased tau O-GlcNAcylation, reduced the number of dystrophic neurons, and protected against the formation of pathological tau species without altering the phosphorylation of non-pathological tau. This indicates that O-GlcNAcylation prevents the aggregation of tau in a manner that does not affect its normal phosphorylation state. Collectively, our results support O-GlcNAcase inhibition as a potential therapeutic strategy for the treatment of Alzheimer's Disease and other tauopathies.


Subject(s)
Acetylglucosamine/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Pyrans/pharmacology , Tauopathies/drug therapy , Thiazoles/pharmacology , tau Proteins/metabolism , Acetylglucosamine/antagonists & inhibitors , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Disease Models, Animal , Female , Glycosylation , Male , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Phosphorylation/drug effects , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/antagonists & inhibitors
12.
FEBS Lett ; 587(22): 3722-8, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24113653

ABSTRACT

Aggregation of tau into paired helical filaments is a pathological process leading to neurotoxicity in Alzheimer's disease and other tauopathies. Tau is posttranslationally modified by O-linked N-acetylglucosamine (O-GlcNAc), and increasing tau O-GlcNAcylation may protect against its aggregation. Research tools to study the relationship between tau aggregation and tau O-GlcNAcylation have not been widely available. Here we describe the generation of a rabbit monoclonal antibody specific for tau O-GlcNAcylated at Ser400 (O-tau(S400)). We show the utility of this antibody for in vitro and in vivo experiments to investigate the function of O-GlcNAc modifications of tau at Ser400.


Subject(s)
Antibodies, Monoclonal/chemistry , Protein Processing, Post-Translational , tau Proteins/immunology , Acetylglucosamine/metabolism , Animals , Antibody Specificity , Glycosylation , HEK293 Cells , Humans , Mice , Mice, Knockout , Protein Binding , Rabbits , Serine/metabolism , tau Proteins/chemistry , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL