Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Mol Ther Methods Clin Dev ; 31: 101158, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38074413

ABSTRACT

Over the last decade, there has been a growing interest in intrabodies and their therapeutic potential. Intrabodies are antibody fragments that are expressed inside a cell to target intracellular antigens. In the context of intracellular protein misfolding and aggregation, such as tau pathology in Alzheimer's disease, intrabodies have become an interesting approach as there is the possibility to target early stages of aggregation. As such, we engineered three anti-tau monoclonal antibodies into single-chain variable fragments for cytoplasmic expression and activity: PT51, PT77, and hTau21. Due to the reducing environment of the cytoplasm, single-chain variable fragment (scFv) aggregation is commonly observed. Therefore, we also performed complementarity-determining region (CDR) grafting into three different stable frameworks to rescue solubility and intracellular binding. All three scFvs retained binding to tau after cytoplasmic expression in HEK293 cells, in at least one of the frameworks. Subsequently, we show their capacity to interfere with either mouse or mutant human tau aggregation in two different primary mouse neuron models and organotypic hippocampal slice cultures. Collectively, our work extends the current knowledge on intracellular tau targeting with intrabodies, providing three scFv intrabodies that can be used as immunological tools to target tau inside cells.

2.
Life (Basel) ; 13(10)2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37895469

ABSTRACT

BACKGROUND: The Tau58/2 and Tau58/4 mouse lines expressing 0N4R tau with a P301S mutation mimic aspects of frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). In a side-by-side comparison, we report the age-dependent development of cognitive, motor, and behavioral deficits in comparison with the spatial-temporal evolution of cellular tau pathology in both models. METHODS: We applied the SHIRPA primary screen and specific neuromotor, behavioral, and cognitive paradigms. The spatiotemporal development of tau pathology was investigated immunohistochemically. Levels of sarkosyl-insoluble paired helical filaments were determined via a MesoScale Discovery biomarker assay. RESULTS: Neuromotor impairments developed from age 3 months in both models. On electron microscopy, spinal cord neurofibrillary pathology was visible in mice aged 3 months; however, AT8 immunoreactivity was not yet observed in Tau58/4 mice. Behavioral abnormalities and memory deficits occurred at a later stage (>9 months) when tau pathology was fully disseminated throughout the brain. Spatiotemporally, tau pathology spread from the spinal cord via the midbrain to the frontal cortex, while the hippocampus was relatively spared, thus explaining the late onset of cognitive deficits. CONCLUSIONS: Our findings indicate the face and construct validity of both Tau58 models, which may provide new, valuable insights into the pathologic effects of tau species in vivo and may consequently facilitate the development of new therapeutic targets to delay or halt neurodegenerative processes occurring in tauopathies.

3.
J Alzheimers Dis ; 93(1): 151-167, 2023.
Article in English | MEDLINE | ID: mdl-36970909

ABSTRACT

BACKGROUND: Clearance of tau seeds by immunization with tau antibodies is currently evaluated as therapeutic strategy to block the spreading of tau pathology in Alzheimer's disease and other tauopathies. Preclinical evaluation of passive immunotherapy is performed in different cellular culture systems and in wild-type and human tau transgenic mouse models. Depending on the preclinical model used, tau seeds or induced aggregates can either be of mouse, human or mixed origin. OBJECTIVE: We aimed to develop human and mouse tau-specific antibodies to discriminate between the endogenous tau and the introduced form in preclinical models. METHODS: Using hybridoma technology, we developed human and mouse tau-specific antibodies that were then used to develop several assays to specifically detect mouse tau. RESULTS: Four antibodies, mTau3, mTau5, mTau8, and mTau9, with a high degree of specificity for mouse tau were identified. Additionally, their potential application in highly sensitive immunoassays to measure tau in mouse brain homogenate and cerebrospinal fluid is illustrated, as well as their application for specific endogenous mouse tau aggregation detection. CONCLUSION: The antibodies reported here can be very important tools to better interpret the results obtained from different model systems as well as to study the role of endogenous tau in tau aggregation and pathology observed in the diverse mouse models available.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Humans , Animals , tau Proteins/metabolism , Tauopathies/pathology , Alzheimer Disease/pathology , Mice, Transgenic , Disease Models, Animal , Antibodies, Monoclonal , Brain/pathology
4.
Med ; 3(12): 860-882.e15, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36257298

ABSTRACT

BACKGROUND: The near impermeability of the blood-brain barrier (BBB) and the unique neuroimmune environment of the CNS prevents the effective use of antibodies in neurological diseases. Delivery of biotherapeutics to the brain can be enabled through receptor-mediated transcytosis via proteins such as the transferrin receptor, although limitations such as the ability to use Fc-mediated effector function to clear pathogenic targets can introduce safety liabilities. Hence, novel delivery approaches with alternative clearance mechanisms are warranted. METHODS: Binders that optimized transport across the BBB, known as transcytosis-enabling modules (TEMs), were identified using a combination of antibody discovery techniques and pharmacokinetic analyses. Functional activity of TEMs were subsequently evaluated by imaging for the ability of myeloid cells to phagocytose target proteins and cells. FINDINGS: We demonstrated significantly enhanced brain exposure of therapeutic antibodies using optimal transferrin receptor or CD98 TEMs. We found that these modules also mediated efficient clearance of tau aggregates and HER2+ tumor cells via a non-classical phagocytosis mechanism through direct engagement of myeloid cells. This mode of clearance potentially avoids the known drawbacks of FcγR-mediated antibody mechanisms in the brain such as the neurotoxic release of proinflammatory cytokines and immune cell exhaustion. CONCLUSIONS: Our study reports a new brain delivery platform that harnesses receptor-mediated transcytosis to maximize brain uptake and uses a non-classical phagocytosis mechanism to efficiently clear pathologic proteins and cells. We believe these findings will transform therapeutic approaches to treat CNS diseases. FUNDING: This research was funded by Janssen, Pharmaceutical Companies of Johnson & Johnson.


Subject(s)
Blood-Brain Barrier , Transcytosis , Blood-Brain Barrier/metabolism , Transcytosis/physiology , Receptors, Transferrin , Biological Transport/physiology , Antibodies
5.
Alzheimers Dement (Amst) ; 13(1): e12204, 2021.
Article in English | MEDLINE | ID: mdl-34095436

ABSTRACT

INTRODUCTION: Diagnosis of Alzheimer's disease (AD) based on amyloid beta (A), pathologic tau (T), and neurodegeneration (N) biomarkers in peripheral fluids promises to accelerate clinical trials and intercept disease earlier. METHODS: Qualification of a Simoa plasma p217+tau assay was performed, followed by clinical utility evaluation in a cohort of 227 subjects with broad A and T spectrum. RESULTS: The p217+tau plasma assay was accurate, precise, dilution linear, and highly sensitive. All measured samples were within linear range of the assay, presented higher concentration in AD versus healthy controls (P < .0001), and plasma and cerebrospinal fluid levels correlated (r2 = 0.35). The plasma p217+tau results were predictive of central T and A status (area under the curve = 0.90 and 0.90, respectively) with low false +/- rates. DISCUSSION: The assay described here exhibits good technical performance and shows potential as a highly accurate peripheral biomarker for A or T status in AD and cognitively normal subjects.

6.
BMC Mol Cell Biol ; 21(1): 81, 2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33183222

ABSTRACT

BACKGROUND: Although several studies demonstrate prion-like properties of Tau fibrils, the effect of size in the seeding capacity of these aggregates is not fully understood. The aim of this study is to characterize Tau seeds by their size and seeding capacity. METHODS: Tau aggregates were isolated from postmortem AD brain tissue and separated from low molecular weight species by sucrose gradient ultracentrifugation. Biochemical characterization of the different fractions was done by non-reducing Western blotting and aggregate-specific immuno-assays using in house developed anti-Tau monoclonal antibodies, including PT76 which binds to an epitope close to the microtubule-binding domain and, hence, also to K18. Seeding efficiency was then assessed in HEK293 cells expressing K18 FRET sensors. RESULTS: We observed that upon sonication of Tau aggregates different size-distributed tau aggregates are obtained. In biochemical assays, these forms show higher signals than the non-sonicated material in some aggregation-specific Tau assays. This could be explained by an increased epitope exposure of the smaller aggregates created by the sonication. By analyzing human brain derived and recombinant (K18) Tau aggregates in a cellular FRET assay, it was observed that, in the absence of transfection reagent, sonicated aggregates showed higher aggregation induction. Preparations also showed altered profiles on native PAGE upon sonication and we could further separate different aggregate species based on their molecular weight via sucrose gradients. CONCLUSIONS: This study further elucidates the molecular properties regarding relative aggregate size and seeding efficiency of sonicated vs. non-sonicated high molecular weight Tau species. This information will provide a better knowledge on how sonication, a commonly used technique in the field of study of Tau aggregation, impacts the aggregates. In addition, the description of PT76-based aggregation specific assay is a valuable tool to quantify K18 and human AD Tau fibrils.


Subject(s)
Alzheimer Disease/metabolism , Protein Aggregation, Pathological/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Epitopes , HEK293 Cells , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Protein Aggregates , Protein Aggregation, Pathological/genetics , Protein Binding , Recombinant Proteins , Sonication , Spectroscopy, Fourier Transform Infrared , tau Proteins/genetics , tau Proteins/ultrastructure
7.
J Alzheimers Dis ; 77(4): 1397-1416, 2020.
Article in English | MEDLINE | ID: mdl-32894244

ABSTRACT

BACKGROUND: As a consequence of the discovery of an extracellular component responsible for the progression of tau pathology, tau immunotherapy is being extensively explored in both preclinical and clinical studies as a disease modifying strategy for the treatment of Alzheimer's disease. OBJECTIVE: Describe the characteristics of the anti-phospho (T212/T217) tau selective antibody PT3 and its humanized variant hPT3. METHODS: By performing different immunization campaigns, a large collection of antibodies has been generated and prioritized. In depth, in vitro characterization using surface plasmon resonance, phospho-epitope mapping, and X-ray crystallography experiments were performed. Further characterization involved immunohistochemical staining on mouse- and human postmortem tissue and neutralization of tau seeding by immunodepletion assays. RESULTS AND CONCLUSION: Various in vitro experiments demonstrated a high intrinsic affinity for PT3 and hPT3 for AD brain-derived paired helical filaments but also to non-aggregated phospho (T212/T217) tau. Further functional analyses in cellular and in vivo models of tau seeding demonstrated almost complete depletion of tau seeds in an AD brain homogenate. Ongoing trials will provide the clinical evaluation of the tau spreading hypothesis in Alzheimer's disease.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal/metabolism , Drug Discovery/methods , tau Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Structure, Tertiary , tau Proteins/chemistry
8.
J Alzheimers Dis ; 77(4): 1417-1430, 2020.
Article in English | MEDLINE | ID: mdl-32831201

ABSTRACT

BACKGROUND: Early and accurate detection and staging is critical to managing Alzheimer's disease (AD) and supporting clinical trials. Cerebrospinal fluid (CSF) biomarkers for amyloid-ß peptides, tau species, and various neurodegenerative and inflammatory analytes are leading the way in this regard, yet there is room for improved sensitivity and specificity. In particular tau is known to be present in many different fragments, conformations, and post-translationally modified forms. While the exact tau species that might best reflect AD pathology is unknown, a growing body of evidence suggests that forms with high levels of phosphorylation in the mid-region may be especially enriched in AD. OBJECTIVE: Develop an assay for measuring p217tau in CSF. METHODS: Here we describe the development and validation of a novel sELISA for measuring CSF tau species containing phosphorylation at threonines 212 & 217, aka p217 + tau, using the PT3 antibody. RESULTS: While the analyte is present at extremely low levels the assay is sufficiently sensitive and specific to quantitate p217 + tau with excellent precision, accuracy, and dilution linearity, allowing good differentiation between diagnostic subgroups. The p217 + tau measurements appear to track AD pathology better than the commonly used p181tau epitope, suggesting superior diagnostic and staging performance. Finally, the assay can also be configured to differentiate antibody-bound versus antibody-free tau, and therefore can be used to measure target engagement by p217 + tau-targeting immunotherapeutics. CONCLUSION: The assay for measuring p217 + tau described here is highly sensitive, accurate, precise, dilution linear, and shows good potential for identifying and staging AD.


Subject(s)
Peptide Fragments/analysis , Peptide Fragments/metabolism , tau Proteins/analysis , tau Proteins/metabolism , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnosis , Biomarkers/cerebrospinal fluid , Cohort Studies , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Female , Humans , Male , Middle Aged , Reproducibility of Results
9.
Neurobiol Dis ; 143: 105011, 2020 09.
Article in English | MEDLINE | ID: mdl-32653674

ABSTRACT

Progressive accumulation of hyperphosphorylated tau is a hallmark of various neurodegenerative disorders including Alzheimer's disease. However, to date, the functional effects of tau pathology on brain network connectivity remain poorly understood. To directly interrogate the impact of tau pathology on functional brain connectivity, we conducted a longitudinal experiment in which we monitored a fibril-seeded hTau.P301L mouse model using correlative whole-brain microscopy and resting-state functional MRI. Despite a progressive aggravation of tau pathology across the brain, the major resting-state networks appeared unaffected up to 15 weeks after seeding. Targeted analyses also showed that the connectivity of regions with high levels of hyperphosphorylated tau was comparable to that observed in controls. In line with the ostensible retention of connectivity, no behavioural changes were detected between seeded and control hTau.P301L mice as determined by three different paradigms. Our data indicate that seeded tau pathology, with accumulation of tau aggregates throughout different regions of the brain, does not alter functional connectivity or behaviour in this mouse model. Additional correlative functional studies on different mouse models should help determine whether this is a generalizable trait of tauopathies.


Subject(s)
Brain/physiopathology , Nerve Net/physiopathology , Neural Pathways/physiopathology , Protein Aggregation, Pathological/physiopathology , tau Proteins/metabolism , Animals , Brain/pathology , Disease Models, Animal , Humans , Magnetic Resonance Imaging , Mice , Nerve Net/pathology , Neural Pathways/pathology , Protein Aggregation, Pathological/pathology
10.
Neurobiol Dis ; 127: 398-409, 2019 07.
Article in English | MEDLINE | ID: mdl-30878534

ABSTRACT

We have exploited whole brain microscopy to map the progressive deposition of hyperphosphorylated tau in intact, cleared mouse brain. We found that the three-dimensional spreading pattern of hyperphosphorylated tau in the brain of an aging Tau.P301L mouse model did not resemble that observed in AD patients. Injection of synthetic or patient-derived tau fibrils in the CA1 region resulted in a more faithful spreading pattern. Atlas-guided volumetric analysis showed a connectome-dependent spreading from the injection site and also revealed hyperphosphorylated tau deposits beyond the direct anatomical connections. In fibril-injected brains, we also detected a persistent subpopulation of rod-like and swollen microglia. Furthermore, we showed that the hyperphosphorylated tau load could be reduced by intracranial co-administration of, and to a lesser extent, by repeated systemic dosing with an antibody targeting the microtubule-binding domain of tau. Thus, the combination of targeted seeding and in toto staging of tau pathology allowed assessing regional vulnerability in a comprehensive manner, and holds potential as a preclinical drug validation tool.


Subject(s)
Brain/metabolism , Microglia/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Disease Models, Animal , Disease Progression , Mice , Mice, Transgenic , Microglia/pathology , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation , Tauopathies/pathology
11.
Acta Neuropathol Commun ; 6(1): 59, 2018 07 12.
Article in English | MEDLINE | ID: mdl-30001207

ABSTRACT

Aggregation of tau protein and spreading of tau aggregates are pivotal pathological processes in a range of neurological disorders. Accumulating evidence suggests that immunotherapy targeting tau may be a viable therapeutic strategy. We have previously described the isolation of antibody CBTAU-22.1 from the memory B-cell repertoire of healthy human donors. CBTAU-22.1 was shown to specifically bind a disease-associated phosphorylated epitope in the C-terminus of tau (Ser422) and to be able to inhibit the spreading of pathological tau aggregates from P301S spinal cord lysates in vitro, albeit with limited potency. Using a combination of rational design and random mutagenesis we have derived a variant antibody with improved affinity while maintaining the specificity of the parental antibody. This affinity improved antibody showed greatly enhanced potency in a cell-based immunodepletion assay using paired helical filaments (PHFs) derived from human Alzheimer's disease (AD) brain tissue. Moreover, the affinity improved antibody limits the in vitro aggregation propensity of full length tau species specifically phosphorylated at position 422 produced by employing a native chemical ligation approach. Together, these results indicate that in addition to being able to inhibit the spreading of pathological tau aggregates, the matured antibody can potentially also interfere with the nucleation of tau which is believed to be the first step of the pathogenic process. Finally, the functionality in a P301L transgenic mice co-injection model highlights the therapeutic potential of human antibody dmCBTAU-22.1.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Antibodies/pharmacology , Brain/metabolism , Serine/metabolism , tau Proteins/immunology , tau Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Antibody Affinity/drug effects , Autopsy , Brain/pathology , Dose-Response Relationship, Drug , Epitopes/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Microscopy, Atomic Force , Middle Aged , Models, Molecular , Mutagenesis , Mutation/genetics , Phosphorylation/physiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/therapy
12.
J Alzheimers Dis ; 65(1): 265-281, 2018.
Article in English | MEDLINE | ID: mdl-30040731

ABSTRACT

The tau spreading hypothesis provides rationale for passive immunization with an anti-tau monoclonal antibody to block seeding by extracellular tau aggregates as a disease-modifying strategy for the treatment of Alzheimer's disease (AD) and potentially other tauopathies. As the biochemical and biophysical properties of the tau species responsible for the spatio-temporal sequences of seeding events are poorly defined, it is not yet clear which epitope is preferred for obtaining optimal therapeutic efficacy. Our internal tau antibody collection has been generated by immunizations with different tau species: aggregated- and non-aggregated tau and human postmortem AD brain-derived tau fibrils. In this communication, we describe and characterize a set of these anti-tau antibodies for their biochemical and biophysical properties, including binding, tissue staining by immunohistochemistry, and epitope. The antibodies bound to different domains of the tau protein and some were demonstrated to be isoform-selective (PT18 and hTau56) or phospho-selective (PT84). Evaluation of the antibodies in cellular- and in vivo seeding assays revealed clear differences in maximal efficacy. Limited proteolysis experiments support the hypothesis that some epitopes are more exposed than others in the tau seeds. Moreover, antibody efficacy seems to depend on the structural properties of fibrils purified from tau Tg mice- and postmortem human AD brain.


Subject(s)
Alzheimer Disease/pathology , Antibodies, Monoclonal/metabolism , Brain/metabolism , tau Proteins/immunology , Animals , Epitope Mapping , Female , HEK293 Cells , Humans , Immunization, Passive , Male , Mice , Mice, Knockout , Mutation/genetics , Surface Plasmon Resonance , tau Proteins/deficiency , tau Proteins/genetics
13.
Acta Neuropathol Commun ; 6(1): 43, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855358

ABSTRACT

Misfolding and aggregation of tau protein are closely associated with the onset and progression of Alzheimer's Disease (AD). By interrogating IgG+ memory B cells from asymptomatic donors with tau peptides, we have identified two somatically mutated VH5-51/VL4-1 antibodies. One of these, CBTAU-27.1, binds to the aggregation motif in the R3 repeat domain and blocks the aggregation of tau into paired helical filaments (PHFs) by sequestering monomeric tau. The other, CBTAU-28.1, binds to the N-terminal insert region and inhibits the spreading of tau seeds and mediates the uptake of tau aggregates into microglia by binding PHFs. Crystal structures revealed that the combination of VH5-51 and VL4-1 recognizes a common Pro-Xn-Lys motif driven by germline-encoded hotspot interactions while the specificity and thereby functionality of the antibodies are defined by the CDR3 regions. Affinity improvement led to improvement in functionality, identifying their epitopes as new targets for therapy and prevention of AD.


Subject(s)
B-Lymphocytes/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/metabolism , tau Proteins/immunology , tau Proteins/metabolism , Adolescent , Adult , Aged , Antibody Specificity , B-Lymphocytes/drug effects , Crystallization , Dose-Response Relationship, Drug , Female , Humans , Immunodominant Epitopes/metabolism , Male , Microglia/metabolism , Microscopy, Atomic Force , Middle Aged , Models, Molecular , Molecular Sequence Data , Protein Aggregates , Young Adult
14.
Acta Neuropathol ; 131(4): 549-69, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26739002

ABSTRACT

Genetic, clinical, histopathological and biomarker data strongly support Beta-amyloid (Aß) induced spreading of Tau-pathology beyond entorhinal cortex (EC), as a crucial process in conversion from preclinical cognitively normal to Alzheimer's Disease (AD), while the underlying mechanism remains unclear. In vivo preclinical models have reproducibly recapitulated Aß-induced Tau-pathology. Tau pathology was thereby also induced by aggregated Aß, in functionally connected brain areas, reminiscent of a prion-like seeding process. In this work we demonstrate, that pre-aggregated Aß can directly induce Tau fibrillization by cross-seeding, in a cell-free assay, comparable to that demonstrated before for alpha-synuclein and Tau. We furthermore demonstrate, in a well-characterized cellular Tau-aggregation assay that Aß-seeds cross-seeded Tau-pathology and strongly catalyzed pre-existing Tau-aggregation, reminiscent of the pathogenetic process in AD. Finally, we demonstrate that heterotypic seeded Tau by pre-aggregated Aß provides efficient seeds for induction and propagation of Tau-pathology in vivo. Prion-like, heterotypic seeding of Tau fibrillization by Aß, providing potent seeds for propagating Tau pathology in vivo, as demonstrated here, provides a compelling molecular mechanism for Aß-induced propagation of Tau-pathology, beyond regions with pre-existing Tau-pathology (entorhinal cortex/locus coeruleus). Cross-seeding along functional connections could thereby resolve the initial spatial dissociation between amyloid- and Tau-pathology, and preferential propagation of Tau-pathology in regions with pre-existing 'silent' Tau-pathology, by conversion of a 'silent' Tau pathology to a 'spreading' Tau-pathology, observed in AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Prion Proteins/metabolism , Protein Aggregation, Pathological/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Analysis of Variance , Animals , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , Presenilin-1/metabolism , Prion Proteins/ultrastructure , Protein Aggregation, Pathological/chemically induced , Protein Aggregation, Pathological/pathology , Tauopathies/genetics , Transfection , tau Proteins/genetics , tau Proteins/ultrastructure
15.
Neurobiol Dis ; 73: 83-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25220759

ABSTRACT

Neurofibrillary tangles composed of hyperphosphorylated fibrillized tau are found in numerous tauopathies including Alzheimer's disease. Increasing evidence suggests that tau pathology can be transmitted from cell-to-cell; however the mechanisms involved in the initiation of tau fibrillization and spreading of disease linked to progression of tau pathology are poorly understood. We show here that intracerebral injections of preformed synthetic tau fibrils into the hippocampus or frontal cortex of young tau transgenic mice expressing mutant human P301L tau induces tau hyperphosphorylation and aggregation around the site of injection, as well as a time-dependent propagation of tau pathology to interconnected brain areas distant from the injection site. Furthermore, we show that the tau pathology as a consequence of injection of tau preformed fibrils into the hippocampus induces selective loss of CA1 neurons. Together, our data confirm previous studies on the seeded induction and the spreading of tau pathology in a different tau transgenic mouse model and reveals neuronal loss associated with seeded tau pathology in tau transgenic mouse brain. These results further validate the utility of the tau seeding model in studying disease transmission, and provide a more complete in vivo tauopathy model with associated neurodegeneration which can be used to investigate the mechanisms involved in tau aggregation and spreading, as well as aid in the search for disease modifying treatments for Alzheimer's disease and related tauopathies.


Subject(s)
Tauopathies , tau Proteins/administration & dosage , tau Proteins/genetics , Age Factors , Analysis of Variance , Animals , Disease Models, Animal , Disease Progression , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Mutation/genetics , Neurofibrillary Tangles/metabolism , Tauopathies/chemically induced , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/chemistry
16.
J Cell Sci ; 128(3): 541­52, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25501810

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with Parkinson's disease, but the precise physiological function of the protein remains ill-defined. Recently, our group proposed a model in which LRRK2 kinase activity is part of an EndoA phosphorylation cycle that facilitates efficient vesicle formation at synapses in the Drosophila melanogaster neuromuscular junctions.Flies harbor only one Lrrk gene, which might encompass the functions of both mammalian LRRK1 and LRRK2. We therefore studied the role of LRRK2 in mammalian synaptic function and provide evidence that knockout or pharmacological inhibition of LRRK2 results in defects in synaptic vesicle endocytosis, altered synaptic morphology and impairments in neurotransmission. In addition, our data indicate that mammalian endophilin A1 (EndoA1,also known as SH3GL2) is phosphorylated by LRRK2 in vitro at T73 and S75, two residues in the BAR domain. Hence, our results indicate that LRRK2 kinase activity has an important role in the regulation of clathrin-mediated endocytosis of synaptic vesicles and subsequent neurotransmission at the synapse.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endocytosis/genetics , Protein Serine-Threonine Kinases/genetics , Synaptic Transmission/genetics , Synaptic Vesicles/genetics , Animals , Cells, Cultured , Clathrin/metabolism , Drosophila melanogaster , Dynamin I/antagonists & inhibitors , Endocytosis/drug effects , Hippocampus/cytology , Hydrazones/pharmacology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Long-Evans , Sucrose/pharmacology , Synaptic Transmission/drug effects
17.
J Pharm Biomed Anal ; 76: 49-58, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23313773

ABSTRACT

After the discovery of kinase activating mutations in leucine-rich repeat kinase 2 (LRRK2) as associated with autosomal dominant forms of Parkinson's disease, inhibition of the kinase is being extensively explored as a disease modifying strategy. As signaling properties and substrate(s) of LRRK2 are poorly documented, autophosphorylation has been an important readout for the enzyme's activity. Western blotting using anti-phospho-S910 or S935 LRRK2 antibodies showed effectiveness in demonstrating inhibitory effects of compounds. In this communication we describe two types of enzyme-linked immunosorbent assays (ELISA) to determine LRRK2 protein levels and kinase activity. Both assays take advantage of the sensitivity of the earlier described total and pS935 antibodies for detection (Nichols et al., Biochem. J. 2010) [10]. The first assay is based on anti-GFP-based capturing of overexpressed LRRK2 and is highly suitable to show cellular effects of kinase inhibitors in a 96-well format. In the other platform anti-LRRK2-based capturing allows detection of endogenously expressed LRRK2 in rat tissue with no significant signal in tissue from LRRK2 knockout rats. Furthermore, both assays showed a significant reduction in pS935 levels on cellular and transgenic R1441C/G LRRK2. With the anti-LRRK2 ELISA we were able to detect LRRK2 phosphorylation in human peripheral blood mononuclear cells (PBMC). To conclude, we report two sensitive assays to monitor LRRK2 expression and kinase activity in samples coming from cellular and in vivo experimental settings. Both can show their value in drug screening and biomarker development but will also be useful in the elucidation of LRRK2-mediated signaling pathways.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Rats , Rats, Long-Evans , Sensitivity and Specificity
18.
Eur J Pharmacol ; 701(1-3): 73-81, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23340224

ABSTRACT

This study elucidates signalling cascades involved in the neurotrophic effects induced by an active compound of Synaptolepis kirkii, a plant that is used against snakebites and for treatment of epilepsy. The active compound of this plant, synaptolepis factor K7 (K7), is suggested to exert anti-tumoral and neurotrophic actions via modulation of PKC. In SH-SY5Y cells synthesis of the neuronal marker growth-associated protein 43 was increased upon 48h treatment with K7. Immunofluorescent staining of neurites revealed an increased neurite formation by synaptolepis factor K7. Short-term signal transduction events were followed at the level of extracellular-regulated kinase phosphorylation. Extracellular-regulated kinase (ERK) phosphorylation was transiently increased upon stimulation with synaptolepis factor K7 (300nM) with a maximal effect at 30min. Use of the general PKC inhibitor bisindolylmaleimide I blocked the K7-induced ERK phosphorylation suggesting involvement of PKC. Conversely, inhibition of conventional PKCs, α, ß and γ by treatment with Go6976 did not inhibit ERK phosphorylation up to 1µM. Use of a specific-PKCε translocation inhibitor peptide or RNAi-mediated knockdown of PKC-epsilon (ε) abolished the K7-induced ERK phosphorylation implicating PKCε in K7 function. This was confirmed by the observed increase in PKCε translocation and autophosphorylation induced by the compound. These data show that synaptolepis factor K7 induces neuronal differentiation of SH-SY5Y cells concomitant with a transient increase in ERK phosphorylation that is mediated by activation of PKCε.


Subject(s)
Plant Extracts/pharmacology , Protein Kinase C-epsilon/metabolism , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Phosphorylation/drug effects , Thymelaeaceae/chemistry , Time Factors
19.
Neuron ; 75(6): 1008-21, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998870

ABSTRACT

LRRK2 is a kinase mutated in Parkinson's disease, but how the protein affects synaptic function remains enigmatic. We identified LRRK2 as a critical regulator of EndophilinA. Using genetic and biochemical studies involving Lrrk loss-of-function mutants and Parkinson-related LRRK2(G2019S) gain-of-kinase function, we show that LRRK2 affects synaptic endocytosis by phosphorylating EndoA at S75, a residue in the BAR domain. We show that LRRK2-mediated EndoA phosphorylation has profound effects on EndoA-dependent membrane tubulation and membrane association in vitro and in vivo and on synaptic vesicle endocytosis at Drosophila neuromuscular junctions in vivo. Our work uncovers a regulatory mechanism that indicates that reduced LRRK2 kinase activity facilitates EndoA membrane association, while increased kinase activity inhibits membrane association. Consequently, both too much and too little LRRK2-dependent EndoA phosphorylation impedes synaptic endocytosis, and we propose a model in which LRRK2 kinase activity is part of an EndoA phosphorylation cycle that facilitates efficient vesicle formation at synapses.


Subject(s)
Acyltransferases/metabolism , Drosophila Proteins/metabolism , Endocytosis/physiology , Neuromuscular Junction/physiology , Protein Serine-Threonine Kinases/metabolism , Acyltransferases/genetics , Animals , Animals, Genetically Modified , Brain/cytology , Brain/metabolism , CHO Cells , Calcium/metabolism , Clathrin/metabolism , Cricetinae , Drosophila , Drosophila Proteins/genetics , Endocytosis/genetics , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mass Spectrometry , Mice , Microscopy, Electron, Transmission , Models, Molecular , Mutation/genetics , Neuromuscular Junction/drug effects , Neuromuscular Junction/ultrastructure , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Serine/genetics , Serine/metabolism , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Synaptic Vesicles/drug effects , Synaptic Vesicles/physiology , Transfection
20.
Neuropharmacology ; 58(1): 135-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19573542

ABSTRACT

CRF-induced ERK phosphorylation has been shown to be an important mechanism underlying expression of pro-opiomelanocortin, a key precursor molecule in the hypothalamic pituitary adrenal axis. In AtT20 cells, CRF signalling has been investigated but the mechanism behind CRF-induced ERK activity is not fully understood. This paper elucidates the signalling cascade involved in this phenomenon. Involvement of CRF(1) receptor on ERK phosphorylation was shown by using CRF and urocortin 1. The lack of inhibitory effect of pertussis toxin and BAPTA-AM excluded involvement of G(i)-coupling and calcium mobilization respectively. In contrast, the process is suggested to be driven by cAMP since treatment of AtT20 cells with forskolin triggered strong ERK phosphorylation. Treatment with PKA inhibitors had a minor effect on CRF-induced ERK signalling while phosphorylation of CREB was completely abolished. This ruled out involvement of PKA and suggested a role for exchange protein directly activated by cAMP (EPAC). Moreover, an activator of EPACs 8-(4-methoxyphenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate mimicked CRF-induced ERK phosphorylation. Gene expression analysis showed high levels of EPAC2 mRNA and protein but low levels of EPAC1. Knockdown of EPAC2 expression by the use of specific siRNAs abolished CRF- and forskolin-induced ERK phosphorylation. The current study demonstrates a clear cAMP-dependent but PKA-independent mechanism underlying CRF-induced ERK activity that proceeds via EPAC2 signalling. Further research will provide more insight in the role of EPAC2 in CRF signalling.


Subject(s)
Carrier Proteins/metabolism , Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Animals , Calcium/metabolism , Carrier Proteins/genetics , Cell Line , Chelating Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...