62 results on '"Jankowsky JL"'
Search Results
2. Erratum: Wild-type microglia do not reverse pathology in mouse models of Rett syndrome (Nature (2015) 521 (E1-E4) DOI:10.1038/nature14444)
- Author
-
Wang, J, Wegener, JE, Huang, TW, Sripathy, S, De Jesus-Cortes, H, Xu, P, Tran, S, Knobbe, W, Leko, V, Britt, J, Starwalt, R, McDaniel, L, Ward, CS, Parra, D, Newcomb, B, Lao, U, Nourigat, C, Flowers, DA, Cullen, S, Jorstad, NL, Yang, Y, Glaskova, L, Vigneau, S, Kozlitina, J, Yetman, MJ, Jankowsky, JL, Reichardt, SD, Reichardt, HM, Gartner, J, Bartolomei, MS, Fang, M, Loeb, K, Keene, CD, Bernstein, I, Goodell, M, Brat, DJ, Huppke, P, Neul, JL, Bedalov, A, and Pieper, AA
- Published
- 2015
3. Wild-type microglia do not reverse pathology in mouse models of Rett syndrome
- Author
-
Wang, J, Wegener, JE, Huang, TW, Sripathy, S, De Jesus-Cortes, H, Xu, P, Tran, S, Knobbe, W, Leko, V, Britt, J, Starwalt, R, McDaniel, L, Ward, CS, Parra, D, Newcomb, B, Lao, U, Nourigat, C, Flowers, DA, Cullen, S, Jorstad, NL, Yang, Y, Glaskova, L, Vigneau, S, Kozlitina, J, Yetman, MJ, Jankowsky, JL, Reichardt, SD, Reichardt, HM, Gärtner, J, Bartolomei, MS, Fang, M, Loeb, K, Keene, CD, Bernstein, I, Goodell, M, Brat, DJ, Huppke, P, Neul, JL, Bedalov, A, and Pieper, AA
- Subjects
Male ,Pediatric ,Transplantation ,Methyl-CpG-Binding Protein 2 ,General Science & Technology ,Prevention ,Neurosciences ,Hematology ,Neurodegenerative ,Stem Cell Research ,Brain Disorders ,Congenital ,Rare Diseases ,Rett Syndrome ,Disease Progression ,Genetics ,Animals ,2.1 Biological and endogenous factors ,Female ,Stem Cell Research - Nonembryonic - Non-Human ,Microglia ,Aetiology - Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by mutations in the X chromosomal gene Methyl-CpG-binding Protein 2 (MECP2) (1). RTT treatment so far is symptomatic. Mecp2 disruption in mice phenocopies major features of the syndrome (2) that can be reversed upon re-expression of Mecp2 (3. It has recently been reported that transplantation of wild type (WT) bone marrow (BMT) into lethally irradiated Mecp2tm1.1Jae/y mice prevented neurologic decline and early death by restoring microglial phagocytic activity against apoptotic targets (4). Based on this report, clinical trials of BMT for patients with RTT have been initiated (5). We aimed to replicate and extend the BMT experiments in three different RTT mouse models but found that despite robust microglial engraftment, BMT from WT donors did not rescue early death or ameliorate neurologic deficits. Furthermore, early and specific genetic expression of Mecp2 in microglia did not rescue Mecp2-deficient mice. In conclusion our experiments do not support BMT as therapy for RTT.
- Published
- 2015
4. Chemogenetic neuronal silencing decouples c-Jun activation from cell death in the temporal cortex.
- Author
-
Wood CA, Somasundaram P, Dundee JM, Rudy MA, Watkins TA, and Jankowsky JL
- Abstract
Initial symptoms of neurodegenerative diseases are often defined by the loss of the most vulnerable neural populations specific to each disorder. In the early stages of Alzheimer's disease, vulnerable circuits in the temporal lobe exhibit diminished activity prior to overt degeneration. It remains unclear whether these functional changes contribute to regional vulnerability or are simply a consequence of pathology. We previously found that entorhinal neurons in the temporal cortex undergo cell death following transient suppression of electrical activity, suggesting a causal role for activity disruption in neurodegeneration. Here we demonstrate that electrical arrest of this circuit stimulates the injury-response transcription factor c-Jun. Entorhinal silencing induces transcriptional changes consistent with c-Jun activation that share characteristics of gene signatures in other neuronal populations vulnerable to Alzheimer's disease. Despite its established role in the neuronal injury response, inhibiting c-Jun failed to ameliorate entorhinal degeneration following activity disruption. Finally, we present preliminary evidence of integrated stress response activity that may serve as an alternative hypothesis to what drives entorhinal degeneration after silencing. Our data demonstrate that c-Jun is activated in response to neuronal silencing in the entorhinal cortex but is decoupled from subsequent neurodegeneration., (© 2024 Federation of European Neuroscience Societies and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
5. Simple improvements in vector design afford substantial gains in AAV delivery of aggregation-slowing Aβ variants.
- Author
-
Borgenheimer E, Trueblood C, Nguyen BL, Lagor WR, and Jankowsky JL
- Abstract
Adeno-associated virus (AAV) gene therapy for neurological disease has gained traction due to stunning advances in capsid evolution for CNS targeting. With AAV brain delivery now in focus, conventional improvements in viral expression vectors offer a complementary route for optimizing gene delivery. We previously introduced a novel AAV gene therapy to slow amyloid aggregation in the brain based on neuronal release of an Aβ sequence variant that inhibited fibrilization of wild-type Aβ. Here we explore three coding elements of the virally delivered DNA plasmid in an effort to maximize the production of therapeutic peptide in the brain. We demonstrate that simply replacing the Gaussia luciferase signal peptide with the mouse immunoglobulin heavy chain signal peptide increased release of variant Aβ by ∼5-fold. Sequence modifications within the expressed minigene further increased peptide release by promoting γ-secretase cleavage. Addition of a cytosolic fusion tag compatible with γ-secretase interaction allowed viral transduction to be tracked by immunostaining, independent from the variant Aβ peptide. Collectively these construct modifications increased neuronal production of therapeutic peptide by 10-fold upon intracranial AAV injection of neonatal mice. These findings demonstrate that modest changes in expression vector design can yield substantial gains in AAV efficiency for therapeutic applications., Competing Interests: The authors declare no competing interests. C.T. is currently a paid employee of Eli Lilly and Co. but was not affiliated with this company or any for-profit entity at the time she contributed to this study. J.L.J. is a co-inventor on pending patent application WO 202213346, Delivery of Abeta variants for aggregation inhibition., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. TMEM106B coding variant is protective and deletion detrimental in a mouse model of tauopathy.
- Author
-
Edwards GA 3rd, Wood CA, He Y, Nguyen Q, Kim PJ, Gomez-Gutierrez R, Park KW, Xu Y, Zurhellen C, Al-Ramahi I, and Jankowsky JL
- Subjects
- Animals, Humans, Mice, Disease Models, Animal, Mice, Knockout, Mice, Transgenic, Mutation, Paralysis genetics, Polymorphism, Single Nucleotide, tau Proteins genetics, tau Proteins metabolism, Membrane Proteins genetics, Nerve Tissue Proteins genetics, Tauopathies pathology
- Abstract
TMEM106B is a risk modifier of multiple neurological conditions, where a single coding variant and multiple non-coding SNPs influence the balance between susceptibility and resilience. Two key questions that emerge from past work are whether the lone T185S coding variant contributes to protection, and if the presence of TMEM106B is helpful or harmful in the context of disease. Here, we address both questions while expanding the scope of TMEM106B study from TDP-43 to models of tauopathy. We generated knockout mice with constitutive deletion of TMEM106B, alongside knock-in mice encoding the T186S knock-in mutation (equivalent to the human T185S variant), and crossed both with a P301S transgenic tau model to study how these manipulations impacted disease phenotypes. We found that TMEM106B deletion accelerated cognitive decline, hind limb paralysis, tau pathology, and neurodegeneration. TMEM106B deletion also increased transcriptional correlation with human AD and the functional pathways enriched in KO:tau mice aligned with those of AD. In contrast, the coding variant protected against tau-associated cognitive decline, synaptic impairment, neurodegeneration, and paralysis without affecting tau pathology. Our findings reveal that TMEM106B is a critical safeguard against tau aggregation, and that loss of this protein has a profound effect on sequelae of tauopathy. Our study further demonstrates that the coding variant is functionally relevant and contributes to neuroprotection downstream of tau pathology to preserve cognitive function., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
7. Generation of a Dcx-CreER T2 knock-in mouse for genetic manipulation of newborn neurons.
- Author
-
Perez GA, Park KW, Lanza D, Cicardo J, Uddin MD, and Jankowsky JL
- Subjects
- Mice, Animals, Mice, Transgenic, Neurogenesis genetics, Brain, Neurons metabolism, Neural Stem Cells metabolism
- Abstract
A wide variety of CreER
T2 driver lines are available for genetic manipulation of adult-born neurons in the mouse brain. These tools have been instrumental in studying fate potential, migration, circuit integration, and morphology of the stem cells supporting lifelong neurogenesis. Despite a wealth of tools, genetic manipulation of adult-born neurons for circuit and behavioral studies has been limited by poor specificity of many driver lines targeting early progenitor cells and by the inaccessibility of lines selective for later stages of neuronal maturation. We sought to address these limitations by creating a new CreERT2 driver line targeted to the endogenous mouse doublecortin locus as a marker of fate-specified neuroblasts and immature neurons. Our new model places a T2A-CreERT2 cassette immediately downstream of the Dcx coding sequence on the X chromosome, allowing expression of both Dcx and CreERT2 proteins in the endogenous spatiotemporal pattern for this gene. We demonstrate that the new mouse line drives expression of a Cre-dependent reporter throughout the brain in neonatal mice and in known neurogenic niches of adult animals. The line has been deposited with the Jackson Laboratory and should provide an accessible tool for studies targeting fate-restricted neuronal precursors., (© 2023 Wiley Periodicals LLC.)- Published
- 2024
- Full Text
- View/download PDF
8. Doxycycline for transgene control disrupts gut microbiome diversity without compromising acute neuroinflammatory response.
- Author
-
Koller EJ, Wood CA, Lai Z, Borgenheimer E, Hoffman KL, and Jankowsky JL
- Subjects
- Mice, Animals, Mice, Transgenic, Lipopolysaccharides, Tetracycline pharmacology, Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Trans-Activators genetics, Inflammation, Transgenes, Doxycycline pharmacology, Gastrointestinal Microbiome
- Abstract
The tetracycline transactivator (tTA) system provides controllable transgene expression through oral administration of the broad-spectrum antibiotic doxycycline. Antibiotic treatment for transgene control in mouse models of disease might have undesirable systemic effects resulting from changes in the gut microbiome. Here we assessed the impact of doxycycline on gut microbiome diversity in a tTA-controlled model of Alzheimer's disease and then examined neuroimmune effects of these microbiome alterations following acute LPS challenge. We show that doxycycline decreased microbiome diversity in both transgenic and wild-type mice and that these changes persisted long after drug withdrawal. Despite the change in microbiome composition, doxycycline treatment had minimal effect on basal transcriptional signatures of inflammation the brain or on the neuroimmune response to LPS challenge. Our findings suggest that central neuroimmune responses may be less affected by doxycycline at doses needed for transgene control than by antibiotic cocktails at doses used for experimental microbiome disruption., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. An automated respiratory data pipeline for waveform characteristic analysis.
- Author
-
Lusk S, Ward CS, Chang A, Twitchell-Heyne A, Fattig S, Allen G, Jankowsky JL, and Ray RS
- Subjects
- Animals, Mice, Humans, Longitudinal Studies, Plethysmography, Software, Respiration
- Abstract
Comprehensive and accurate analysis of respiratory and metabolic data is crucial to modelling congenital, pathogenic and degenerative diseases converging on autonomic control failure. A lack of tools for high-throughput analysis of respiratory datasets remains a major challenge. We present Breathe Easy, a novel open-source pipeline for processing raw recordings and associated metadata into operative outcomes, publication-worthy graphs and robust statistical analyses including QQ and residual plots for assumption queries and data transformations. This pipeline uses a facile graphical user interface for uploading data files, setting waveform feature thresholds and defining experimental variables. Breathe Easy was validated against manual selection by experts, which represents the current standard in the field. We demonstrate Breathe Easy's utility by examining a 2-year longitudinal study of an Alzheimer's disease mouse model to assess contributions of forebrain pathology in disordered breathing. Whole body plethysmography has become an important experimental outcome measure for a variety of diseases with primary and secondary respiratory indications. Respiratory dysfunction, while not an initial symptom in many of these disorders, often drives disability or death in patient outcomes. Breathe Easy provides an open-source respiratory analysis tool for all respiratory datasets and represents a necessary improvement upon current analytical methods in the field. KEY POINTS: Respiratory dysfunction is a common endpoint for disability and mortality in many disorders throughout life. Whole body plethysmography in rodents represents a high face-value method for measuring respiratory outcomes in rodent models of these diseases and disorders. Analysis of key respiratory variables remains hindered by manual annotation and analysis that leads to low throughput results that often exclude a majority of the recorded data. Here we present a software suite, Breathe Easy, that automates the process of data selection from raw recordings derived from plethysmography experiments and the analysis of these data into operative outcomes and publication-worthy graphs with statistics. We validate Breathe Easy with a terabyte-scale Alzheimer's dataset that examines the effects of forebrain pathology on respiratory function over 2 years of degeneration., (© 2023 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society.)
- Published
- 2023
- Full Text
- View/download PDF
10. The TMEM106B T186S coding variant increases neurite arborization and synaptic density in primary hippocampal neurons.
- Author
-
Nguyen Q, Wood CA, Kim PJ, and Jankowsky JL
- Abstract
The lysosomal protein TMEM106B was identified as a risk modifier of multiple dementias including frontotemporal dementia and Alzheimer's disease. The gene comes in two major haplotypes, one associated with disease risk, and by comparison, the other with resilience. Only one coding polymorphism distinguishes the two alleles, a threonine-to-serine substitution at residue 185 (186 in mouse), that is inherited in disequilibrium with multiple non-coding variants. Transcriptional studies suggest synaptic, neuronal, and cognitive preservation in human subjects with the protective haplotype, while murine in vitro studies reveal dramatic effects of TMEM106B deletion on neuronal development. Despite this foundation, the field has not yet resolved whether coding variant is biologically meaningful, and if so, whether it has any specific effect on neuronal phenotypes. Here we studied how loss of TMEM106B or expression of the lone coding variant in isolation affected transcriptional signatures in the mature brain and neuronal structure during development in primary neurons. Homozygous expression of the TMEM106B T186S variant in knock-in mice increased cortical expression of genes associated with excitatory synaptic function and axon outgrowth, and promoted neurite branching, dendritic spine density, and synaptic density in primary hippocampal neurons. In contrast, constitutive TMEM106B deletion affected transcriptional signatures of myelination without altering neuronal development in vitro . Our findings show that the T186S variant is functionally relevant and may contribute to disease resilience during neurodevelopment., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Nguyen, Wood, Kim and Jankowsky.)
- Published
- 2023
- Full Text
- View/download PDF
11. TMEM106B regulates microglial proliferation and survival in response to demyelination.
- Author
-
Zhang T, Pang W, Feng T, Guo J, Wu K, Nunez Santos M, Arthanarisami A, Nana AL, Nguyen Q, Kim PJ, Jankowsky JL, Seeley WW, and Hu F
- Subjects
- Humans, Mice, Animals, Mice, Knockout, Brain metabolism, Cell Proliferation, Membrane Proteins genetics, Membrane Proteins metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Membrane Glycoproteins metabolism, Receptors, Immunologic metabolism, Microglia metabolism, Demyelinating Diseases genetics, Demyelinating Diseases metabolism
- Abstract
TMEM106B, a lysosomal transmembrane protein, has been closely associated with brain health. Recently, an intriguing link between TMEM106B and brain inflammation has been discovered, but how TMEM106B regulates inflammation is unknown. Here, we report that TMEM106B deficiency in mice leads to reduced microglia proliferation and activation and increased microglial apoptosis in response to demyelination. We also found an increase in lysosomal pH and a decrease in lysosomal enzyme activities in TMEM106B-deficient microglia. Furthermore, TMEM106B loss results in a significant decrease in the protein levels of TREM2, an innate immune receptor essential for microglia survival and activation. Specific ablation of TMEM106B in microglia results in similar microglial phenotypes and myelination defects in mice, supporting the idea that microglial TMEM106B is critical for proper microglial activities and myelination. Moreover, the TMEM106B risk allele is associated with myelin loss and decreased microglial numbers in humans. Collectively, our study unveils a previously unknown role of TMEM106B in promoting microglial functionality during demyelination.
- Published
- 2023
- Full Text
- View/download PDF
12. TMEM106B coding variant is protective and deletion detrimental in a mouse model of tauopathy.
- Author
-
Edwards GA, Wood CA, Nguyen Q, Kim PJ, Gomez-Gutierrez R, Park KW, Zurhellen C, Al-Ramahi I, and Jankowsky JL
- Abstract
TMEM106B is a risk modifier for a growing list of age-associated dementias including Alzheimer’s and frontotemporal dementia, yet its function remains elusive. Two key questions that emerge from past work are whether the conservative T185S coding variant found in the minor haplotype contributes to protection, and whether the presence of TMEM106B is helpful or harmful in the context of disease. Here we address both issues while extending the testbed for study of TMEM106B from models of TDP to tauopathy. We show that TMEM106B deletion accelerates cognitive decline, hindlimb paralysis, neuropathology, and neurodegeneration. TMEM106B deletion also increases transcriptional overlap with human AD, making it a better model of disease than tau alone. In contrast, the coding variant protects against tau-associated cognitive decline, neurodegeneration, and paralysis without affecting tau pathology. Our findings show that the coding variant contributes to neuroprotection and suggest that TMEM106B is a critical safeguard against tau aggregation.
- Published
- 2023
- Full Text
- View/download PDF
13. Correction: Temporal and spatially controlled APP transgene expression using Cre-dependent alleles.
- Author
-
Koller EJ, Comstock M, Bean JC, Escobedo G, Park KW, and Jankowsky JL
- Published
- 2023
- Full Text
- View/download PDF
14. Activity disruption causes degeneration of entorhinal neurons in a mouse model of Alzheimer's circuit dysfunction.
- Author
-
Zhao R, Grunke SD, Wood CA, Perez GA, Comstock M, Li MH, Singh AK, Park KW, and Jankowsky JL
- Subjects
- Mice, Animals, Neurons physiology, Hippocampus metabolism, Entorhinal Cortex, Alzheimer Disease metabolism
- Abstract
Neurodegenerative diseases are characterized by selective vulnerability of distinct cell populations; however, the cause for this specificity remains elusive. Here, we show that entorhinal cortex layer 2 (EC2) neurons are unusually vulnerable to prolonged neuronal inactivity compared with neighboring regions of the temporal lobe, and that reelin + stellate cells connecting EC with the hippocampus are preferentially susceptible within the EC2 population. We demonstrate that neuronal death after silencing can be elicited through multiple independent means of activity inhibition, and that preventing synaptic release, either alone or in combination with electrical shunting, is sufficient to elicit silencing-induced degeneration. Finally, we discovered that degeneration following synaptic silencing is governed by competition between active and inactive cells, which is a circuit refinement process traditionally thought to end early in postnatal life. Our data suggests that the developmental window for wholesale circuit plasticity may extend into adulthood for specific brain regions. We speculate that this sustained potential for remodeling by entorhinal neurons may support lifelong memory but renders them vulnerable to prolonged activity changes in disease., Competing Interests: RZ, SG, CW, GP, MC, ML, AS, KP, JJ No competing interests declared, (© 2022, Zhao, Grunke, Wood et al.)
- Published
- 2022
- Full Text
- View/download PDF
15. Temporal and spatially controlled APP transgene expression using Cre-dependent alleles.
- Author
-
Koller EJ, Comstock M, Bean JC, Escobedo G, Park KW, and Jankowsky JL
- Subjects
- Alleles, Animals, Humans, Mice, Mice, Transgenic, Tetracycline pharmacology, Transgenes, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Integrases metabolism
- Abstract
Although a large number of mouse models have been made to study Alzheimer's disease, only a handful allow experimental control over the location or timing of the protein being used to drive pathology. Other fields have used the Cre and the tamoxifen-inducible CreER driver lines to achieve precise spatial and temporal control over gene deletion and transgene expression, yet these tools have not been widely used in studies of neurodegeneration. Here, we describe two strategies for harnessing the wide range of Cre and CreER driver lines to control expression of disease-associated amyloid precursor protein (APP) in modeling Alzheimer's amyloid pathology. We show that CreER-based spatial and temporal control over APP expression can be achieved with existing lines by combining a Cre driver with a tetracycline-transactivator (tTA)-dependent APP responder using a Cre-to-tTA converter line. We then describe a new mouse line that places APP expression under direct control of Cre recombinase using an intervening lox-stop-lox cassette. Mating this allele with a CreER driver allows both spatial and temporal control over APP expression, and with it, amyloid onset. This article has an associated First Person interview with the first author of the paper., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
16. Gene therapy using Aβ variants for amyloid reduction.
- Author
-
Park KW, Wood CA, Li J, Taylor BC, Oh S, Young NL, and Jankowsky JL
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease pathology, Amyloid beta-Peptides genetics, Amyloid beta-Peptides metabolism, Animals, Dependovirus genetics, Female, Genetic Vectors genetics, Humans, Male, Mice, Mice, Inbred ICR, Mice, Transgenic, Plaque, Amyloid genetics, Plaque, Amyloid metabolism, Alzheimer Disease therapy, Amyloid beta-Peptides antagonists & inhibitors, Brain metabolism, Genetic Therapy, Genetic Vectors administration & dosage, Mutation, Plaque, Amyloid therapy
- Abstract
Numerous aggregation inhibitors have been developed with the goal of blocking or reversing toxic amyloid formation in vivo. Previous studies have used short peptide inhibitors targeting different amyloid β (Aβ) amyloidogenic regions to prevent aggregation. Despite the specificity that can be achieved by peptide inhibitors, translation of these strategies has been thwarted by two key obstacles: rapid proteolytic degradation in the bloodstream and poor transfer across the blood-brain barrier. To circumvent these problems, we have created a minigene to express full-length Aβ variants in the mouse brain. We identify two variants, F20P and F19D/L34P, that display four key properties required for therapeutic use: neither peptide aggregates on its own, both inhibit aggregation of wild-type Aβ in vitro, promote disassembly of pre-formed fibrils, and diminish toxicity of Aβ oligomers. We used intraventricular injection of adeno-associated virus (AAV) to express each variant in APP/PS1 transgenic mice. Lifelong expression of F20P, but not F19D/L34P, diminished Aβ levels, plaque burden, and plaque-associated neuroinflammation. Our findings suggest that AAV delivery of Aβ variants may offer a novel therapeutic strategy for Alzheimer's disease. More broadly our work offers a framework for identifying and delivering peptide inhibitors tailored to other protein-misfolding diseases., Competing Interests: Declaration of interests K.-W.P. completed the bulk of this study while employed at Baylor College of Medicine performing work that was fully funded by NIH and a non-profit research foundation. In July 2020, K.-W.P. left Baylor to become CEO of Aβrain, a biotechnology company he founded to pursue AAV-based therapeutics for protein aggregation disorders. He is currently a paid employee and sole owner of Aβrain. The other authors declare no competing interests., (Copyright © 2021 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
17. Type I interferon response drives neuroinflammation and synapse loss in Alzheimer disease.
- Author
-
Roy ER, Wang B, Wan YW, Chiu G, Cole A, Yin Z, Propson NE, Xu Y, Jankowsky JL, Liu Z, Lee VM, Trojanowski JQ, Ginsberg SD, Butovsky O, Zheng H, and Cao W
- Subjects
- Alzheimer Disease chemically induced, Alzheimer Disease pathology, Animals, Complement C3 immunology, Disease Models, Animal, Humans, Inflammation chemically induced, Inflammation immunology, Inflammation pathology, Interferon-beta adverse effects, Interferon-beta pharmacology, Mice, Microglia immunology, Microglia pathology, Synapses pathology, Up-Regulation drug effects, Up-Regulation immunology, Alzheimer Disease immunology, Amyloid immunology, Interferon-beta immunology, Synapses immunology
- Abstract
Type I interferon (IFN) is a key cytokine that curbs viral infection and cell malignancy. Previously, we demonstrated a potent IFN immunogenicity of nucleic acid-containing (NA-containing) amyloid fibrils in the periphery. Here, we investigated whether IFN is associated with β-amyloidosis inside the brain and contributes to neuropathology. An IFN-stimulated gene (ISG) signature was detected in the brains of multiple murine Alzheimer disease (AD) models, a phenomenon also observed in WT mouse brain challenged with generic NA-containing amyloid fibrils. In vitro, microglia innately responded to NA-containing amyloid fibrils. In AD models, activated ISG-expressing microglia exclusively surrounded NA+ amyloid β plaques, which accumulated in an age-dependent manner. Brain administration of rIFN-β resulted in microglial activation and complement C3-dependent synapse elimination in vivo. Conversely, selective IFN receptor blockade effectively diminished the ongoing microgliosis and synapse loss in AD models. Moreover, we detected activated ISG-expressing microglia enveloping NA-containing neuritic plaques in postmortem brains of patients with AD. Gene expression interrogation revealed that IFN pathway was grossly upregulated in clinical AD and significantly correlated with disease severity and complement activation. Therefore, IFN constitutes a pivotal element within the neuroinflammatory network of AD and critically contributes to neuropathogenic processes.
- Published
- 2020
- Full Text
- View/download PDF
18. Cross-species genetic screens to identify kinase targets for APP reduction in Alzheimer's disease.
- Author
-
Huichalaf CH, Al-Ramahi I, Park KW, Grunke SD, Lu N, de Haro M, El-Zein K, Gallego-Flores T, Perez AM, Jung SY, Botas J, Zoghbi HY, and Jankowsky JL
- Subjects
- Alzheimer Disease genetics, Amyloidosis therapy, Animals, Brain metabolism, Brain pathology, Cell Line, Tumor, Disease Models, Animal, Drosophila, Genetic Testing, Genetic Therapy, Humans, Mice, Mice, Transgenic, NIH 3T3 Cells, Phosphorylation, Plaque, Amyloid pathology, Protein Kinase C beta genetics, Protein Kinase C beta metabolism, Protein Kinases genetics, Protein Kinases metabolism, RNA Interference, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Amyloid beta-Protein Precursor metabolism, Amyloidosis metabolism, Protein Kinase C beta antagonists & inhibitors
- Abstract
An early hallmark of Alzheimer's disease is the accumulation of amyloid-β (Aβ), inspiring numerous therapeutic strategies targeting this peptide. An alternative approach is to destabilize the amyloid beta precursor protein (APP) from which Aβ is derived. We interrogated innate pathways governing APP stability using a siRNA screen for modifiers whose own reduction diminished APP in human cell lines and transgenic Drosophila. As proof of principle, we validated PKCβ-a known modifier identified by the screen-in an APP transgenic mouse model. PKCβ was genetically targeted using a novel adeno-associated virus shuttle vector to deliver microRNA-adapted shRNA via intracranial injection. In vivo reduction of PKCβ initially diminished APP and delayed plaque formation. Despite persistent PKCβ suppression, the effect on APP and amyloid diminished over time. Our study advances this approach for mining druggable modifiers of disease-associated proteins, while cautioning that prolonged in vivo validation may be needed to reveal emergent limitations on efficacy., (© The Author(s) 2019. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
19. Brain-wide distribution of reporter expression in five transgenic tetracycline-transactivator mouse lines.
- Author
-
Lillehaug S, Yetman MJ, Puchades MA, Checinska MM, Kleven H, Jankowsky JL, Bjaalie JG, and Leergaard TB
- Subjects
- Animals, Imaging, Three-Dimensional, Mice, Promoter Regions, Genetic, Tetracycline, Trans-Activators physiology, Brain Mapping methods, Gene Expression Regulation physiology, Genes, Reporter, Mice, Transgenic anatomy & histology, Mice, Transgenic genetics
- Abstract
The spatial pattern of transgene expression in tetracycline-controlled mouse models is governed primarily by the driver line used to introduce the tetracycline-controlled transactivator (tTA). Detailed maps showing where each tTA driver activates expression are therefore essential for designing and using tet-regulated models, particularly in brain research where cell type and regional specificity determine the circuits affected by conditional gene expression. We have compiled a comprehensive online repository of serial microscopic images showing brain-wide reporter expression for five commonly used tTA driver lines. We have spatially registered all images to a common three-dimensional mouse brain anatomical reference atlas for direct comparison of spatial distribution across lines. The high-resolution images and associated metadata are shared via the web page of the EU Human Brain Project. Images can be inspected using an interactive viewing tool that includes an optional overlay feature providing anatomical delineations and reference atlas coordinates. Interactive viewing is supplemented by semi-quantitative analyses of expression levels within anatomical subregions for each tTA driver line.
- Published
- 2019
- Full Text
- View/download PDF
20. Combination anti-Aβ treatment maximizes cognitive recovery and rebalances mTOR signaling in APP mice.
- Author
-
Chiang ACA, Fowler SW, Savjani RR, Hilsenbeck SG, Wallace CE, Cirrito JR, Das P, and Jankowsky JL
- Subjects
- Amyloid beta-Peptides metabolism, Animals, Axons metabolism, Behavior, Animal, Biomarkers metabolism, Drug Therapy, Combination, Immunization, Passive, Lysosomes metabolism, Mice, Inbred C57BL, Mice, Transgenic, Plaque, Amyloid metabolism, Plaque, Amyloid pathology, Solubility, Synapses metabolism, Transgenes, Alzheimer Disease drug therapy, Alzheimer Disease physiopathology, Amyloid beta-Peptides antagonists & inhibitors, Cognition, Signal Transduction, TOR Serine-Threonine Kinases metabolism
- Abstract
Drug development for Alzheimer's disease has endeavored to lower amyloid β (Aβ) by either blocking production or promoting clearance. The benefit of combining these approaches has been examined in mouse models and shown to improve pathological measures of disease over single treatment; however, the impact on cellular and cognitive functions affected by Aβ has not been tested. We used a controllable APP transgenic mouse model to test whether combining genetic suppression of Aβ production with passive anti-Aβ immunization improved functional outcomes over either treatment alone. Compared with behavior before treatment, arresting further Aβ production (but not passive immunization) was sufficient to stop further decline in spatial learning, working memory, and associative memory, whereas combination treatment reversed each of these impairments. Cognitive improvement coincided with resolution of neuritic dystrophy, restoration of synaptic density surrounding deposits, and reduction of hyperactive mammalian target of rapamycin signaling. Computational modeling corroborated by in vivo microdialysis pointed to the reduction of soluble/exchangeable Aβ as the primary driver of cognitive recovery., (© 2018 Chiang et al.)
- Published
- 2018
- Full Text
- View/download PDF
21. Discrete Pools of Oligomeric Amyloid-β Track with Spatial Learning Deficits in a Mouse Model of Alzheimer Amyloidosis.
- Author
-
Chiang ACA, Fowler SW, Reddy R, Pletnikova O, Troncoso JC, Sherman MA, Lesne SE, and Jankowsky JL
- Subjects
- Aged, Aged, 80 and over, Alzheimer Disease pathology, Animals, Brain pathology, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Transgenic, Middle Aged, Motor Activity physiology, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Brain metabolism, Spatial Learning physiology, Spatial Memory physiology
- Abstract
Despite increasing appreciation that oligomeric amyloid-β (Aβ) may contribute to cognitive decline of Alzheimer disease, defining the most critical forms has been thwarted by the changeable nature of these aggregates and the varying methods used for detection. Herein, using a broad approach, we quantified Aβ oligomers during the evolution of cognitive deficits in an aggressive model of Aβ amyloidosis. Amyloid precursor protein/tetracycline transactivator mice underwent behavioral testing at 3, 6, 9, and 12 months of age to evaluate spatial learning and memory, followed by histologic assessment of amyloid burden and biochemical characterization of oligomeric Aβ species. Transgenic mice displayed progressive impairments in acquisition and immediate recall of the trained platform location. Biochemical analysis of cortical extracts from behaviorally tested mice revealed distinct age-dependent patterns of accumulation in multiple oligomeric species. Dot blot analysis demonstrated that nonfibrillar Aβ oligomers were highly soluble and extracted into a fraction enriched for extracellular proteins, whereas prefibrillar species required high-detergent conditions to retrieve, consistent with membrane localization. Low-detergent extracts tested by 82E1 enzyme-linked immunosorbent assay confirmed the presence of bona fide Aβ oligomers, whereas immunoprecipitation-Western blotting using high-detergent extracts revealed a variety of SDS-stable low-n species. These findings show that different Aβ oligomers vary in solubility, consistent with distinct localization, and identify nonfibrillar Aβ oligomer-positive aggregates as tracking most closely with cognitive decline in this model., (Copyright © 2018 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
22. Practical considerations for choosing a mouse model of Alzheimer's disease.
- Author
-
Jankowsky JL and Zheng H
- Subjects
- Alzheimer Disease pathology, Amyloid beta-Protein Precursor genetics, Animals, Cerebral Amyloid Angiopathy genetics, Cognitive Dysfunction genetics, Humans, Mice, Mice, Knockout, Neurodegenerative Diseases genetics, Neurofibrillary Tangles, Plaque, Amyloid, Alzheimer Disease genetics, Disease Models, Animal, Mice, Transgenic
- Abstract
Alzheimer's disease (AD) is behaviorally identified by progressive memory impairment and pathologically characterized by the triad of β-amyloid plaques, neurofibrillary tangles, and neurodegeneration. Genetic mutations and risk factors have been identified that are either causal or modify the disease progression. These genetic and pathological features serve as basis for the creation and validation of mouse models of AD. Efforts made in the past quarter-century have produced over 100 genetically engineered mouse lines that recapitulate some aspects of AD clinicopathology. These models have been valuable resources for understanding genetic interactions that contribute to disease and cellular reactions that are engaged in response. Here we focus on mouse models that have been widely used stalwarts of the field or that are recently developed bellwethers of the future. Rather than providing a summary of each model, we endeavor to compare and contrast the genetic approaches employed and to discuss their respective advantages and limitations. We offer a critical account of the variables which may contribute to inconsistent findings and the factors that should be considered when choosing a model and interpreting the results. We hope to present an insightful review of current AD mouse models and to provide a practical guide for selecting models best matched to the experimental question at hand.
- Published
- 2017
- Full Text
- View/download PDF
23. Combination of Aβ Suppression and Innate Immune Activation in the Brain Significantly Attenuates Amyloid Plaque Deposition.
- Author
-
Verbeeck C, Carrano A, Chakrabarty P, Jankowsky JL, and Das P
- Subjects
- Alzheimer Disease immunology, Animals, Anti-Bacterial Agents pharmacology, Brain immunology, Brain pathology, Dependovirus, Doxycycline pharmacology, Genetic Therapy methods, Interleukin-6 administration & dosage, Interleukin-6 immunology, Mice, Mice, Transgenic, Alzheimer Disease pathology, Amyloid beta-Peptides antagonists & inhibitors, Brain drug effects, Immunity, Innate drug effects
- Abstract
Anti-Aβ clinical trials are currently under way to determine whether preventing amyloid deposition will be beneficial in arresting progression of Alzheimer disease. Both clinical and preclinical studies suggest that antiamyloid strategies are only effective if started at early stages of the disease process in a primary prevention strategy. Because this approach will be difficult to deploy, strategies for secondary prevention aimed at later stages of disease are also needed. In this study, we asked whether combining innate immune activation in the brain with concurrent Aβ suppression could enhance plaque clearance and could improve pathologic outcomes in mice with moderate amyloid pathologic disorder. Starting at 5 months of age, tet-off amyloid precursor protein transgenic mice were treated with doxycycline (dox) to suppress further amyloid precursor protein/Aβ production, and at the same time mice were intracranially injected with adeno-associated virus 1 expressing murine IL-6 (AAV1-mIL-6). Three months later, mice treated with the combination of Aβ suppression and AAV1-mIL-6 showed significantly less plaque pathologic disorder than dox or AAV1-mIL-6 only groups. The combination of AAV1-mIL-6 + dox treatment lowered total plaque burden by >60% versus untreated controls. Treatment with either dox or AAV1-mIL-6 alone was less effective than the combination. Our results suggest a synergistic mechanism by which the up-regulation of mIL-6 was able to improve plaque clearance in the setting of Aβ suppression., (Copyright © 2017 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
24. Neuronal overexpression of human VAPB slows motor impairment and neuromuscular denervation in a mouse model of ALS.
- Author
-
Kim JY, Jang A, Reddy R, Yoon WH, and Jankowsky JL
- Subjects
- Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Animals, Denervation, Disease Models, Animal, Female, Gene Expression Regulation, Humans, Male, Mice, Mice, Transgenic, Motor Neuron Disease genetics, Motor Neurons metabolism, Motor Neurons pathology, Mutation, Neuromuscular Diseases genetics, Neurons metabolism, Spinal Cord metabolism, Superoxide Dismutase-1 genetics, Vesicular Transport Proteins genetics, Vesicular Transport Proteins metabolism, Neuromuscular Diseases metabolism, Vesicular Transport Proteins biosynthesis
- Abstract
Four mutations in the VAMP/synaptobrevin-associated protein B (VAPB) gene have been linked to amyotrophic lateral sclerosis (ALS) type 8. The mechanism by which VAPB mutations cause motor neuron disease is unclear, but studies of the most common P56S variant suggest both loss of function and dominant-negative sequestration of wild-type protein. Diminished levels of VAPB and its proteolytic cleavage fragment have also been reported in sporadic ALS cases, suggesting that VAPB loss of function may be a common mechanism of disease. Here, we tested whether neuronal overexpression of wild-type human VAPB would attenuate disease in a mouse model of familial ALS1. We used neonatal intraventricular viral injections to express VAPB or YFP throughout the brain and spinal cord of superoxide dismutase (SOD1) G93A transgenic mice. Lifelong elevation of neuronal VAPB slowed the decline of neurological impairment, delayed denervation of hindlimb muscles, and prolonged survival of spinal motor neurons. Collectively, these changes produced a slight but significant extension in lifespan, even in this highly aggressive model of disease. Our findings lend support for a protective role of VAPB in neuromuscular health.
- Published
- 2016
- Full Text
- View/download PDF
25. Impaired Recall of Positional Memory following Chemogenetic Disruption of Place Field Stability.
- Author
-
Zhao R, Grunke SD, Keralapurath MM, Yetman MJ, Lam A, Lee TC, Sousounis K, Jiang Y, Swing DA, Tessarollo L, Ji D, and Jankowsky JL
- Subjects
- Animals, Entorhinal Cortex physiology, Female, Humans, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Models, Neurological, Perforant Pathway physiology, Hippocampus physiology, Nerve Net physiology, Spatial Memory physiology, Temporal Lobe physiology
- Abstract
The neural network of the temporal lobe is thought to provide a cognitive map of our surroundings. Functional analysis of this network has been hampered by coarse tools that often result in collateral damage to other circuits. We developed a chemogenetic system to temporally control electrical input into the hippocampus. When entorhinal input to the perforant path was acutely silenced, hippocampal firing patterns became destabilized and underwent extensive remapping. We also found that spatial memory acquired prior to neural silencing was impaired by loss of input through the perforant path. Together, our experiments show that manipulation of entorhinal activity destabilizes spatial coding and disrupts spatial memory. Moreover, we introduce a chemogenetic model for non-invasive neuronal silencing that offers multiple advantages over existing strategies in this setting., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Transgene expression in the Nop-tTA driver line is not inherently restricted to the entorhinal cortex.
- Author
-
Yetman MJ, Lillehaug S, Bjaalie JG, Leergaard TB, and Jankowsky JL
- Subjects
- Amyloid beta-Protein Precursor metabolism, Animals, Brain metabolism, Galactosides metabolism, Genes, Reporter, Humans, Immunohistochemistry, Indoles metabolism, Lac Operon, Mice, Mice, Inbred C57BL, Mice, Transgenic, Promoter Regions, Genetic, Entorhinal Cortex metabolism, Kallikreins metabolism, Tetracycline metabolism, Trans-Activators metabolism
- Abstract
The entorhinal cortex (EC) plays a central role in episodic memory and is among the earliest sites of neurodegeneration and neurofibrillary tangle formation in Alzheimer's disease. Given its importance in memory and dementia, the ability to selectively modulate gene expression or neuronal function in the EC is of widespread interest. To this end, several recent studies have taken advantage of a transgenic line in which the tetracycline transactivator (tTA) was placed under control of the neuropsin (Nop) promoter to limit transgene expression within the medial EC and pre-/parasubiculum. Although the utility of this driver is contingent on its spatial specificity, no detailed neuroanatomical analysis of its expression has yet been conducted. We therefore undertook a systematic analysis of Nop-tTA expression using a lacZ reporter and have made the complete set of histological sections available through the Rodent Brain Workbench tTA atlas, www.rbwb.org . Our findings confirm that the highest density of tTA expression is found in the EC and pre-/parasubiculum, but also reveal considerable expression in several other cortical areas. Promiscuous transgene expression may account for the appearance of pathological protein aggregates outside of the EC in mouse models of Alzheimer's disease using this driver, as we find considerable overlap between sites of delayed amyloid deposition and regions with sparse β-galactosidase reporter labeling. While different tet-responsive lines can display individual expression characteristics, our results suggest caution when designing experiments that depend on precise localization of gene products controlled by the Nop-tTA or other spatially restrictive transgenic drivers.
- Published
- 2016
- Full Text
- View/download PDF
27. Humanized Tau Mice with Regionalized Amyloid Exhibit Behavioral Deficits but No Pathological Interaction.
- Author
-
Yetman MJ, Fowler SW, and Jankowsky JL
- Subjects
- Amyloid genetics, Amyloid beta-Protein Precursor metabolism, Animals, Conditioning, Psychological, Fear psychology, Female, Gene Knockout Techniques, Humans, Male, Maze Learning, Mice, Mice, Transgenic, Phosphorylation, tau Proteins deficiency, tau Proteins metabolism, Amyloid metabolism, Behavior, Animal, tau Proteins genetics
- Abstract
Alzheimer's disease (AD) researchers have struggled for decades to draw a causal link between extracellular Aβ aggregation and intraneuronal accumulation of microtubule-associated protein tau. The amyloid cascade hypothesis posits that Aβ deposition promotes tau hyperphosphorylation, tangle formation, cell loss, vascular damage, and dementia. While the genetics of familial AD and the pathological staging of sporadic disease support this sequence of events, attempts to examine the molecular mechanism in transgenic animal models have largely relied on models of other inherited tauopathies as the basis for testing the interaction with Aβ. In an effort to more accurately model the relationship between Aβ and wild-type tau in AD, we intercrossed mice that overproduce human Aβ with a tau substitution model in which all 6 isoforms of the human protein are expressed in animals lacking murine tau. We selected an amyloid model in which pathology was biased towards the entorhinal region so that we could further examine whether the anticipated changes in tau phosphorylation occurred at the site of Aβ deposition or in synaptically connected regions. We found that Aβ and tau had independent effects on locomotion, learning, and memory, but found no behavioral evidence for an interaction between the two transgenes. Moreover, we saw no indication of amyloid-induced changes in the phosphorylation or aggregation of human tau either within the entorhinal area or elsewhere. These findings suggest that robust amyloid pathology within the medial temporal lobe has little effect on the metabolism of wild type human tau in this model.
- Published
- 2016
- Full Text
- View/download PDF
28. Astrocyte-Microglia Cross Talk through Complement Activation Modulates Amyloid Pathology in Mouse Models of Alzheimer's Disease.
- Author
-
Lian H, Litvinchuk A, Chiang AC, Aithmitti N, Jankowsky JL, and Zheng H
- Subjects
- Alzheimer Disease genetics, Amyloid beta-Protein Precursor genetics, Animals, Cells, Cultured, Complement C3 genetics, Complement C3 metabolism, Disease Models, Animal, Female, Glial Fibrillary Acidic Protein genetics, Glial Fibrillary Acidic Protein metabolism, Humans, I-kappa B Proteins genetics, I-kappa B Proteins metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phagocytosis drug effects, Phagocytosis genetics, Presenilin-1 genetics, Signal Transduction drug effects, Signal Transduction genetics, Up-Regulation genetics, Alzheimer Disease immunology, Alzheimer Disease pathology, Astrocytes metabolism, Complement Activation genetics, Microglia metabolism
- Abstract
Increasing evidence supports a role of neuroinflammation in the pathogenesis of Alzheimer's disease (AD). Previously, we identified a neuron-glia signaling pathway whereby Aβ acts as an upstream activator of astroglial nuclear factor kappa B (NF-κB), leading to the release of complement C3, which acts on the neuronal C3a receptor (C3aR) to influence dendritic morphology and cognitive function. Here we report that astrocytic complement activation also regulates Aβ dynamics in vitro and amyloid pathology in AD mouse models through microglial C3aR. We show that in primary microglial cultures, acute C3 or C3a activation promotes, whereas chronic C3/C3a treatment attenuates, microglial phagocytosis and that the effect of chronic C3 exposure can be blocked by cotreatment with a C3aR antagonist and by genetic deletion of C3aR. We further demonstrate that Aβ pathology and neuroinflammation in amyloid precursor protein (APP) transgenic mice are worsened by astroglial NF-κB hyperactivation and resulting C3 elevation, whereas treatment with the C3aR antagonist (C3aRA) ameliorates plaque load and microgliosis. Our studies define a complement-dependent intercellular cross talk in which neuronal overproduction of Aβ activates astroglial NF-κB to elicit extracellular release of C3. This promotes a pathogenic cycle by which C3 in turn interacts with neuronal and microglial C3aR to alter cognitive function and impair Aβ phagocytosis. This feedforward loop can be effectively blocked by C3aR inhibition, supporting the therapeutic potential of C3aR antagonists under chronic neuroinflammation conditions., Significance Statement: The complement pathway is activated in Alzheimer's disease. Here we show that the central complement factor C3 secreted from astrocytes interacts with microglial C3a receptor (C3aR) to mediate β-amyloid pathology and neuroinflammation in AD mouse models. Our study provides support for targeting C3aR as a potential therapy for Alzheimer's disease., (Copyright © 2016 the authors 0270-6474/16/360577-13$15.00/0.)
- Published
- 2016
- Full Text
- View/download PDF
29. Widespread Neuronal Transduction of the Rodent CNS via Neonatal Viral Injection.
- Author
-
Kim JY, Grunke SD, and Jankowsky JL
- Subjects
- Animals, Animals, Newborn virology, Dependovirus genetics, Genetic Vectors administration & dosage, Injections, Intraventricular, Mice, Dependovirus physiology, Neurons metabolism, Transduction, Genetic
- Abstract
The rapid pace of neuroscience research demands equally efficient and flexible methods for genetically manipulating and visualizing selected neurons within the rodent brain. The use of viral vectors for gene delivery saves the time and cost of traditional germline transgenesis and offers the versatility of readily available reagents that can be easily customized to meet individual experimental needs. Here, we present a protocol for widespread neuronal transduction based on intraventricular viral injection of the neonatal mouse brain. Injections can be done either free-hand or assisted by a stereotaxic device to produce lifelong expression of virally delivered transgenes.
- Published
- 2016
- Full Text
- View/download PDF
30. Corrigendum: Wild-type microglia do not reverse pathology in mouse models of Rett syndrome.
- Author
-
Wang J, Wegener JE, Huang TW, Sripathy S, De Jesus-Cortes H, Xu P, Tran S, Knobbe W, Leko V, Britt J, Starwalt R, McDaniel L, Ward CS, Parra D, Newcomb B, Lao U, Nourigat C, Flowers DA, Cullen S, Jorstad NL, Yang Y, Glaskova L, Vigneau S, Kozlitina J, Yetman MJ, Jankowsky JL, Reichardt SD, Reichardt HM, Gärtner J, Bartolomei MS, Fang M, Loeb K, Keene CD, Bernstein I, Goodell M, Brat DJ, Huppke P, Neul JL, Bedalov A, and Pieper AA
- Published
- 2015
- Full Text
- View/download PDF
31. Quaternary Structure Defines a Large Class of Amyloid-β Oligomers Neutralized by Sequestration.
- Author
-
Liu P, Reed MN, Kotilinek LA, Grant MK, Forster CL, Qiang W, Shapiro SL, Reichl JH, Chiang AC, Jankowsky JL, Wilmot CM, Cleary JP, Zahs KR, and Ashe KH
- Subjects
- Alzheimer Disease pathology, Amyloid beta-Peptides chemistry, Amyloid beta-Peptides classification, Animals, Humans, Mice, Plaque, Amyloid chemistry, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Plaque, Amyloid metabolism, Protein Aggregation, Pathological metabolism, Protein Structure, Quaternary
- Abstract
The accumulation of amyloid-β (Aβ) as amyloid fibrils and toxic oligomers is an important step in the development of Alzheimer's disease (AD). However, there are numerous potentially toxic oligomers and little is known about their neurological effects when generated in the living brain. Here we show that Aβ oligomers can be assigned to one of at least two classes (type 1 and type 2) based on their temporal, spatial, and structural relationships to amyloid fibrils. The type 2 oligomers are related to amyloid fibrils and represent the majority of oligomers generated in vivo, but they remain confined to the vicinity of amyloid plaques and do not impair cognition at levels relevant to AD. Type 1 oligomers are unrelated to amyloid fibrils and may have greater potential to cause global neural dysfunction in AD because they are dispersed. These results refine our understanding of the pathogenicity of Aβ oligomers in vivo., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
32. Wild-type microglia do not reverse pathology in mouse models of Rett syndrome.
- Author
-
Wang J, Wegener JE, Huang TW, Sripathy S, De Jesus-Cortes H, Xu P, Tran S, Knobbe W, Leko V, Britt J, Starwalt R, McDaniel L, Ward CS, Parra D, Newcomb B, Lao U, Nourigat C, Flowers DA, Cullen S, Jorstad NL, Yang Y, Glaskova L, Vingeau S, Kozlitina J, Yetman MJ, Jankowsky JL, Reichardt SD, Reichardt HM, Gärtner J, Bartolomei MS, Fang M, Loeb K, Keene CD, Bernstein I, Goodell M, Brat DJ, Huppke P, Neul JL, Bedalov A, and Pieper AA
- Subjects
- Animals, Female, Male, Disease Progression, Methyl-CpG-Binding Protein 2 metabolism, Microglia cytology, Microglia physiology, Rett Syndrome pathology
- Published
- 2015
- Full Text
- View/download PDF
33. NFκB-activated astroglial release of complement C3 compromises neuronal morphology and function associated with Alzheimer's disease.
- Author
-
Lian H, Yang L, Cole A, Sun L, Chiang AC, Fowler SW, Shim DJ, Rodriguez-Rivera J, Taglialatela G, Jankowsky JL, Lu HC, and Zheng H
- Subjects
- Alzheimer Disease pathology, Amyloid beta-Protein Precursor genetics, Animals, Brain metabolism, Humans, Mice, Mice, Transgenic, Microscopy, Confocal, Neurons pathology, Receptors, Complement antagonists & inhibitors, Signal Transduction, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Astrocytes metabolism, Complement C3 metabolism, NF-kappa B metabolism, Neurons metabolism, Receptors, Complement metabolism
- Abstract
Abnormal NFκB activation has been implicated in Alzheimer's disease (AD). However, the signaling pathways governing NFκB regulation and function in the brain are poorly understood. We identify complement protein C3 as an astroglial target of NFκB and show that C3 release acts through neuronal C3aR to disrupt dendritic morphology and network function. Exposure to Aβ activates astroglial NFκB and C3 release, consistent with the high levels of C3 expression in brain tissue from AD patients and APP transgenic mice, where C3aR antagonist treatment rescues cognitive impairment. Therefore, dysregulation of neuron-glia interaction through NFκB/C3/C3aR signaling may contribute to synaptic dysfunction in AD, and C3aR antagonists may be therapeutically beneficial., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. Cerebral vascular leak in a mouse model of amyloid neuropathology.
- Author
-
Tanifum EA, Starosolski ZA, Fowler SW, Jankowsky JL, and Annapragada AV
- Subjects
- Amyloid beta-Protein Precursor genetics, Animals, Brain pathology, Cerebral Amyloid Angiopathy genetics, Contrast Media, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Transgenic, Nanoparticles, Tomography, X-Ray Computed, Amyloid analysis, Blood-Brain Barrier pathology, Brain blood supply, Cerebral Amyloid Angiopathy pathology, Cerebral Arteries pathology
- Abstract
In Alzheimer's disease (AD), there is increasing evidence of blood-brain barrier (BBB) compromise, usually observed as 'microbleeds' correlated with amyloid plaque deposition and apoE-ɛ4 status, raising the possibility of nanotherapeutic delivery. Molecular probes have been used to study neurovascular leak, but this approach does not adequately estimate vascular permeability of nanoparticles. We therefore characterized cerebrovascular leaks in live APP+ transgenic animals using a long circulating ∼100 nm nanoparticle computed tomography (CT) contrast agent probe. Active leaks fell into four categories: (1) around the dorsomedial cerebellar artery (DMCA), (2) around other major vessels, (3) nodular leaks in the cerebral cortex, and (4) diffuse leaks. Cortical leaks were uniformly more frequent in the transgenic animals than in age-matched controls. Leaks around vessels other than the DMCA were more frequent in older transgenics compared with younger ones. All other leaks were equally prevalent across genotypes independent of age. Ten days after injection, 4 to 5 μg of the dose was estimated to be present in the brain, roughly a half of which was in locations other than the leaky choroid plexus, and associated with amyloid deposition in older animals. These results suggest that amyloid deposition and age increase delivery of nanoparticle-borne reagents to the brain, in therapeutically relevant amounts.
- Published
- 2014
- Full Text
- View/download PDF
35. Intracerebroventricular viral injection of the neonatal mouse brain for persistent and widespread neuronal transduction.
- Author
-
Kim JY, Grunke SD, Levites Y, Golde TE, and Jankowsky JL
- Subjects
- Animals, Animals, Newborn, Brain cytology, Dependovirus genetics, Female, Genetic Vectors, Injections, Intraventricular, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Neurons virology, Pregnancy, Transduction, Genetic, Transgenes, Brain physiology, Brain virology, Dependovirus physiology, Gene Transfer Techniques, Neurons physiology
- Abstract
With the pace of scientific advancement accelerating rapidly, new methods are needed for experimental neuroscience to quickly and easily manipulate gene expression in the mouse brain. Here we describe a technique first introduced by Passini and Wolfe for direct intracranial delivery of virally-encoded transgenes into the neonatal mouse brain. In its most basic form, the procedure requires only an ice bucket and a microliter syringe. However, the protocol can also be adapted for use with stereotaxic frames to improve consistency for researchers new to the technique. The method relies on the ability of adeno-associated virus (AAV) to move freely from the cerebral ventricles into the brain parenchyma while the ependymal lining is still immature during the first 12-24 hr after birth. Intraventricular injection of AAV at this age results in widespread transduction of neurons throughout the brain. Expression begins within days of injection and persists for the lifetime of the animal. Viral titer can be adjusted to control the density of transduced neurons, while co-expression of a fluorescent protein provides a vital label of transduced cells. With the rising availability of viral core facilities to provide both off-the-shelf, pre-packaged reagents and custom viral preparation, this approach offers a timely method for manipulating gene expression in the mouse brain that is fast, easy, and far less expensive than traditional germline engineering.
- Published
- 2014
- Full Text
- View/download PDF
36. Impairments in experience-dependent scaling and stability of hippocampal place fields limit spatial learning in a mouse model of Alzheimer's disease.
- Author
-
Zhao R, Fowler SW, Chiang AC, Ji D, and Jankowsky JL
- Subjects
- Action Potentials, Alzheimer Disease, Animals, Disease Models, Animal, Electrodes, Implanted, Environment, Female, Male, Maze Learning physiology, Mice, Inbred C57BL, Mice, Transgenic, Amyloidosis physiopathology, Hippocampus physiopathology, Neurons physiology, Spatial Learning physiology, Spatial Navigation physiology
- Abstract
Impaired spatial memory characterizes many mouse models for Alzheimer's disease, but we understand little about how this trait arises. Here, we use a transgenic model of amyloidosis to examine the relationship between behavioral performance in tests of spatial navigation and the function of hippocampal place cells. We find that amyloid precursor protein (APP) mice require considerably more training than controls to reach the same level of performance in a water maze task, and recall the trained location less well 24 h later. At a single cell level, place fields from control mice become more stable and spatially restricted with repeated exposure to a new environment, while those in APP mice improve less over time, ultimately producing a spatial code of lower resolution, accuracy, and reliability than controls. The limited refinement of place fields in APP mice likely contributes to their delayed water maze acquisition, and provides evidence for circuit dysfunction underlying cognitive impairment., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
37. Genetic modulation of soluble Aβ rescues cognitive and synaptic impairment in a mouse model of Alzheimer's disease.
- Author
-
Fowler SW, Chiang AC, Savjani RR, Larson ME, Sherman MA, Schuler DR, Cirrito JR, Lesné SE, and Jankowsky JL
- Subjects
- Alanine administration & dosage, Alanine analogs & derivatives, Amyloid Precursor Protein Secretases antagonists & inhibitors, Amyloid beta-Protein Precursor genetics, Animals, Azepines administration & dosage, Cognition Disorders therapy, Disease Models, Animal, Doxycycline pharmacology, Exploratory Behavior drug effects, Exploratory Behavior physiology, Humans, Maze Learning drug effects, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutation, Plaque, Amyloid chemically induced, Plaque, Amyloid metabolism, Synapses drug effects, Alzheimer Disease complications, Alzheimer Disease genetics, Alzheimer Disease pathology, Amyloid Precursor Protein Secretases metabolism, Cognition Disorders genetics, Cognition Disorders metabolism, Synapses pathology
- Abstract
An unresolved debate in Alzheimer's disease (AD) is whether amyloid plaques are pathogenic, causing overt physical disruption of neural circuits, or protective, sequestering soluble forms of amyloid-β (Aβ) that initiate synaptic damage and cognitive decline. Few animal models of AD have been capable of isolating the relative contribution made by soluble and insoluble forms of Aβ to the behavioral symptoms and biochemical consequences of the disease. Here we use a controllable transgenic mouse model expressing a mutant form of amyloid precursor protein (APP) to distinguish the impact of soluble Aβ from that of deposited amyloid on cognitive function and synaptic structure. Rapid inhibition of transgenic APP modulated the production of Aβ without affecting pre-existing amyloid deposits and restored cognitive performance to the level of healthy controls in Morris water maze, radial arm water maze, and fear conditioning. Selective reduction of Aβ with a γ-secretase inhibitor provided similar improvement, suggesting that transgene suppression restored cognition, at least in part by lowering Aβ. Cognitive improvement coincided with reduced levels of synaptotoxic Aβ oligomers, greater synaptic density surrounding amyloid plaques, and increased expression of presynaptic and postsynaptic markers. Together these findings indicate that transient Aβ species underlie much of the cognitive and synaptic deficits observed in this model and demonstrate that significant functional and structural recovery can be attained without removing deposited amyloid., (Copyright © 2014 the authors 0270-6474/14/347871-15$15.00/0.)
- Published
- 2014
- Full Text
- View/download PDF
38. Specificity and efficiency of reporter expression in adult neural progenitors vary substantially among nestin-CreER(T2) lines.
- Author
-
Sun MY, Yetman MJ, Lee TC, Chen Y, and Jankowsky JL
- Subjects
- Animals, Brain metabolism, Cell Lineage genetics, Chloride Channels genetics, Chloride Channels metabolism, Collagen Type IV metabolism, Galactosides genetics, Galactosides metabolism, Integrases metabolism, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Nerve Tissue Proteins metabolism, Receptors, Estrogen genetics, Adult Stem Cells metabolism, Brain cytology, Integrases genetics, Nestin genetics
- Abstract
Transgenic lines expressing a controllable form of Cre recombinase have become valuable tools for manipulating gene expression in adult neural progenitors and their progeny. Neural progenitors express several proteins that distinguish them from mature neurons, and the promoters for these genes have been co-opted to produce selective transgene expression within this population. To date, nine CreER(T2) transgenic lines have been designed using the nestin promoter; however, only a subset are capable of eliciting expression within both neurogenic zones of the adult brain. Here we compare three such nestin-CreER(T2) lines to evaluate specificity of expression and efficiency of recombination. Each line was examined by using three different Cre reporter strains that varied in sensitivity. We found that all three nestin-CreER(T2) strains induced reporter expression within the main neurogenic areas, albeit to varying degrees depending on the reporter. Unexpectedly, we found that two of the three lines induced substantial reporter expression outside of neurogenic areas. These lines produced strong labeling in cerebellar granule neurons, with additional expression in the cortex, hippocampus, striatum, and thalamus. Reporter expression in the third nestin-CreER(T2) line was considerably more specific, but was also less efficient, labeling a smaller percentage of the target population than the other two drivers. Our findings suggest that each nestin-CreER(T2) line may best serve different experimental needs, depending on whether specificity or efficiency is of greatest concern. Our study further demonstrates that each new pair of driver and responder lines should be evaluated independently, as both components can significantly influence the resulting expression pattern., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
39. Genetic suppression of transgenic APP rescues Hypersynchronous network activity in a mouse model of Alzeimer's disease.
- Author
-
Born HA, Kim JY, Savjani RR, Das P, Dabaghian YA, Guo Q, Yoo JW, Schuler DR, Cirrito JR, Zheng H, Golde TE, Noebels JL, and Jankowsky JL
- Subjects
- Amyloid Precursor Protein Secretases antagonists & inhibitors, Animals, Disease Models, Animal, Entropy, Female, Gene Knock-In Techniques, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Models, Neurological, Neural Inhibition physiology, Presenilin-1 genetics, Seizures chemically induced, Seizures physiopathology, Suppression, Genetic, Transgenes physiology, Alzheimer Disease genetics, Alzheimer Disease physiopathology, Amyloid beta-Protein Precursor genetics, Cerebral Cortex physiopathology, Electroencephalography
- Abstract
Alzheimer's disease (AD) is associated with an elevated risk for seizures that may be fundamentally connected to cognitive dysfunction. Supporting this link, many mouse models for AD exhibit abnormal electroencephalogram (EEG) activity in addition to the expected neuropathology and cognitive deficits. Here, we used a controllable transgenic system to investigate how network changes develop and are maintained in a model characterized by amyloid β (Aβ) overproduction and progressive amyloid pathology. EEG recordings in tet-off mice overexpressing amyloid precursor protein (APP) from birth display frequent sharp wave discharges (SWDs). Unexpectedly, we found that withholding APP overexpression until adulthood substantially delayed the appearance of epileptiform activity. Together, these findings suggest that juvenile APP overexpression altered cortical development to favor synchronized firing. Regardless of the age at which EEG abnormalities appeared, the phenotype was dependent on continued APP overexpression and abated over several weeks once transgene expression was suppressed. Abnormal EEG discharges were independent of plaque load and could be extinguished without altering deposited amyloid. Selective reduction of Aβ with a γ-secretase inhibitor has no effect on the frequency of SWDs, indicating that another APP fragment or the full-length protein was likely responsible for maintaining EEG abnormalities. Moreover, transgene suppression normalized the ratio of excitatory to inhibitory innervation in the cortex, whereas secretase inhibition did not. Our results suggest that APP overexpression, and not Aβ overproduction, is responsible for EEG abnormalities in our transgenic mice and can be rescued independently of pathology.
- Published
- 2014
- Full Text
- View/download PDF
40. Wild-type neural progenitors divide and differentiate normally in an amyloid-rich environment.
- Author
-
Yetman MJ and Jankowsky JL
- Subjects
- Animals, Cell Survival genetics, Female, Hippocampus metabolism, Hippocampus pathology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Inbred ICR, Mice, Transgenic, Neurons metabolism, Amyloid beta-Peptides genetics, Cell Differentiation genetics, Cell Division genetics, Neurogenesis genetics, Neurons pathology
- Abstract
Adult neurogenesis is modulated by a balance of extrinsic signals and intrinsic responses that maintain production of new granule cells in the hippocampus. Disorders that disrupt the proliferative niche can impair this process, and alterations in adult neurogenesis have been described in human autopsy tissue and transgenic mouse models of Alzheimer's disease. Because exogenous application of aggregated Aβ peptide is neurotoxic in vitro and extracellular Aβ deposits are the main pathological feature recapitulated by mouse models, cell-extrinsic effects of Aβ accumulation were thought to underlie the breakdown of hippocampal neurogenesis observed in Alzheimer's models. We tested this hypothesis using a bigenic mouse in which transgenic expression of APP was restricted to mature projection neurons. These mice allowed us to examine how wild-type neural progenitor cells responded to high levels of Aβ released from neighboring granule neurons. We find that the proliferation, determination, and survival of hippocampal adult-born granule neurons are unaffected in the APP bigenic mice, despite abundant amyloid pathology and robust neuroinflammation. Our findings suggest that Aβ accumulation is insufficient to impair adult hippocampal neurogenesis, and that factors other than amyloid pathology may account for the neurogenic deficits observed in transgenic models with more widespread APP expression.
- Published
- 2013
- Full Text
- View/download PDF
41. Capsid serotype and timing of injection determines AAV transduction in the neonatal mice brain.
- Author
-
Chakrabarty P, Rosario A, Cruz P, Siemienski Z, Ceballos-Diaz C, Crosby K, Jansen K, Borchelt DR, Kim JY, Jankowsky JL, Golde TE, and Levites Y
- Subjects
- Animals, Animals, Newborn, Astrocytes virology, Brain cytology, Brain virology, Green Fluorescent Proteins genetics, Injections, Mice, Time Factors, Viral Tropism, Brain metabolism, Capsid metabolism, Dependovirus classification, Dependovirus genetics, Serotyping, Transduction, Genetic methods
- Abstract
Adeno-associated virus (AAV) mediated gene expression is a powerful tool for gene therapy and preclinical studies. A comprehensive analysis of CNS cell type tropism, expression levels and biodistribution of different capsid serotypes has not yet been undertaken in neonatal rodents. Our previous studies show that intracerebroventricular injection with AAV2/1 on neonatal day P0 results in widespread CNS expression but the biodistribution is limited if injected beyond neonatal day P1. To extend these observations we explored the effect of timing of injection on tropism and biodistribution of six commonly used pseudotyped AAVs delivered in the cerebral ventricles of neonatal mice. We demonstrate that AAV2/8 and 2/9 resulted in the most widespread biodistribution in the brain. Most serotypes showed varying biodistribution depending on the day of injection. Injection on neonatal day P0 resulted in mostly neuronal transduction, whereas administration in later periods of development (24-84 hours postnatal) resulted in more non-neuronal transduction. AAV2/5 showed widespread transduction of astrocytes irrespective of the time of injection. None of the serotypes tested showed any microglial transduction. This study demonstrates that both capsid serotype and timing of injection influence the regional and cell-type distribution of AAV in neonatal rodents, and emphasizes the utility of pseudotyped AAV vectors for translational gene therapy paradigms.
- Published
- 2013
- Full Text
- View/download PDF
42. Viral transduction of the neonatal brain delivers controllable genetic mosaicism for visualising and manipulating neuronal circuits in vivo.
- Author
-
Kim JY, Ash RT, Ceballos-Diaz C, Levites Y, Golde TE, Smirnakis SM, and Jankowsky JL
- Subjects
- Animals, Animals, Newborn, Genetic Vectors genetics, Mice, Brain, Dependovirus genetics, Mosaicism, Transduction, Genetic methods
- Abstract
The neonatal intraventricular injection of adeno-associated virus has been shown to transduce neurons widely throughout the brain, but its full potential for experimental neuroscience has not been adequately explored. We report a detailed analysis of the method's versatility with an emphasis on experimental applications where tools for genetic manipulation are currently lacking. Viral injection into the neonatal mouse brain is fast, easy, and accesses regions of the brain including the cerebellum and brainstem that have been difficult to target with other techniques such as electroporation. We show that viral transduction produces an inherently mosaic expression pattern that can be exploited by varying the titer to transduce isolated neurons or densely-packed populations. We demonstrate that the expression of virally-encoded proteins is active much sooner than previously believed, allowing genetic perturbation during critical periods of neuronal plasticity, but is also long-lasting and stable, allowing chronic studies of aging. We harness these features to visualise and manipulate neurons in the hindbrain that have been recalcitrant to approaches commonly applied in the cortex. We show that viral labeling aids the analysis of postnatal dendritic maturation in cerebellar Purkinje neurons by allowing individual cells to be readily distinguished, and then demonstrate that the same sparse labeling allows live in vivo imaging of mature Purkinje neurons at a resolution sufficient for complete analytical reconstruction. Given the rising availability of viral constructs, packaging services, and genetically modified animals, these techniques should facilitate a wide range of experiments into brain development, function, and degeneration., (© 2013 Federation of European Neuroscience Societies and Blackwell Publishing Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
43. Strain background influences neurotoxicity and behavioral abnormalities in mice expressing the tetracycline transactivator.
- Author
-
Han HJ, Allen CC, Buchovecky CM, Yetman MJ, Born HA, Marin MA, Rodgers SP, Song BJ, Lu HC, Justice MJ, Probst FJ, and Jankowsky JL
- Subjects
- Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Analysis of Variance, Animals, Anti-Bacterial Agents pharmacology, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Chromosome Mapping, Conditioning, Psychological physiology, Dentate Gyrus metabolism, Dentate Gyrus pathology, Disease Models, Animal, Doxycycline pharmacology, Exploratory Behavior physiology, Fear physiology, Female, Male, Maze Learning physiology, Mental Disorders pathology, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Mutation genetics, Neurotoxicity Syndromes pathology, Species Specificity, tau Proteins genetics, Mental Disorders genetics, Mental Disorders metabolism, Neurotoxicity Syndromes genetics, Neurotoxicity Syndromes metabolism, Tetracycline metabolism, Trans-Activators genetics
- Abstract
The tet-off system has been widely used to create transgenic models of neurological disorders including Alzheimer's, Parkinson's, Huntington's, and prion disease. The utility of this system lies in the assumption that the tetracycline transactivator (TTA) acts as an inert control element and does not contribute to phenotypes under study. Here we report that neuronal expression of TTA can affect hippocampal cytoarchitecture and behavior in a strain-dependent manner. While studying neurodegeneration in two tet-off Alzheimer's disease models, we unexpectedly discovered neuronal loss within the dentate gyrus of single transgenic TTA controls. Granule neurons appeared most sensitive to TTA exposure during postnatal development, and doxycycline treatment during this period was neuroprotective. TTA-induced degeneration could be rescued by moving the transgene onto a congenic C57BL/6J background and recurred on reintroduction of either CBA or C3H/He backgrounds. Quantitative trait analysis of B6C3 F2 TTA mice identified a region on Chromosome 14 that contains a major modifier of the neurodegenerative phenotype. Although B6 mice were resistant to degeneration, they were not ideal for cognitive testing. F1 offspring of TTA C57BL/6J and 129X1/SvJ, FVB/NJ, or DBA/1J showed improved spatial learning, but TTA expression caused subtle differences in contextual fear conditioning on two of these backgrounds, indicating that strain and genotype can interact independently under different behavioral settings. All model systems have limitations that should be recognized and mitigated where possible; our findings stress the importance of mapping the effects caused by TTA alone when working with tet-off models.
- Published
- 2012
- Full Text
- View/download PDF
44. Transgenic APP expression during postnatal development causes persistent locomotor hyperactivity in the adult.
- Author
-
Rodgers SP, Born HA, Das P, and Jankowsky JL
- Subjects
- Aging, Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloid beta-Protein Precursor metabolism, Animals, Animals, Genetically Modified, Brain metabolism, Disease Models, Animal, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Neurons metabolism, Transgenes, Amyloid beta-Protein Precursor genetics, Locomotion genetics
- Abstract
Background: Transgenic mice expressing disease-associated proteins have become standard tools for studying human neurological disorders. Transgenes are often expressed using promoters chosen to drive continuous high-level expression throughout life rather than temporal and spatial fidelity to the endogenous gene. This approach has allowed us to recapitulate diseases of aging within the two-year lifespan of the laboratory mouse, but has the potential for creating aberrant phenotypes by mechanisms unrelated to the human disorder., Results: We show that overexpression of the Alzheimer's-related amyloid precursor protein (APP) during early postnatal development leads to severe locomotor hyperactivity that can be significantly attenuated by delaying transgene onset until adulthood. Our data suggest that exposure to transgenic APP during maturation influences the development of neuronal circuits controlling motor activity. Both when matched for total duration of APP overexpression and when matched for cortical amyloid burden, animals exposed to transgenic APP as juveniles are more active in locomotor assays than animals in which APP overexpression was delayed until adulthood. In contrast to motor activity, the age of APP onset had no effect on thigmotaxis in the open field as a rough measure of anxiety, suggesting that the interaction between APP overexpression and brain development is not unilateral., Conclusions: Our findings indicate that locomotor hyperactivity displayed by the tet-off APP transgenic mice and several other transgenic models of Alzheimer's disease may result from overexpression of mutant APP during postnatal brain development. Our results serve as a reminder of the potential for unexpected interactions between foreign transgenes and brain development to cause long-lasting effects on neuronal function in the adult. The tet-off APP model provides an easy means of avoiding developmental confounds by allowing transgene expression to be delayed until the mice reach adulthood.
- Published
- 2012
- Full Text
- View/download PDF
45. Neuronal aggregates are associated with phenotypic onset in the R6/2 Huntington's disease transgenic mouse.
- Author
-
Cowin RM, Roscic A, Bui N, Graham D, Paganetti P, Jankowsky JL, Weiss A, and Paylor R
- Subjects
- Age of Onset, Animals, Brain metabolism, Disease Models, Animal, Fear physiology, Humans, Huntingtin Protein, Male, Mice, Mice, Transgenic, Motor Activity physiology, Motor Skills physiology, Mutation, Nerve Tissue Proteins genetics, Peptides genetics, Phenotype, Sensory Gating physiology, Trinucleotide Repeat Expansion genetics, Trinucleotide Repeat Expansion physiology, Huntington Disease genetics, Huntington Disease physiopathology, Nerve Tissue Proteins physiology, Neurons metabolism, Proteins metabolism
- Abstract
Huntington's disease (HD) is caused by the expansion of the polyglutamine tract expressed in the huntingtin protein. Data from patients show a strong negative correlation between CAG repeat size and age of disease onset. Recent studies in mixed background C57×CBA R6/2 mice suggest the inverse correlation observed in the human disease may not be replicated in some animal models of HD. To further clarify the relationship between repeat length and age of onset, congenic C57BL6/J R6/2 transgenic mice expressing 110, 260 or 310 CAG were tested in a comprehensive behavioral battery at multiple ages. Data confirmed the findings of earlier studies and indicate that on a pure C57BL6/J genetic background, R6/2 mice with larger repeats exhibit a delay in phenotypic onset with increasing polyglutamine size (6 weeks in 110 CAG and 17 weeks in 310 CAG mice). Further analysis confirmed a decrease in transgene transcript expression in 310 CAG mice as well as differential aggregated protein localization in association with repeat length. Mice expressing 110 CAG developed aggregates that localized almost exclusively to the nucleus of neuronal cells in the striatum and cortex. In contrast, tissue from 310 CAG mice exhibited predominantly extranuclear inclusions. Novel mutant protein analysis obtained using time-resolved fluorescence resonance energy transfer (FRET) revealed that soluble protein levels decreased with disease onset in R6/2 mice while aggregated protein levels increased. We believe that these data suggest a role for aggregation and inclusion localization in HD pathogenesis and propose a mechanism for the age of onset delay observed in R6/2 mice., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
46. Robust amyloid clearance in a mouse model of Alzheimer's disease provides novel insights into the mechanism of amyloid-beta immunotherapy.
- Author
-
Wang A, Das P, Switzer RC 3rd, Golde TE, and Jankowsky JL
- Subjects
- Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Protein Precursor metabolism, Analysis of Variance, Animals, Blotting, Western, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Hippocampus metabolism, Immunohistochemistry, Mice, Mice, Transgenic, Treatment Outcome, Alzheimer Disease therapy, Amyloid beta-Peptides metabolism, Hippocampus pathology, Immunization, Passive methods, Plaque, Amyloid metabolism
- Abstract
Many new therapeutics for Alzheimer's disease delay the accumulation of amyloid-β (Aβ) in transgenic mice, but evidence for clearance of preexisting plaques is often lacking. Here, we demonstrate that anti-Aβ immunotherapy combined with suppression of Aβ synthesis allows significant removal of antecedent deposits. We treated amyloid-bearing tet-off APP (amyloid precursor protein) mice with doxycycline to suppress transgenic Aβ production before initiating a 12 week course of passive immunization. Animals remained on doxycycline for 3 months afterward to assess whether improvements attained during combined treatment could be maintained by monotherapy. This strategy reduced amyloid load by 52% and Aβ42 content by 28% relative to pretreatment levels, with preferential clearance of small deposits and diffuse Aβ surrounding fibrillar cores. We demonstrate that peripherally administered anti-Aβ antibody crossed the blood-brain barrier, bound to plaques, and was still be found associated with a subset of amyloid deposits many months after the final injection. Antibody accessed the brain independent of plasma Aβ levels, where it enhanced microglial internalization of aggregated Aβ. Our data support a mechanism by which passive immunization acts centrally to stimulate microglial phagocytosis of aggregated Aβ, but is opposed by the continued aggregation of newly secreted Aβ. By arresting the production of Aβ, combination therapy allows microglial clearance to work from a static amyloid burden toward a significant reduction in plaque load. Our findings suggest that combining two therapeutic approaches currently in clinical trials may improve neuropathological outcome over either alone.
- Published
- 2011
- Full Text
- View/download PDF
47. Remote sites of structural atrophy predict later amyloid formation in a mouse model of Alzheimer's disease.
- Author
-
Badea A, Johnson GA, and Jankowsky JL
- Subjects
- Alzheimer Disease metabolism, Amyloid metabolism, Animals, Atrophy pathology, Brain metabolism, Disease Models, Animal, Female, Image Interpretation, Computer-Assisted, Male, Mice, Mice, Transgenic, Alzheimer Disease pathology, Brain pathology, Magnetic Resonance Imaging methods, Plaque, Amyloid pathology
- Abstract
Magnetic resonance (MR) imaging can provide a longitudinal view of neurological disease through repeated imaging of patients at successive stages of impairment. Until recently, the difficulty of manual delineation has limited volumetric analyses of MR data sets to a few select regions and a small number of subjects. Increased throughput offered by faster imaging methods, automated segmentation, and deformation-based morphometry have recently been applied to overcome this limitation with mouse models of neurological conditions. We use automated analyses to produce an unbiased view of volumetric changes in a transgenic mouse model for Alzheimer's disease (AD) at two points in the progression of disease: immediately before and shortly after the onset of amyloid formation. In addition to the cortex and hippocampus, where atrophy has been well documented in AD patients, we identify volumetric losses in the pons and substantia nigra where neurodegeneration has not been carefully examined. We find that deficits in cortical volume precede amyloid formation in this mouse model, similar to presymptomatic atrophy seen in patients with familial AD. Unexpectedly, volumetric losses identified by MR outside of the forebrain predict locations of future amyloid formation, such as the inferior colliculus and spinal nuclei, which develop pathology at very late stages of disease. Our work provides proof-of-principle that MR microscopy can expand our view of AD by offering a complete and unbiased examination of volumetric changes that guide us in revisiting the canonical neuropathology., (Copyright 2009 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
48. GABA transporter function, oligomerization state, and anchoring: correlates with subcellularly resolved FRET.
- Author
-
Moss FJ, Imoukhuede PI, Scott K, Hu J, Jankowsky JL, Quick MW, and Lester HA
- Subjects
- Animals, Biological Transport, Cell Line, Cell Membrane, Fluorescence Resonance Energy Transfer, GABA Plasma Membrane Transport Proteins genetics, Humans, Mice, GABA Plasma Membrane Transport Proteins chemistry, GABA Plasma Membrane Transport Proteins metabolism, gamma-Aminobutyric Acid metabolism
- Abstract
The mouse gamma-aminobutyric acid (GABA) transporter mGAT1 was expressed in neuroblastoma 2a cells. 19 mGAT1 designs incorporating fluorescent proteins were functionally characterized by [(3)H]GABA uptake in assays that responded to several experimental variables, including the mutations and pharmacological manipulation of the cytoskeleton. Oligomerization and subsequent trafficking of mGAT1 were studied in several subcellular regions of live cells using localized fluorescence, acceptor photobleach Förster resonance energy transfer (FRET), and pixel-by-pixel analysis of normalized FRET (NFRET) images. Nine constructs were functionally indistinguishable from wild-type mGAT1 and provided information about normal mGAT1 assembly and trafficking. The remainder had compromised [(3)H]GABA uptake due to observable oligomerization and/or trafficking deficits; the data help to determine regions of mGAT1 sequence involved in these processes. Acceptor photobleach FRET detected mGAT1 oligomerization, but richer information was obtained from analyzing the distribution of all-pixel NFRET amplitudes. We also analyzed such distributions restricted to cellular subregions. Distributions were fit to either two or three Gaussian components. Two of the components, present for all mGAT1 constructs that oligomerized, may represent dimers and high-order oligomers (probably tetramers), respectively. Only wild-type functioning constructs displayed three components; the additional component apparently had the highest mean NFRET amplitude. Near the cell periphery, wild-type functioning constructs displayed the highest NFRET. In this subregion, the highest NFRET component represented approximately 30% of all pixels, similar to the percentage of mGAT1 from the acutely recycling pool resident in the plasma membrane in the basal state. Blocking the mGAT1 C terminus postsynaptic density 95/discs large/zona occludens 1 (PDZ)-interacting domain abolished the highest amplitude component from the NFRET distributions. Disrupting the actin cytoskeleton in cells expressing wild-type functioning transporters moved the highest amplitude component from the cell periphery to perinuclear regions. Thus, pixel-by-pixel NFRET analysis resolved three distinct forms of GAT1: dimers, high-order oligomers, and transporters associated via PDZ-mediated interactions with the actin cytoskeleton and/or with the exocyst.
- Published
- 2009
- Full Text
- View/download PDF
49. Rodent A beta modulates the solubility and distribution of amyloid deposits in transgenic mice.
- Author
-
Jankowsky JL, Younkin LH, Gonzales V, Fadale DJ, Slunt HH, Lester HA, Younkin SG, and Borchelt DR
- Subjects
- Amino Acid Sequence, Animals, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, Peptides chemistry, Presenilin-1 metabolism, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Solubility, Transgenes, Amyloid chemistry, Amyloid beta-Peptides chemistry
- Abstract
The amino acid sequence of amyloid precursor protein (APP) is highly conserved, and age-related A beta aggregates have been described in a variety of vertebrate animals, with the notable exception of mice and rats. Three amino acid substitutions distinguish mouse and human A beta that might contribute to their differing properties in vivo. To examine the amyloidogenic potential of mouse A beta, we studied several lines of transgenic mice overexpressing wild-type mouse amyloid precursor protein (moAPP) either alone or in conjunction with mutant PS1 (PS1dE9). Neither overexpression of moAPP alone nor co-expression with PS1dE9 caused mice to develop Alzheimer-type amyloid pathology by 24 months of age. We further tested whether mouse A beta could accelerate the deposition of human A beta by crossing the moAPP transgenic mice to a bigenic line expressing human APPswe with PS1dE9. The triple transgenic animals (moAPP x APPswe/PS1dE9) produced 20% more A beta but formed amyloid deposits no faster and to no greater extent than APPswe/PS1dE9 siblings. Instead, the additional mouse A beta increased the detergent solubility of accumulated amyloid and exacerbated amyloid deposition in the vasculature. These findings suggest that, although mouse A beta does not influence the rate of amyloid formation, the incorporation of A beta peptides with differing sequences alters the solubility and localization of the resulting aggregates.
- Published
- 2007
- Full Text
- View/download PDF
50. Alzheimer's-type amyloidosis in transgenic mice impairs survival of newborn neurons derived from adult hippocampal neurogenesis.
- Author
-
Verret L, Jankowsky JL, Xu GM, Borchelt DR, and Rampon C
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Amyloidosis genetics, Amyloidosis metabolism, Animals, Cell Survival genetics, Cellular Senescence genetics, Hippocampus metabolism, Humans, Male, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Transgenic, Neurons metabolism, Alzheimer Disease pathology, Amyloidosis pathology, Hippocampus cytology, Hippocampus pathology, Neurons pathology
- Abstract
Alzheimer's disease (AD) is characterized by severe neuronal loss in several brain regions important for learning and memory. Of the structures affected by AD, the hippocampus is unique in continuing to produce new neurons throughout life. Mounting evidence indicates that hippocampal neurogenesis contributes to the processing and storage of new information and that deficits in the production of new neurons may impair learning and memory. Here, we examine whether the overproduction of amyloid-beta (Abeta) peptide in a mouse model for AD might be detrimental to newborn neurons in the hippocampus. We used transgenic mice overexpressing familial AD variants of amyloid precursor protein (APP) and/or presenilin-1 to test how the level (moderate or high) and the aggregation state (soluble or deposited) of Abeta impacts the proliferation and survival of new hippocampal neurons. Although proliferation and short-term survival of neural progenitors in the hippocampus was unaffected by APP/Abeta overproduction, survival of newborn cells 4 weeks later was dramatically diminished in transgenic mice with Alzheimer's-type amyloid pathology. Phenotypic analysis of the surviving population revealed a specific reduction in newborn neurons. Our data indicate that overproduction of Abeta and the consequent appearance of amyloid plaques cause an overall reduction in the number of adult-generated hippocampal neurons. Diminished capacity for hippocampal neuron replacement may contribute to the cognitive decline observed in these mice.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.