Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
NPJ Parkinsons Dis ; 7(1): 30, 2021 Mar 19.
Article in English | MEDLINE | ID: mdl-33741985

ABSTRACT

Parkinson's disease (PD) and related synucleinopathies are characterized by chronic neuroinflammation leading to the premise that anti-inflammatory therapies could ameliorate synucleinopathy and associated sequelae. To test this idea, we used recombinant adeno-associated viruses (AAV) to express the anti-inflammatory cytokine, Interleukin (Il)-10, in Line M83 transgenic mice that expresses the PD-associated A53T mutant human α-synuclein (αSyn). Contrary to our expectations, we observed that intraspinal Il-10 expression initiated at birth upregulated microgliosis and led to early death in homozygous M83+/+ mice. We further observed that Il-10 preconditioning led to reduced lifespan in the hemizygous M83+/- mice injected with preformed αSyn aggregates in hindlimb muscles. To determine the mechanistic basis for these adverse effects, we took advantage of the I87A variant Il-10 (vIl-10) that has predominantly immunosuppressive properties. Sustained intraspinal expression of vIl-10 in preformed αSyn-aggregate seeded M83+/- mice resulted in earlier death, accelerated αSyn pathology, pronounced microgliosis, and increased apoptosis compared to control mice. AAV-vIl-10 expression robustly induced p62 and neuronal LC3B accumulation in these mice, indicating that Il-10 signaling mediated preconditioning of the neuraxis can potentially exacerbate αSyn accumulation through autophagy dysfunction in the neurons. Together, our data demonstrate unexpected adverse effects of both Il-10 and its immunosuppressive variant, vIl-10, in a mouse model of PD, highlighting the pleiotropic functions of immune mediators and their complex role in non-cell autonomous signaling in neurodegenerative proteinopathies.

2.
Mol Ther ; 29(2): 859-872, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33128896

ABSTRACT

Immunotherapies designed to treat neurodegenerative tauopathies that primarily engage extracellular tau may have limited efficacy as tau is primarily intracellular. We generated tau-targeting single-chain variable fragments (scFvs) and intrabodies (iBs) from the phosphorylated tau-specific antibodies CP13 and PHF1 and the pan-tau antibody Tau5. Recombinant adeno-associated virus (rAAV) was utilized to express these antibody fragments in homozygous JNPL3 P301L tau mice. Two iBs (CP13i, PHF1i) and one scFv (PHF1s) abrogated tau pathology and delayed time to severe hindlimb paralysis. In a second tauopathy model (rTg4510), CP13i and PHF1i reduced tau pathology, but cognate scFvs did not. These data demonstrate that (1) disease-modifying efficacy does not require antibody effector functions, (2) the intracellular targeting of tau with phosphorylated tau-specific iBs is more effective than extracellular targeting with the scFvs, and (3) robust effects on tau pathology before neurodegeneration only resulted in modest disease modification as assessed by delay of severe motor phenotype.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Secretory Pathway/drug effects , Single-Chain Antibodies/pharmacology , tau Proteins/antagonists & inhibitors , Animals , Combined Modality Therapy , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Transgenic , Neurodegenerative Diseases/etiology , Treatment Outcome , tau Proteins/metabolism
3.
Alzheimers Res Ther ; 12(1): 132, 2020 10 17.
Article in English | MEDLINE | ID: mdl-33069251

ABSTRACT

BACKGROUND: Self-assembly of the amyloid-ß (Aß) peptide into aggregates, from small oligomers to amyloid fibrils, is fundamentally linked with Alzheimer's disease (AD). However, it is clear that not all forms of Aß are equally harmful and that linking a specific aggregate to toxicity also depends on the assays and model systems used (Haass et al., J Biol. Chem 269:17741-17748, 1994; Borchelt et al., Neuron 17:1005-1013, 1996). Though a central postulate of the amyloid cascade hypothesis, there remain many gaps in our understanding regarding the links between Aß deposition and neurodegeneration. METHODS: In this study, we examined familial mutations of Aß that increase aggregation and oligomerization, E22G and ΔE22, and induce cerebral amyloid angiopathy, E22Q and D23N. We also investigated synthetic mutations that stabilize dimerization, S26C, and a phospho-mimetic, S8E, and non-phospho-mimetic, S8A. To that end, we utilized BRI2-Aß fusion technology and rAAV2/1-based somatic brain transgenesis in mice to selectively express individual mutant Aß species in vivo. In parallel, we generated PhiC31-based transgenic Drosophila melanogaster expressing wild-type (WT) and Aß40 and Aß42 mutants, fused to the Argos signal peptide to assess the extent of Aß42-induced toxicity as well as to interrogate the combined effect of different Aß40 and Aß42 species. RESULTS: When expressed in the mouse brain for 6 months, Aß42 E22G, Aß42 E22Q/D23N, and Aß42WT formed amyloid aggregates consisting of some diffuse material as well as cored plaques, whereas other mutants formed predominantly diffuse amyloid deposits. Moreover, while Aß40WT showed no distinctive phenotype, Aß40 E22G and E22Q/D23N formed unique aggregates that accumulated in mouse brains. This is the first evidence that mutant Aß40 overexpression leads to deposition under certain conditions. Interestingly, we found that mutant Aß42 E22G, E22Q, and S26C, but not Aß40, were toxic to the eye of Drosophila. In contrast, flies expressing a copy of Aß40 (WT or mutants), in addition to Aß42WT, showed improved phenotypes, suggesting possible protective qualities for Aß40. CONCLUSIONS: These studies suggest that while some Aß40 mutants form unique amyloid aggregates in mouse brains, they do not exacerbate Aß42 toxicity in Drosophila, which highlights the significance of using different systems for a better understanding of AD pathogenicity and more accurate screening for new potential therapies.


Subject(s)
Alzheimer Disease , Drosophila , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Brain/metabolism , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Mice , Peptide Fragments/toxicity
4.
Mol Neurodegener ; 15(1): 15, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32122372

ABSTRACT

BACKGROUND: Recombinant adeno-associated virus (rAAV) is widely used in the neuroscience field to manipulate gene expression in the nervous system. However, a limitation to the use of rAAV vectors is the time and expense needed to produce them. To overcome this limitation, we evaluated whether unpurified rAAV vectors secreted into the media following scalable PEI transfection of HEK293T cells can be used in lieu of purified rAAV. METHODS: We packaged rAAV2-EGFP vectors in 30 different wild-type and mutant capsids and subsequently collected the media containing secreted rAAV. Genomic titers of each rAAV vector were assessed and the ability of each unpurified virus to transduce primary mixed neuroglial cultures (PNGCs), organotypic brain slice cultures (BSCs) and the mouse brain was evaluated. RESULTS: There was ~ 40-fold wide variance in the average genomic titers of the rAAV2-EGFP vector packaged in the 30 different capsids, ranging from a low ~ 4.7 × 1010 vector genomes (vg)/mL for rAAV2/5-EGFP to a high of ~ 2.0 × 1012 vg/mL for a capsid mutant of rAAV2/8-EGFP. In PNGC studies, we observed a wide range of transduction efficiency among the 30 capsids evaluated, with the rAAV2/6-EGFP vector demonstrating the highest overall transduction efficiency. In BSC studies, we observed robust transduction by wild-type capsid vectors rAAV2/6, 2/8 and 2/9, and by capsid mutants of rAAV2/1, 2/6, and 2/8. In the in vivo somatic brain transgenesis (SBT) studies, we found that intra-cerebroventricular injection of media containing unpurified rAAV2-EGFP vectors packaged with select mutant capsids resulted in abundant EGFP positive neurons and astrocytes in the hippocampus and forebrain of non-transgenic mice. We demonstrate that unpurified rAAV can express transgenes at equivalent levels to lysate-purified rAAV both in vitro and in vivo. We also show that unpurified rAAV is sufficient to drive tau pathology in BSC and neuroinflammation in vivo, recapitulating previous studies using purified rAAV. CONCLUSIONS: Unpurified rAAV vectors secreted into the media can efficiently transduce brain cells in vitro and in vivo, providing a cost-effective way to manipulate gene expression. The use of unpurified virus will greatly reduce costs of exploratory studies and further increase the utility of rAAV vectors for standard laboratory use.


Subject(s)
Dependovirus , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Transduction, Genetic/methods , Animals , Brain , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Mice , Neuroglia , Neurons
5.
Brain Struct Funct ; 225(1): 427-439, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31894407

ABSTRACT

Imaging biomarkers for immune activation may be valuable for early-stage detection, therapeutic testing, and research on neurodegenerative conditions. In the present study, we determined whether diffusion magnetic resonance imaging-derived free water signal is a sensitive marker for neuroinflammatory effects of interferon-gamma (Ifn-γ). Neonatal wild-type mice were injected in the cerebral ventricles with recombinant adeno-associated viruses expressing the inflammatory cytokine Ifn-γ. Groups of mice expressing Ifn-γ and age-matched controls were imaged at 1, 5 and 8 months. Mice deficient in Ifngr1-/- and Stat1-/- were scanned at 5 months as controls for the signaling cascades activated by Ifn-γ. The results indicate that Ifn-γ affected fractional anisotropy (FA), mean diffusivity (MD), and free water (FW) in white matter structures, midline cortical areas, and medial thalamic areas. In these structures, FA and MD decreased progressively from 1 to 8 months of age, while FW increased significantly. The observed reductions in FA and MD and increased FW with elevated brain Ifn-γ was not observed in Ifngr1-/- or Stat1-/- mice. These results suggest that the observed microstructure changes involve the Ifn-gr1 and Stat1 signaling. Interestingly, increases in FW were observed in midbrain of Ifngr1-/- mice, which suggests alternative Ifn-γ signaling in midbrain. Although initial evidence is offered in relation to the sensitivity of the FW signal to neurodegenerative and/or inflammatory patterns specific to Ifn-γ, further research is needed to determine applicability and specificity across animal models of neuroinflammatory and degenerative disorders.


Subject(s)
Brain/diagnostic imaging , Brain/pathology , Diffusion Magnetic Resonance Imaging/methods , Encephalitis/diagnostic imaging , Encephalitis/pathology , Interferon-gamma/metabolism , White Matter/diagnostic imaging , White Matter/pathology , Animals , Anisotropy , Brain/drug effects , Brain/metabolism , Encephalitis/chemically induced , Female , Interferon-gamma/administration & dosage , Interferon-gamma/genetics , Male , Mice, Knockout , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Water/analysis , White Matter/drug effects , White Matter/metabolism
6.
J Exp Med ; 216(3): 539-555, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30770411

ABSTRACT

It has been challenging to produce ex vivo models of the inclusion pathologies that are hallmark pathologies of many neurodegenerative diseases. Using three-dimensional mouse brain slice cultures (BSCs), we have developed a paradigm that rapidly and robustly recapitulates mature neurofibrillary inclusion and Lewy body formation found in Alzheimer's and Parkinson's disease, respectively. This was achieved by transducing the BSCs with recombinant adeno-associated viruses (rAAVs) that express α-synuclein or variants of tau. Notably, the tauopathy BSC model enables screening of small molecule therapeutics and tracking of neurodegeneration. More generally, the rAAV BSC "toolkit" enables efficient transduction and transgene expression from neurons, microglia, astrocytes, and oligodendrocytes, alone or in combination, with transgene expression lasting for many months. These rAAV-based BSC models provide a cost-effective and facile alternative to in vivo studies, and in the future can become a widely adopted methodology to explore physiological and pathological mechanisms related to brain function and dysfunction.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Dependovirus/genetics , Parkinson Disease/pathology , Alzheimer Disease/virology , Animals , Brain/metabolism , Brain/virology , Drug Evaluation, Preclinical/methods , Gene Expression , Humans , Mice, Inbred C3H , Mice, Transgenic , Microorganisms, Genetically-Modified , Mutation , Neurons/pathology , Organ Culture Techniques , Parkinson Disease/virology , Transduction, Genetic , Transgenes , alpha-Synuclein/genetics , tau Proteins/genetics
7.
Neurobiol Dis ; 120: 98-106, 2018 12.
Article in English | MEDLINE | ID: mdl-30195075

ABSTRACT

Mechanisms underlying α-synuclein (αSyn) mediated neurodegeneration are poorly understood. Intramuscular (IM) injection of αSyn fibrils in human A53T transgenic M83+/- mice produce a rapid model of α-synucleinopathy with highly predictable onset of motor impairment. Using varying doses of αSyn seeds, we show that αSyn-induced phenotype is largely dose-independent. We utilized the synchrony of this IM model to explore the temporal sequence of αSyn pathology, neurodegeneration and neuroinflammation. Longitudinal tracking showed that while motor neuron death and αSyn pathology occur within 2 months post IM, astrogliosis appears at a later timepoint, implying neuroinflammation is a consequence, rather than a trigger, in this prionoid model of synucleinopathy. Initiating at 3 months post IM, immune activation dominates the pathologic landscape in terminal IM-seeded M83+/- mice, as revealed by unbiased transcriptomic analyses. Our findings provide insights into the role of neuroinflammation in αSyn mediated proteostasis and neurodegeneration, which will be key in designing potential therapies.


Subject(s)
Motor Neurons/metabolism , Nerve Degeneration/metabolism , alpha-Synuclein/biosynthesis , Animals , Brain/immunology , Brain/metabolism , Brain/pathology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Transgenic , Motor Neurons/immunology , Motor Neurons/pathology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , alpha-Synuclein/immunology
8.
J Exp Med ; 215(9): 2247-2264, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30158114

ABSTRACT

There is considerable interest in harnessing innate immunity to treat Alzheimer's disease (AD). Here, we explore whether a decoy receptor strategy using the ectodomain of select TLRs has therapeutic potential in AD. AAV-mediated expression of human TLR5 ectodomain (sTLR5) alone or fused to human IgG4 Fc (sTLR5Fc) results in robust attenuation of amyloid ß (Aß) accumulation in a mouse model of Alzheimer-type Aß pathology. sTLR5Fc binds to oligomeric and fibrillar Aß with high affinity, forms complexes with Aß, and blocks Aß toxicity. Oligomeric and fibrillar Aß modulates flagellin-mediated activation of human TLR5 but does not, by itself, activate TLR5 signaling. Genetic analysis shows that rare protein coding variants in human TLR5 may be associated with a reduced risk of AD. Further, transcriptome analysis shows altered TLR gene expression in human AD. Collectively, our data suggest that TLR5 decoy receptor-based biologics represent a novel and safe Aß-selective class of biotherapy in AD.


Subject(s)
Alzheimer Disease , Immunoglobulin Fc Fragments/therapeutic use , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Toll-Like Receptor 5/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/immunology , Animals , Female , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/immunology
9.
J Exp Med ; 215(1): 283-301, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29208777

ABSTRACT

Processing of amyloid-ß (Aß) precursor protein (APP) by γ-secretase produces multiple species of Aß: Aß40, short Aß peptides (Aß37-39), and longer Aß peptides (Aß42-43). γ-Secretase modulators, a class of Alzheimer's disease therapeutics, reduce production of the pathogenic Aß42 but increase the relative abundance of short Aß peptides. To evaluate the pathological relevance of these peptides, we expressed Aß36-40 and Aß42-43 in Drosophila melanogaster to evaluate inherent toxicity and potential modulatory effects on Aß42 toxicity. In contrast to Aß42, the short Aß peptides were not toxic and, when coexpressed with Aß42, were protective in a dose-dependent fashion. In parallel, we explored the effects of recombinant adeno-associated virus-mediated expression of Aß38 and Aß40 in mice. When expressed in nontransgenic mice at levels sufficient to drive Aß42 deposition, Aß38 and Aß40 did not deposit or cause behavioral alterations. These studies indicate that treatments that lower Aß42 by raising the levels of short Aß peptides could attenuate the toxic effects of Aß42.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Amyloid/genetics , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Drosophila melanogaster , Eye/metabolism , Eye/pathology , Eye/ultrastructure , Female , Locomotion , Mice , Mice, Transgenic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Phenotype , Protein Aggregates , Protein Aggregation, Pathological/metabolism
10.
Neurobiol Dis ; 110: 133-141, 2018 02.
Article in English | MEDLINE | ID: mdl-29196213

ABSTRACT

Brain expression of AAV-Ifn-γ leads to reactive gliosis, nigrostriatal degeneration and midbrain calcification in wild type mice. This mouse model phenocopies idiopathic basal ganglia calcification which is associated with Parkinsonian symptoms. To understand how the nigro-striatal pathway is selectively vulnerable to Ifn-γ, we determined if the phenotype is driven by canonical signaling intermediates, Ifngr1 and Stat1. Using focused bioinformatic analysis and rotarod testing, we show that neuroinflammation and motor abnormalities precede the appearance of midbrain neuropathologies in the brains of Ifn-γ mouse model. To test whether canonical Ifn-γ signaling is a key driver of progressive nigrostriatal degeneration, we overexpressed Ifn-γ in the brains of Ifngr1-/- and Stat1-/- mice. Expression of Ifn-γ in Ifngr1-/- mice did not result in any neuroinflammation, midbrain calcinosis or nigrostriatal degenerative pathology. Interestingly, in Stat1-/- mice, Ifn-γ expression resulted in gliosis without recapitulating the neurodegenerative phenotype. Overall, our data shows that canonical Ifn-γ signaling triggers midbrain calcinosis and nigrostriatal neurodegeneration, providing mechanistic insights into cytokine-driven selective neuronal vulnerability. Our study establishes the broader relevance of inflammatory signaling in neurodegenerative diseases and can potentially identify novel immunological targets for Parkinsonian syndromes.


Subject(s)
Brain/pathology , Interferon-gamma/metabolism , Nerve Degeneration/metabolism , Receptors, Interferon/metabolism , STAT1 Transcription Factor/metabolism , Animals , Basal Ganglia Diseases/metabolism , Basal Ganglia Diseases/pathology , Brain/metabolism , Calcinosis/metabolism , Calcinosis/pathology , Mice , Mice, Knockout , Nerve Degeneration/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Signal Transduction/physiology , Interferon gamma Receptor
11.
Mol Ther Methods Clin Dev ; 3: 16026, 2016.
Article in English | MEDLINE | ID: mdl-27308302

ABSTRACT

Recombinant adeno-associated viruses (rAAV) have been widely used in gene therapy applications for central nervous system diseases. Though rAAV can efficiently target neurons and astrocytes in mouse brains, microglia, the immune cells of the brain, are refractile to rAAV. To identify AAV capsids with microglia-specific transduction properties, we initially screened the most commonly used serotypes, AAV1-9 and rh10, on primary mouse microglia cultures. While these capsids were not permissive, we then tested the microglial targeting properties of a newly characterized set of modified rAAV6 capsid variants with high tropism for monocytes. Indeed, these newly characterized rAAV6 capsid variants, specially a triply mutated Y731F/Y705F/T492V form, carrying a self-complementary genome and microglia-specific promoters (F4/80 or CD68) could efficiently and selectively transduce microglia in vitro. Delivery of these constructs in mice brains resulted in microglia-specific expression of green fluorescent protein, albeit at modest levels. We further show that CD68 promoter-driven expression of the inflammatory cytokine, interleukin-6, using this capsid variant leads to increased astrogliosis in the brains of wild-type mice. Our study describes the first instance of AAV-targeted microglial gene expression leading to functional modulation of the innate immune system in mice brains. This provides the rationale for utilizing these unique capsid/promoter combinations for microglia-specific gene targeting for modeling or functional studies.

12.
Mol Neurodegener ; 10: 25, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26152284

ABSTRACT

BACKGROUND: UBQLN2 mutations have recently been associated with familial forms of amyotrophic lateral sclerosis (ALS) and ALS-dementia. UBQLN2 encodes for ubiquilin-2, a member of the ubiquitin-like protein family which facilitates delivery of ubiquitinated proteins to the proteasome for degradation. To study the potential role of ubiquilin-2 in ALS, we used recombinant adeno-associated viral (rAAV) vectors to express UBQLN2 and three of the identified ALS-linked mutants (P497H, P497S, and P506T) in primary neuroglial cultures and in developing neonatal mouse brains. RESULTS: In primary cultures rAAV2/8-mediated expression of UBQLN2 mutants resulted in inclusion bodies and insoluble aggregates. Intracerebroventricular injection of FVB mice at post-natal day 0 with rAAV2/8 expressing wild type or mutant UBQLN2 resulted in widespread, sustained expression of ubiquilin-2 in brain. In contrast to wild type, mutant UBQLN2 expression induced significant pathology with large neuronal, cytoplasmic inclusions and ubiquilin-2-positive aggregates in surrounding neuropil. Ubiquilin-2 inclusions co-localized with ubiquitin, p62/SQSTM, optineurin, and occasionally TDP-43, but were negative for α-synuclein, neurofilament, tau, and FUS. Mutant UBLQN2 expression also resulted in Thioflavin-S-positive inclusions/aggregates. Mice expressing mutant forms of UBQLN2 variably developed a motor phenotype at 3-4 months, including nonspecific clasping and rotarod deficits. CONCLUSIONS: These findings demonstrate that UBQLN2 mutants (P497H, P497S, and P506T) induce proteinopathy and cause behavioral deficits, supporting a "toxic" gain-of-function, which may contribute to ALS pathology. These data establish also that our rAAV model can be used to rapidly assess the pathological consequences of various UBQLN2 mutations and provides an agile system to further interrogate the molecular mechanisms of ubiquilins in neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Cell Cycle Proteins/physiology , Mutation, Missense , Point Mutation , Ubiquitins/physiology , Adaptor Proteins, Signal Transducing , Animals , Animals, Newborn , Autophagy-Related Proteins , Brain/metabolism , Brain/pathology , Cell Cycle Proteins/genetics , Dependovirus/genetics , Disease Models, Animal , Genes, Synthetic , Genetic Vectors/toxicity , HEK293 Cells , Hand Strength , Humans , Inclusion Bodies/ultrastructure , Injections, Intraventricular , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rotarod Performance Test , Transduction, Genetic , Transgenes , Ubiquitination , Ubiquitins/genetics
13.
Mol Neurodegener ; 10: 32, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26223783

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder that is pathologically characterized by loss of dopaminergic neurons from the substantia nigra, the presence of aggregated α-synuclein (αS) and evidence of neuroinflammation. Experimental studies have shown that the cerebral injection of recombinant fibrillar αS, especially in αS transgenic mouse models, can induce the formation and spread of αS inclusion pathology. However, studies reporting this phenomenon did not consider the presence of lipopolysaccharide (LPS) in the injected αS, produced in E. coli, as a potential confound. The objectives of this study are to develop a method to remove the LPS contamination and investigate the differences in pathologies induced by αS containing LPS or αS highly purified of LPS. RESULTS AND CONCLUSIONS: We were able to remove >99.5% of the LPS contamination from the αS preparations through the addition of a cation exchange step during purification. The αS pathology induced by injection of fibrils produced from αS containing LPS or purified of LPS, showed a similar distribution pattern; however, there was less spread into the cortex of the mice injected with αS containing higher levels of LPS. As previously reported, injection of αS fibrils could induce astrogliosis, and αS inclusions were present within astrocytes in mice injected with fibrils comprised of αS with or without cation exchange purification. Furthermore, we identified the presence of αS pathology in ependymal cells in both groups of mice, which suggests the involvement of a novel mechanism for spread in this model of αS pathology.


Subject(s)
Endotoxins/pharmacology , Parkinsonian Disorders/chemically induced , alpha-Synuclein/toxicity , Animals , Astrocytes/pathology , Cell Count , Cells, Cultured , Chromatography, Ion Exchange , Disease Progression , Drug Contamination , Endotoxins/isolation & purification , Entorhinal Cortex/drug effects , Entorhinal Cortex/pathology , Escherichia coli/chemistry , Escherichia coli/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Humans , Hydrophobic and Hydrophilic Interactions , Inclusion Bodies/chemistry , Inflammation , Injections , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Parkinsonian Disorders/pathology , Plaque, Amyloid/chemistry , Protein Conformation , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity , alpha-Synuclein/administration & dosage , alpha-Synuclein/isolation & purification
14.
Mol Neurodegener ; 10: 29, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26169917

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) 42 has been implicated as the initiating molecule in the pathogenesis of Alzheimer's disease (AD); thus, therapeutic strategies that target Aß42 are of great interest. γ-Secretase modulators (GSMs) are small molecules that selectively decrease Aß42. We have previously reported that many acidic steroids are GSMs with potencies ranging in the low to mid micromolar concentration with 5ß-cholanic acid being the most potent steroid identified GSM with half maximal effective concentration (EC50) of 5.7 µM. RESULTS: We find that the endogenous cholesterol metabolite, 3ß-hydroxy-5-cholestenoic acid (CA), is a steroid GSM with enhanced potency (EC50 of 250 nM) relative to 5ß-cholanic acid. CA i) is found in human plasma at ~100-300 nM concentrations ii) has the typical acidic GSM signature of decreasing Aß42 and increasing Aß38 levels iii) is active in in vitro γ-secretase assay iv) is made in the brain. To test if CA acts as an endogenous GSM, we used Cyp27a1 knockout (Cyp27a1-/-) and Cyp7b1 knockout (Cyp7b1-/-) mice to investigate if manipulation of cholesterol metabolism pathways relevant to CA formation would affect brain Aß42 levels. Our data show that Cyp27a1-/- had increased brain Aß42, whereas Cyp7b1-/- mice had decreased brain Aß42 levels; however, peripheral dosing of up to 100 mg/kg CA did not affect brain Aß levels. Structure-activity relationship (SAR) studies with multiple known and novel CA analogs studies failed to reveal CA analogs with increased potency. CONCLUSION: These data suggest that CA may act as an endogenous GSM within the brain. Although it is conceptually attractive to try and increase the levels of CA in the brain for prevention of AD, our data suggest that this will not be easily accomplished.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cholesterol/analogs & derivatives , Peptide Fragments/metabolism , Animals , Blood-Brain Barrier , CHO Cells , Cells, Cultured , Cholestanetriol 26-Monooxygenase/deficiency , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/chemistry , Cholesterol/metabolism , Cholesterol/pharmacology , Cholic Acids/pharmacology , Coculture Techniques , Cricetinae , Cricetulus , Cytochrome P450 Family 7 , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Neuroglia/metabolism , Neurons/metabolism , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Structure-Activity Relationship
15.
FASEB J ; 29(10): 4384-98, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26156074

ABSTRACT

Inflammatory activation precedes and correlates with accumulating τ lesions in Alzheimer's disease and tauopathies. However, the relationship between neuroinflammation and etiology of pathologic τ remains elusive. To evaluate whether inflammatory signaling may promote or accelerate neurofibrillary tangle pathology, we explored the effect of recombinant adeno-associated virus (rAAV)-mediated overexpression of a master inflammatory cytokine, IFN-γ, on τ phosphorylation. In initial studies in primary neuroglial cultures, rAAV-mediated expression of IFN-γ did not alter endogenous τ production or paired helical filament τ phosphorylation. Next, we tested the effect of rAAV-mediated expression of IFN-γ in the brains of 2 mouse models of tauopathy: JNPL3 and rTg4510. In both models, IFN-γ increased 1) signal transducer and activator of transcription 1 levels and gliosis, and 2) hyperphosphorylation and conformational alterations of soluble τ compared with control cohorts. However, sarkosyl-insoluble phosphorylated τ levels and ubiquitin staining were unaltered in the IFN-γ cohorts. Notably, IFN-γ-induced τ hyperphosphorylation was associated with release of the inhibitory effect of glycogen synthase kinase 3ß function by decreasing Ser9 phosphorylation. Our data suggest that type II IFN signaling can promote τ phosphorylation by modulating cellular kinase activity, though this is insufficient in accelerating neuritic tangle pathology.


Subject(s)
Interferon-gamma/metabolism , Neurofibrillary Tangles/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Cells, Cultured , Dependovirus/genetics , Enzyme Activation , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunoblotting , Immunohistochemistry , Interferon-gamma/genetics , Male , Mice, Inbred C3H , Mice, Transgenic , Neurofibrillary Tangles/genetics , Neuroglia/metabolism , Phosphorylation , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Tauopathies/genetics
16.
PLoS One ; 10(6): e0128619, 2015.
Article in English | MEDLINE | ID: mdl-26046535

ABSTRACT

The signal peptide peptidases (SPPs) are biomedically important proteases implicated as therapeutic targets for hepatitis C (human SPP, (hSPP)), plasmodium (Plasmodium SPP (pSPP)), and B-cell immunomodulation and neoplasia (signal peptide peptidase like 2a, (SPPL2a)). To date, no drug-like, selective inhibitors have been reported. We use a recombinant substrate based on the amino-terminus of BRI2 fused to amyloid ß 1-25 (Aß1-25) (FBA) to develop facile, cost-effective SPP/SPPL protease assays. Co-transfection of expression plasmids expressing the FBA substrate with SPP/SPPLs were conducted to evaluate cleavage, which was monitored by ELISA, Western Blot and immunoprecipitation/MALDI-TOF Mass spectrometry (IP/MS). No cleavage is detected in the absence of SPP/SPPL overexpression. Multiple γ-secretase inhibitors (GSIs) and (Z-LL)2 ketone differentially inhibited SPP/SPPL activity; for example, IC50 of LY-411,575 varied from 51±79 nM (on SPPL2a) to 5499±122 nM (on SPPL2b), while Compound E showed inhibition only on hSPP with IC50 of 1465±93 nM. Data generated were predictive of effects observed for endogenous SPPL2a cleavage of CD74 in a murine B-Cell line. Thus, it is possible to differentially inhibit SPP family members. These SPP/SPPL cleavage assays will expedite the search for selective inhibitors. The data also reinforce similarities between SPP family member cleavage and cleavage catalyzed by γ-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Protease Inhibitors/chemistry , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/metabolism , Amino Acid Sequence , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Azepines/chemistry , Azepines/metabolism , Benzodiazepinones/chemistry , Benzodiazepinones/metabolism , Cell Line , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunoprecipitation , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protease Inhibitors/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
17.
EMBO J ; 34(12): 1674-86, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25964433

ABSTRACT

The biological underpinnings linking stress to Alzheimer's disease (AD) risk are poorly understood. We investigated how corticotrophin releasing factor (CRF), a critical stress response mediator, influences amyloid-ß (Aß) production. In cells, CRF treatment increases Aß production and triggers CRF receptor 1 (CRFR1) and γ-secretase internalization. Co-immunoprecipitation studies establish that γ-secretase associates with CRFR1; this is mediated by ß-arrestin binding motifs. Additionally, CRFR1 and γ-secretase co-localize in lipid raft fractions, with increased γ-secretase accumulation upon CRF treatment. CRF treatment also increases γ-secretase activity in vitro, revealing a second, receptor-independent mechanism of action. CRF is the first endogenous neuropeptide that can be shown to directly modulate γ-secretase activity. Unexpectedly, CRFR1 antagonists also increased Aß. These data collectively link CRF to increased Aß through γ-secretase and provide mechanistic insight into how stress may increase AD risk. They also suggest that direct targeting of CRF might be necessary to effectively modulate this pathway for therapeutic benefit in AD, as CRFR1 antagonists increase Aß and in some cases preferentially increase Aß42 via complex effects on γ-secretase.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/biosynthesis , Corticotropin-Releasing Hormone/metabolism , Models, Biological , Stress, Physiological/physiology , Alzheimer Disease/etiology , Analysis of Variance , Animals , Blotting, Western , Cyclic AMP/metabolism , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Hypothalamo-Hypophyseal System/physiology , Immunoprecipitation , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Pituitary-Adrenal System/physiology , Real-Time Polymerase Chain Reaction , Receptors, Corticotropin-Releasing Hormone/metabolism
18.
Neuron ; 85(3): 519-33, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25619653

ABSTRACT

Anti-inflammatory strategies are proposed to have beneficial effects in Alzheimer's disease. To explore how anti-inflammatory cytokine signaling affects Aß pathology, we investigated the effects of adeno-associated virus (AAV2/1)-mediated expression of Interleukin (IL)-10 in the brains of APP transgenic mouse models. IL-10 expression resulted in increased Aß accumulation and impaired memory in APP mice. A focused transcriptome analysis revealed changes consistent with enhanced IL-10 signaling and increased ApoE expression in IL-10-expressing APP mice. ApoE protein was selectively increased in the plaque-associated insoluble cellular fraction, likely because of direct interaction with aggregated Aß in the IL-10-expressing APP mice. Ex vivo studies also show that IL-10 and ApoE can individually impair glial Aß phagocytosis. Our observations that IL-10 has an unexpected negative effect on Aß proteostasis and cognition in APP mouse models demonstrate the complex interplay between innate immunity and proteostasis in neurodegenerative diseases, an interaction we call immunoproteostasis.


Subject(s)
Amyloid beta-Protein Precursor , Cognition Disorders/metabolism , Immunoproteins/biosynthesis , Interleukin-10/biosynthesis , Plaque, Amyloid/metabolism , Proteostasis Deficiencies/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Cells, Cultured , Cognition Disorders/immunology , HEK293 Cells , Humans , Mice , Mice, Transgenic , Plaque, Amyloid/immunology , Proteostasis Deficiencies/immunology
19.
Mol Ther ; 23(1): 53-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25228069

ABSTRACT

The architecture of the spinal cord makes efficient delivery of recombinant adeno-associated virus (rAAV) vectors throughout the neuraxis challenging. We describe a paradigm in which small amounts of virus delivered intraspinally to newborn mice result in robust rAAV-mediated transgene expression in the spinal cord. We compared the efficacy of rAAV2/1, 2/5, 2/8, and 2/9 encoding EGFP delivered to the hindlimb muscle (IM), cisterna magna (ICM), or lumbar spinal cord (IS) of neonatal pups. IS injection of all four capsids resulted in robust transduction of the spinal cord with rAAV2/5, 2/8, and 2/9 vectors appearing to be transported to brain. ICM injection resulted in widespread expression of EGFP in the brain, and upper spinal cord. IM injection resulted in robust muscle expression, with only rAAV2/8 and 2/9 transducing spinal motor and sensory neurons. As proof of concept, we use the IS paradigm to express murine Interleukin (IL)-10 in the spinal cord of the SOD1-G93A transgenic mouse model of amyotrophic lateral sclerosis. We show that expression of IL-10 in the spinal axis of SOD1-G93A mice altered the immune milieu and significantly prolonged survival. These data establish an efficient paradigm for somatic transgene delivery of therapeutic biologics to the spinal cord of mice.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Brain/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Spinal Cord/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Animals, Newborn , Brain/pathology , Capsid/metabolism , Cisterna Magna/metabolism , Cisterna Magna/pathology , Dependovirus/metabolism , Gene Expression , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Injections, Spinal , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transduction, Genetic
20.
Acta Neuropathol ; 127(5): 645-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24659240

ABSTRACT

In order to further evaluate the parameters whereby intracerebral administration of recombinant α-synuclein (αS) induces pathological phenotypes in mice, we conducted a series of studies where αS fibrils were injected into the brains of M83 (A53T) and M47 (E46K) αS transgenic (Tg) mice, and non-transgenic (nTg) mice. Using multiple markers to assess αS inclusion formation, we find that injected fibrillar human αS induced widespread cerebral αS inclusion formation in the M83 Tg mice, but in both nTg and M47 Tg mice, induced αS inclusion pathology is largely restricted to the site of injection. Furthermore, mouse αS fibrils injected into nTg mice brains also resulted in inclusion pathology restricted to the site of injection with no evidence for spread. We find no compelling evidence for extensive spread of αS pathology within white matter tracts, and we attribute previous reports of white matter tract spreading to cross-reactivity of the αS pSer129/81A antibody with phosphorylated neurofilament subunit L. These studies suggest that, with the exception of the M83 Tg mice which appear to be uniquely susceptible to induction of inclusion pathology by exogenous forms of αS, there are significant barriers in mice to widespread induction of αS pathology following intracerebral administration of amyloidogenic αS.


Subject(s)
Amyloidosis/metabolism , Amyloidosis/pathology , Brain/metabolism , Brain/pathology , alpha-Synuclein/metabolism , Animals , Cells, Cultured , Escherichia coli , Gene Transfer Techniques , Humans , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Neural Pathways/metabolism , Neural Pathways/pathology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Recombinant Proteins/metabolism , Species Specificity , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...