Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Neurosci ; 42(40): 7673-7688, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36333098

ABSTRACT

As the CNS-resident macrophages and member of the myeloid lineage, microglia fulfill manifold functions important for brain development and homeostasis. In the context of neurodegenerative diseases, they have been implicated in degenerative and regenerative processes. The discovery of distinct activation patterns, including increased phagocytosis, indicated a damaging role of myeloid cells in multiple system atrophy (MSA), a devastating, rapidly progressing atypical parkinsonian disorder. Here, we analyzed the gene expression profile of microglia in a mouse model of MSA (MBP29-hα-syn) and identified a disease-associated expression profile and upregulation of the colony-stimulating factor 1 (Csf1). Thus, we hypothesized that CSF1 receptor-mediated depletion of myeloid cells using PLX5622 modifies the disease progression and neuropathological phenotype in this mouse model. Intriguingly, sex-balanced analysis of myeloid cell depletion in MBP29-hα-syn mice revealed a two-faced outcome comprising an improved survival rate accompanied by a delayed onset of neurological symptoms in contrast to severely impaired motor functions. Furthermore, PLX5622 reversed gene expression profiles related to myeloid cell activation but reduced gene expression associated with transsynaptic signaling and signal release. While transcriptional changes were accompanied by a reduction of dopaminergic neurons in the SNpc, striatal neuritic density was increased upon myeloid cell depletion in MBP29-hα-syn mice. Together, our findings provide insight into the complex, two-faced role of myeloid cells in the context of MSA emphasizing the importance to carefully balance the beneficial and adverse effects of CSF1R inhibition in different models of neurodegenerative disorders before its clinical translation.SIGNIFICANCE STATEMENT Myeloid cells have been implicated as detrimental in the disease pathogenesis of multiple system atrophy. However, long-term CSF1R-dependent depletion of these cells in a mouse model of multiple system atrophy demonstrates a two-faced effect involving an improved survival associated with a delayed onset of disease and reduced inflammation which was contrasted by severely impaired motor functions, synaptic signaling, and neuronal circuitries. Thus, this study unraveled a complex role of myeloid cells in multiple system atrophy, which indicates important functions beyond the previously described disease-associated, destructive phenotype and emphasized the need of further investigation to carefully and individually fine-tune immunologic processes in different neurodegenerative diseases.


Subject(s)
Multiple System Atrophy , Animals , Mice , Multiple System Atrophy/genetics , Longevity , Organic Chemicals/pharmacology , Microglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Disease Models, Animal , Myeloid Cells/metabolism , Receptors, Colony-Stimulating Factor
2.
Neurobiol Dis ; 178: 106010, 2023 03.
Article in English | MEDLINE | ID: mdl-36702318

ABSTRACT

Mutations or triplication of the alpha synuclein (ASYN) gene contribute to synucleinopathies including Parkinson's disease (PD), Dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Recent evidence suggests that ASYN also plays an important role in amyloid-induced neurotoxicity, although the mechanism(s) remains unknown. One hypothesis is that accumulation of ASYN alters endolysosomal pathways to impact axonal trafficking and processing of the amyloid precursor protein (APP). To define an axonal function for ASYN, we used a transgenic mouse model of synucleinopathy that expresses a GFP-human ASYN (GFP-hASYN) transgene and an ASYN knockout (ASYN-/-) mouse model. Our results demonstrate that expression of GFP-hASYN in primary neurons derived from a transgenic mouse impaired axonal trafficking and processing of APP. In addition, axonal transport of BACE1, Rab5, Rab7, lysosomes and mitochondria were also reduced in these neurons. Interestingly, axonal transport of these organelles was also affected in ASYN-/- neurons, suggesting that ASYN plays an important role in maintaining normal axonal transport function. Therefore, selective impairment of trafficking and processing of APP by ASYN may act as a potential mechanism to induce pathological features of Alzheimer's disease (AD) in PD patients.


Subject(s)
Parkinson Disease , Synucleinopathies , Humans , Mice , Animals , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/genetics , Aspartic Acid Endopeptidases , Parkinson Disease/genetics , Mice, Transgenic , Lysosomes/metabolism
3.
J Neurovirol ; 29(5): 564-576, 2023 10.
Article in English | MEDLINE | ID: mdl-37801175

ABSTRACT

Central nervous system (CNS) dysfunction remains prevalent in people with HIV (PWH) despite effective antiretroviral therapy (ART). There is evidence that low-level HIV infection and ART drugs may contribute to CNS damage in the brain of PWH with suppressed viral loads. As cannabis is used at a higher rate in PWH compared to the general population, there is interest in understanding how HIV proteins and ART drugs interact with the endocannabinoid system (ECS) and inflammation in the CNS. Therefore, we investigated the effects of the HIV envelope protein gp120 and tenofovir alafenamide (TAF) on cannabinoid receptor 1 (CB1R), glial fibrillary acidic protein (GFAP), and IBA1 in the brain and on locomotor activity in mice. The gp120 transgenic (tg) mouse model was administered TAF daily for 30 days and then analyzed using the open field test before being euthanized, and their brains were analyzed for CB1R, GFAP, and IBA1 expression using immunohistochemical approaches. CB1R expression levels were significantly increased in CA1, CA2/3, and dentate gyrus of gp120tg mice compared to wt littermates; TAF reversed these effects. As expected, TAF showed a medium effect of enhancing GFAP in the frontal cortex of gp120tg mice in the frontal cortex. TAF had minimal effect on IBA1 signal. TAF showed medium to large effects on fine movements, rearing, total activity, total distance, and lateral activity in the open-field test. These findings suggest that TAF may reverse gp120-induced effects on CB1R expression and, unlike tenofovir disoproxil fumarate (TDF), may not affect gliosis in the brain.


Subject(s)
Anti-HIV Agents , HIV Infections , Humans , Mice , Animals , HIV Infections/drug therapy , HIV Infections/genetics , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , HIV Envelope Protein gp120/genetics , Adenine/pharmacology , Mice, Transgenic , Hippocampus , Receptors, Cannabinoid/therapeutic use
4.
Neurobiol Dis ; 152: 105277, 2021 05.
Article in English | MEDLINE | ID: mdl-33516874

ABSTRACT

The microtubule-associated protein tau is implicated in multiple degenerative diseases including retinal diseases such as glaucoma; however, the way tau initiates retinopathy is unclear. Previous retinal assessments in mouse models of tauopathy suggest that mutations in four-repeat (4R) tau are associated with disease-induced retinal dysfunction, while shifting tau isoform ratio to favor three-repeat (3R) tau production enhanced photoreceptor function. To further understand how alterations in tau expression impact the retina, we analyzed the retinas of transgenic mice overexpressing mutant 3R tau (m3R tau-Tg), a model known to exhibit Pick's Disease pathology in the brain. Analysis of retinal cross-sections from young (3 month) and adult (9 month) mice detected asymmetric 3R tau immunoreactivity in m3R tau-Tg retina, concentrated in the retinal ganglion and amacrine cells of the dorsal retinal periphery. Accumulation of hyperphosphorylated tau was detected specifically in the detergent insoluble fraction of the adult m3R tau-Tg retina. RNA-seq analysis highlighted biological pathways associated with tauopathy that were uniquely altered in m3R tau-Tg retina. The upregulation of transcript encoding apoptotic protease caspase-2 coincided with increased immunostaining in predominantly 3R tau positive retinal regions. In adult m3R tau-Tg, the dorsal peripheral retina of the adult m3R tau-Tg exhibited decreased cell density in the ganglion cell layer (GCL) and reduced thickness of the inner plexiform layer (IPL) compared to the ventral peripheral retina. Together, these data indicate that mutant 3R tau may mediate toxicity in retinal ganglion cells (RGC) by promoting caspase-2 expression which results in RGC degeneration. The m3R tau-Tg line has the potential to be used to assess tau-mediated RGC degeneration and test novel therapeutics for degenerative diseases such as glaucoma.


Subject(s)
Caspase 2/metabolism , Retinal Diseases/pathology , Retinal Ganglion Cells/pathology , Tauopathies/pathology , tau Proteins/metabolism , Animals , Cell Death , Female , Humans , Male , Mice , Mice, Transgenic , Mutation , Protein Isoforms , Retinal Diseases/metabolism , Retinal Ganglion Cells/metabolism , tau Proteins/genetics
5.
Traffic ; 19(11): 840-853, 2018 11.
Article in English | MEDLINE | ID: mdl-30120810

ABSTRACT

The cytosolic chaperonin T-complex protein (TCP) 1-ring complex (TRiC) has been shown to exert neuroprotective effects on axonal transport through clearance of mutant Huntingtin (mHTT) in Huntington's disease. However, it is presently unknown if TRiC also has any effect on axonal transport in wild-type neurons. Here, we examined how TRiC impacted the retrograde axonal transport of brain-derived neurotrophic factor (BDNF). We found that expression of a single TRiC subunit significantly enhanced axonal transport of BDNF, leading to an increase in instantaneous velocity with a concomitant decrease in pauses for retrograde BDNF transport. The transport enhancing effect by TRiC was dependent on endogenous tau expression because no effect was seen in neurons from tau knockout mice. We showed that TRiC regulated the level of cyclin-dependent kinase 5 (CDK5)/p35 positively, contributing to TRiC-mediated tau phosphorylation (ptau). Expression of a single TRiC subunit increased the level of ptau while downregulation of the TRiC complex decreased ptau. We further demonstrated that TRiC-mediated increase in ptau induced detachment of tau from microtubules. Our study has thus revealed that TRiC-mediated increase in tau phosphorylation impacts retrograde axonal transport.


Subject(s)
Axonal Transport , Chaperonin Containing TCP-1/metabolism , tau Proteins/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , CHO Cells , Cells, Cultured , Chaperonin Containing TCP-1/genetics , Cricetinae , Cricetulus , Cyclin-Dependent Kinase 5/metabolism , HEK293 Cells , Humans , Microtubules/metabolism , Phosphorylation , Rats
6.
J Neurosci ; 38(4): 1000-1014, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29246926

ABSTRACT

Dementia with Lewy bodies, Parkinson's disease, and Multiple System Atrophy are age-related neurodegenerative disorders characterized by progressive accumulation of α-synuclein (α-syn) and jointly termed synucleinopathies. Currently, no disease-modifying treatments are available for these disorders. Previous preclinical studies demonstrate that active and passive immunizations targeting α-syn partially ameliorate behavioral deficits and α-syn accumulation; however, it is unknown whether combining humoral and cellular immunization might act synergistically to reduce inflammation and improve microglial-mediated α-syn clearance. Since combined delivery of antigen plus rapamycin (RAP) in nanoparticles is known to induce antigen-specific regulatory T cells (Tregs), we adapted this approach to α-syn using the antigen-presenting cell-targeting glucan microparticle (GP) vaccine delivery system. PDGF-α-syn transgenic (tg) male and female mice were immunized with GP-alone, GP-α-syn (active humoral immunization), GP+RAP, or GP+RAP/α-syn (combined active humoral and Treg) and analyzed using neuropathological and biochemical markers. Active immunization resulted in higher serological total IgG, IgG1, and IgG2a anti-α-syn levels. Compared with mice immunized with GP-alone or GP-α-syn, mice vaccinated with GP+RAP or GP+RAP/α-syn displayed increased numbers of CD25-, FoxP3-, and CD4-positive cells in the CNS. GP-α-syn or GP+RAP/α-syn immunizations resulted in a 30-45% reduction in α-syn accumulation, neuroinflammation, and neurodegeneration. Mice immunized with GP+RAP/α-syn further rescued neurons and reduced neuroinflammation. Levels of TGF-ß1 were increased with GP+RAP/α-syn immunization, while levels of TNF-α and IL-6 were reduced. We conclude that the observed effects of GP+RAP/α-syn immunization support the hypothesis that cellular immunization may enhance the effects of active immunotherapy for the treatment of synucleinopathies.SIGNIFICANCE STATEMENT We show that a novel vaccination modality combining an antigen-presenting cell-targeting glucan particle (GP) vaccine delivery system with encapsulated antigen (α-synuclein) + rapamycin (RAP) induced both strong anti-α-synuclein antibody titers and regulatory T cells (Tregs). This vaccine, collectively termed GP+RAP/α-syn, is capable of triggering neuroprotective Treg responses in synucleinopathy models, and the combined vaccine is more effective than the humoral or cellular immunization alone. Together, these results support the further development of this multifunctional vaccine approach for the treatment of synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple systems atrophy.


Subject(s)
Neurodegenerative Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Vaccination/methods , alpha-Synuclein/immunology , Animals , Female , Glucans/administration & dosage , Glucans/immunology , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunosuppressive Agents/administration & dosage , Male , Mice , Mice, Transgenic , Nanoparticles , Sirolimus/administration & dosage , alpha-Synuclein/administration & dosage
7.
Neurobiol Dis ; 127: 163-177, 2019 07.
Article in English | MEDLINE | ID: mdl-30849508

ABSTRACT

Neurodegenerative disorders of the aging population are characterized by progressive accumulation of neuronal proteins such as α-synuclein (α-syn) in Parkinson's Disease (PD) and Amyloid ß (Aß) and Tau in Alzheimer's disease (AD) for which no treatments are currently available. The ability to regulate the expression at the gene transcription level would be beneficial for reducing the accumulation of these proteins or regulating expression levels of other genes in the CNS. Short interfering RNA molecules can bind specifically to target RNAs and deliver them for degradation. This approach has shown promise therapeutically in vitro and in vivo in mouse models of PD and AD and other neurological disorders; however, delivery of the siRNA to the CNS in vivo has been achieved primarily through intra-cerebral or intra-thecal injections that may be less amenable for clinical translation; therefore, alternative approaches for delivery of siRNAs to the brain is needed. Recently, we described a small peptide from the envelope protein of the rabies virus (C2-9r) that was utilized to deliver an siRNA targeting α-syn across the blood brain barrier (BBB) following intravenous injection. This approach showed reduced expression of α-syn and neuroprotection in a toxic mouse model of PD. However, since receptor-mediated delivery is potentially saturable, each allowing the delivery of a limited number of molecules, we identified an alternative peptide for the transport of nucleotides across the BBB based on the apolipoprotein B (apoB) protein targeted to the family of low-density lipoprotein receptors (LDL-R). We used an 11-amino acid sequence from the apoB protein (ApoB11) that, when coupled with a 9-amino acid arginine linker, can transport siRNAs across the BBB to neuronal and glial cells. To examine the value of this peptide mediated oligonucleotide delivery system for PD, we delivered an siRNA targeting the α-syn (siα-syn) in a transgenic mouse model of PD. We found that ApoB11 was effective (comparable to C2-9r) at mediating the delivery of siα-syn into the CNS, co-localized to neurons and glial cells and reduced levels of α-syn protein translation and accumulation. Delivery of ApoB11/siα-syn was accompanied by protection from degeneration of selected neuronal populations in the neocortex, limbic system and striato-nigral system and reduced neuro-inflammation. Taken together, these results suggest that systemic delivery of oligonucleotides targeting α-syn using ApoB11 might be an interesting alternative strategy worth considering for the experimental treatment of synucleinopathies.


Subject(s)
Lewy Body Disease/therapy , Nerve Degeneration/therapy , alpha-Synuclein/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Genetic Vectors , Lewy Body Disease/genetics , Lewy Body Disease/metabolism , Mice , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurons/metabolism , RNA, Small Interfering/administration & dosage , Receptors, LDL/genetics , Receptors, LDL/metabolism , alpha-Synuclein/genetics
10.
Alzheimers Dement ; 15(9): 1133-1148, 2019 09.
Article in English | MEDLINE | ID: mdl-31378574

ABSTRACT

INTRODUCTION: Immunotherapeutic approaches targeting amyloid ß (Aß) protein and tau in Alzheimer's disease and α-synuclein (α-syn) in Parkinson's disease are being developed for treating dementia with Lewy bodies. However, it is unknown if single or combined immunotherapies targeting Aß and/or α-syn may be effective. METHODS: Amyloid precursor protein/α-syn tg mice were immunized with AFFITOPEs® (AFF) peptides specific to Aß (AD02) or α-syn (PD-AFF1) and the combination. RESULTS: AD02 more effectively reduced Aß and pTau burden; however, the combination exhibited some additive effects. Both AD02 and PD-AFF1 effectively reduced α-syn, ameliorated degeneration of pyramidal neurons, and reduced neuroinflammation. PD-AFF1 more effectively ameliorated cholinergic and dopaminergic fiber loss; the combined immunization displayed additive effects. AD02 more effectively improved buried pellet test behavior, whereas PD-AFF1 more effectively improved horizontal beam test; the combined immunization displayed additive effects. DISCUSSION: Specific active immunotherapy targeting Aß and/or α-syn may be of potential interest for the treatment of dementia with Lewy bodies.


Subject(s)
Amyloid beta-Peptides/immunology , Immunotherapy , Lewy Body Disease/immunology , alpha-Synuclein/immunology , Alzheimer Disease , Animals , Humans , Immunologic Factors , Mice , Parkinson Disease
11.
Acta Neuropathol ; 136(1): 69-87, 2018 07.
Article in English | MEDLINE | ID: mdl-29934874

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia in the elderly affecting more than 5 million people in the U.S. AD is characterized by the accumulation of ß-amyloid (Aß) and Tau in the brain, and is manifested by severe impairments in memory and cognition. Therefore, removing tau pathology has become one of the main therapeutic goals for the treatment of AD. Tau (tubulin-associated unit) is a major neuronal cytoskeletal protein found in the CNS encoded by the gene MAPT. Alternative splicing generates two major isoforms of tau containing either 3 or 4 repeat (R) segments. These 3R or 4RTau species are differentially expressed in neurodegenerative diseases. Previous studies have been focused on reducing Tau accumulation with antibodies against total Tau, 4RTau or phosphorylated isoforms. Here, we developed a brain penetrating, single chain antibody that specifically recognizes a pathogenic 3RTau. This single chain antibody was modified by the addition of a fragment of the apoB protein to facilitate trafficking into the brain, once in the CNS these antibody fragments reduced the accumulation of 3RTau and related deficits in a transgenic mouse model of tauopathy. NMR studies showed that the single chain antibody recognized an epitope at aa 40-62 of 3RTau. This single chain antibody reduced 3RTau transmission and facilitated the clearance of Tau via the endosomal-lysosomal pathway. Together, these results suggest that targeting 3RTau with highly specific, brain penetrating, single chain antibodies might be of potential value for the treatment of tauopathies such as Pick's Disease.


Subject(s)
Alzheimer Disease/drug therapy , DNA Repeat Expansion/genetics , Pick Disease of the Brain/drug therapy , Single-Chain Antibodies/therapeutic use , tau Proteins/genetics , tau Proteins/immunology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Apolipoproteins B/metabolism , Brain/drug effects , Brain/metabolism , Cell Line, Transformed , Coculture Techniques , Disease Models, Animal , Exploratory Behavior/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neuroblastoma/pathology , Phosphorylation , Pick Disease of the Brain/genetics , Pick Disease of the Brain/pathology , Protein Transport/drug effects , Protein Transport/genetics , rab5 GTP-Binding Proteins/metabolism , tau Proteins/metabolism
12.
J Neurosci ; 36(30): 7971-84, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27466341

ABSTRACT

UNLABELLED: Alzheimer's disease (AD) is characterized by the progressive accumulation of amyloid ß (Aß) and microtubule associate protein tau, leading to the selective degeneration of neurons in the neocortex, limbic system, and nucleus basalis, among others. Recent studies have shown that α-synuclein (α-syn) also accumulates in the brains of patients with AD and interacts with Aß and tau, forming toxic hetero-oligomers. Although the involvement of α-syn has been investigated extensively in Lewy body disease, less is known about the role of this synaptic protein in AD. Here, we found that reducing endogenous α-syn in an APP transgenic mouse model of AD prevented the degeneration of cholinergic neurons, ameliorated corresponding deficits, and recovered the levels of Rab3a and Rab5 proteins involved in intracellular transport and sorting of nerve growth factor and brain-derived neurotrophic factor. Together, these results suggest that α-syn might participate in mechanisms of vulnerability of selected neuronal populations in AD and that reducing α-syn might be a potential approach to protecting these populations from the toxic effects of Aß. SIGNIFICANCE STATEMENT: Reducing endogenous α-synuclein (α-syn) in an APP transgenic mouse model of Alzheimer's disease (AD) prevented the degeneration of cholinergic neurons, ameliorated corresponding deficits, and recovered the levels of Rab3a and Rab5 proteins involved in intracellular transport and sorting of nerve growth factor and brain-derived neurotrophic factor. These results suggest that α-syn might participate in mechanisms of vulnerability of selected neuronal populations in AD and that reducing α-syn might be a potential approach to protecting these populations from the toxic effects of amyloid ß.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Neurons/metabolism , Neurons/pathology , alpha-Synuclein/metabolism , Animals , Brain/pathology , Down-Regulation/genetics , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , alpha-Synuclein/genetics , rab3A GTP-Binding Protein/metabolism , rab5 GTP-Binding Proteins/metabolism
13.
Neurobiol Dis ; 104: 85-96, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28476636

ABSTRACT

Disorders with progressive accumulation of α-synuclein (α-syn) are a common cause of dementia and parkinsonism in the aging population. Accumulation and propagation of α-syn play a role in the pathogenesis of these disorders. Previous studies have shown that immunization with antibodies that recognize C-terminus of α-syn reduces the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy. These studies employed antibodies that recognize epitopes within monomeric and aggregated α-syn that were generated through active immunization or administered via passive immunization. However, it is possible that more specific effects might be achieved with antibodies recognizing selective species of the α-syn aggregates. In this respect we recently developed antibodies that differentially recognized various oligomers (Syn-O1, -O2, and -O4) and fibrilar (Syn-F1 and -F2) forms of α-syn. For this purpose wild-type α-syn transgenic (line 61) mice were immunized with these 5 different antibodies and neuropathologically and biochemically analyzed to determine which was most effective at reducing α-syn accumulation and related deficits. We found that Syn-O1, -O4 and -F1 antibodies were most effective at reducing accumulation of α-syn oligomers in multiple brain regions and at preventing neurodegeneration. Together this study supports the notion that selective antibodies against α-syn might be suitable for development new treatments for synucleinopathies such as PD and DLB.


Subject(s)
Dementia/therapy , Immunotherapy/methods , Parkinsonian Disorders/therapy , alpha-Synuclein/immunology , alpha-Synuclein/metabolism , Analysis of Variance , Animals , Antibodies/therapeutic use , Calcium-Binding Proteins/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Dementia/genetics , Dementia/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/physiology , Female , Glial Fibrillary Acidic Protein/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microscopy, Confocal , Neuroblastoma/pathology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/immunology , Synaptophysin/metabolism , alpha-Synuclein/genetics
14.
J Neurovirol ; 23(2): 290-303, 2017 04.
Article in English | MEDLINE | ID: mdl-28105557

ABSTRACT

Despite the success of antiretroviral therapies to control systemic HIV-1 infection, the prevalence of HIV-associated neurocognitive disorders (HANDs) has not decreased among aging patients with HIV. Autophagy pathway alterations, triggered by HIV-1 proteins including gp120, Tat, and Nef, might contribute to the neurodegenerative process in aging patients with HAND. Although no treatments are currently available to manage HAND, we have previously shown that sunitinib, an anticancer drug that blocks receptor tyrosine-kinase and cyclin kinase pathways, might be of interest. Studies in cancer models suggest that sunitinib might also modulate autophagy, which is dysregulated in our models of Tat-induced neurotoxicity. We evaluated the efficacy of sunitinib to promote autophagy in the CNS and ameliorate neurodegeneration using LC3-GFP-expressing neuronal cells challenged with low concentrations of Tat and using inducible Tat transgenic mice. In neuronal cultures challenged with low levels of Tat, sunitinib increased markers of autophagy such as LC3-II and reduced p62 accumulation in a dose-dependent manner. In vivo, sunitinib treatment restored LC3-II, p62, and endophilin B1 (EndoB1) levels in doxycycline-induced Tat transgenic mice. Moreover, in these animals, sunitinib reduced the hyperactivation of CDK5, tau hyperphosphorylation, and p35 cleavage to p25. Restoration of CDK5 and autophagy were associated with reduced neurodegeneration and behavioral alterations. Alterations in autophagy in the Tat tg mice were associated with reduced levels of a CDK5 substrate, EndoB1, and levels of total EndoB1 were normalized by sunitinib treatment. We conclude that sunitinib might ameliorate Tat-mediated autophagy alterations and may decrease neurodegeneration in aging patients with HAND.


Subject(s)
Antineoplastic Agents/pharmacology , Cognitive Dysfunction/drug therapy , HIV Infections/drug therapy , Indoles/pharmacology , Pyrroles/pharmacology , Transgenes , tat Gene Products, Human Immunodeficiency Virus/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Autophagy/drug effects , Autophagy/genetics , Cognitive Dysfunction/complications , Cognitive Dysfunction/genetics , Cognitive Dysfunction/virology , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Regulation , HIV Infections/complications , HIV Infections/genetics , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , HIV-1/metabolism , Humans , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/virology , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Sunitinib , tat Gene Products, Human Immunodeficiency Virus/metabolism
15.
J Neuroinflammation ; 13(1): 120, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27220536

ABSTRACT

BACKGROUND: HIV-associated neurocognitive disorders (HAND) continue to be a common morbidity associated with chronic HIV infection. It has been shown that HIV proteins (e.g., gp120) released from infected microglial/macrophage cells can cause neuronal damage by triggering inflammation and oxidative stress, activating aberrant kinase pathways, and by disrupting mitochondrial function and biogenesis. Previous studies have shown that FK506, an immunophilin ligand that modulates inflammation and mitochondrial function and inhibits calcineurin, is capable of rescuing the neurodegenerative pathology in models of Parkinson's disease, Alzheimer's disease, and Huntington's disease. In this context, the main objective of this study was to evaluate if FK506 could rescue the neuronal degeneration and mitochondrial alterations in a transgenic (tg) animal model of HIV1-gp120 neurotoxicity. METHODS: GFAP-gp120 tg mice were treated with FK506 and analyzed for neuropathology, behavior, mitochondrial markers, and calcium flux by two-photon microscopy. RESULTS: We found that FK506 reduced the neuronal cell loss and neuro-inflammation in the gp120 tg mice. Moreover, while vehicle-treated gp120 tg mice displayed damaged mitochondria and increased neuro-inflammatory markers, FK506 rescued the morphological mitochondrial alterations and neuro-inflammation while increasing levels of optic atrophy 1 and mitofusin 1. By two-photon microscopy, calcium levels were not affected in the gp120 tg mice and no effects of FK506 were detected. However, at a functional level, FK506 ameliorated the gp120 tg mice hyperactivity in the open field. CONCLUSIONS: Together, these results suggest that FK506 might be potentially neuroprotective in patients with HAND by mitigating inflammation and mitochondrial alterations.


Subject(s)
HIV Envelope Protein gp120/toxicity , Immunosuppressive Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Tacrolimus/therapeutic use , Analysis of Variance , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Encephalitis/drug therapy , Interleukin-6/metabolism , Mice , Microfilament Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Tissue Proteins/metabolism , Neurotoxicity Syndromes/complications , Tacrolimus Binding Proteins/metabolism , Treatment Outcome
16.
BMC Neurosci ; 16: 85, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26611895

ABSTRACT

BACKGROUND: Tauopathies are a group of neurodegenerative disorders with accumulation of three-repeat (3R) or four-repeat (4R) Tau. While 3R tau is found in Pick's disease and Alzheimer's disease (AD), 4R tau is more abundant in corticobasal degeneration, progressive supranuclear palsy, and AD. We have previously shown that Cerebrolysin™ (CBL), a neuropeptide mixture with neurotrophic effects, ameliorates the pathology in amyloid precursor protein transgenic (tg) mouse model of AD and 4R tau, however it is unclear if CBL ameliorates the deficits and neuropathology in the mouse model of Pick's disease over expressing 3R tau. RESULTS: Mice expressing 3R tau (L266V and G272V mutations) under the mThy-1 promoter were treated with CBL in two separate groups, the first was 3 months old (treated for 3 months, IP) and the second was 6 months old (treated for 3 months, IP) at the start of the treatment. We found that although the levels of total 3R tau were unchanged, CBL reduced the levels of hyper-phosphorylated tau in both groups of mice. This was accompanied by reduced neurodegenerative pathology in the neocortex and hippocampus in both groups and by improvements in the behavioral deficits in the nest-building test and water maze in the 3-6 month group. CONCLUSION: Taken together these results support the notion that CBL may be beneficial in other taupathy models by reducing the levels of aberrantly phosphorylated tau.


Subject(s)
Amino Acids/pharmacology , Neuroprotective Agents/pharmacology , Pick Disease of the Brain/drug therapy , Tauopathies/drug therapy , Aging/drug effects , Aging/metabolism , Aging/pathology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Phosphorylation/drug effects , Pick Disease of the Brain/metabolism , Pick Disease of the Brain/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/genetics , tau Proteins/metabolism
17.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33651103

ABSTRACT

A potent γ-secretase modulator (GSM) has been developed to circumvent problems associated with γ-secretase inhibitors (GSIs) and to potentially enable use in primary prevention of early-onset familial Alzheimer's disease (EOFAD). Unlike GSIs, GSMs do not inhibit γ-secretase activity but rather allosterically modulate γ-secretase, reducing the net production of Aß42 and to a lesser extent Aß40, while concomitantly augmenting production of Aß38 and Aß37. This GSM demonstrated robust time- and dose-dependent efficacy in acute, subchronic, and chronic studies across multiple species, including primary and secondary prevention studies in a transgenic mouse model. The GSM displayed a >40-fold safety margin in rats based on a comparison of the systemic exposure (AUC) at the no observed adverse effect level (NOAEL) to the 50% effective AUC or AUCeffective, the systemic exposure required for reducing levels of Aß42 in rat brain by 50%.


Subject(s)
Alzheimer Disease/enzymology , Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/metabolism , Phenethylamines/administration & dosage , Pyridazines/administration & dosage , Signal Transduction/drug effects , Amyloid beta-Peptides/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/metabolism , Neuroblastoma/pathology , Peptide Fragments/metabolism , Rats , Rats, Sprague-Dawley , Treatment Outcome
18.
Acta Neuropathol Commun ; 8(1): 123, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753049

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

19.
Front Neurosci ; 14: 286, 2020.
Article in English | MEDLINE | ID: mdl-32296304

ABSTRACT

Progressive accumulation of the pre-synaptic protein α-synuclein (α-syn) has been strongly associated with the pathogenesis of neurodegenerative disorders of the aging population such as Alzheimer's disease (AD), Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy. While the precise mechanisms are not fully understood, alterations in kinase pathways including that of mitogen activated protein kinase (MAPK) p38 have been proposed to play a role. In AD, p38α activation has been linked to neuro-inflammation while alterations in p38γ have been associated with tau phosphorylation. Although p38 has been studied in AD, less is known about its role in DLB/PD and other α-synucleinopathies. For this purpose, we investigated the expression of the p38 family in brains from α-syn overexpressing transgenic mice (α-syn Tg: Line 61) and patients with DLB/PD. Immunohistochemical analysis revealed that in healthy human controls and non-Tg mice, p38α associated with neurons and astroglial cells and p38γ localized to pre-synaptic terminals. In DLB and α-syn Tg brains, however, p38α levels were increased in astroglial cells while p38γ immunostaining was redistributed from the synaptic terminals to the neuronal cell bodies. Double immunolabeling further showed that p38γ colocalized with α-syn aggregates in DLB patients, and immunoblot and qPCR analysis confirmed the increased levels of p38α and p38γ. α1-syntrophin, a synaptic target of p38γ, was present in the neuropil and some neuronal cell bodies in human controls and non-Tg mice. In DLB and and Tg mice, however, α1-syntrophin was decreased in the neuropil and instead colocalized with α-syn in intra-neuronal inclusions. In agreement with these findings, in vitro studies showed that α-syn co-immunoprecipitates with p38γ, but not p38α. These results suggest that α-syn might interfere with the p38γ pathway and play a role in the mechanisms of synaptic dysfunction in DLB/PD.

20.
J Alzheimers Dis ; 72(2): 575-585, 2019.
Article in English | MEDLINE | ID: mdl-31594233

ABSTRACT

Extracellular vesicles (EVs) are a heterogeneous group of secreted particles consisting of microvesicles, which are released by budding of the cellular membrane, and exosomes, which are secreted through exocytosis from multivesicular bodies. EV cargo consists of a wide range of proteins and nucleic acids that can be transferred between cells. Importantly, EVs may be pathogenically involved in neurodegenerative diseases such as Alzheimer's disease (AD). While EVs derived from AD neurons have been found to be neurotoxic in vitro, little is known about the pathological consequences of AD EVs in vivo. Furthermore, although all known familial AD (fAD) mutations involve either amyloid-ß protein precursor (AßPP) or the machinery that processes AßPP, hyperphosphorylation of the microtubule associated protein tau appears to play a critical role in fAD-associated neurodegeneration, and previous reports suggest EVs may propagate tau pathology in the AD brain. Therefore, we hypothesized that fAD EVs may have a mechanistic involvement in the development of fAD-associated tau pathology. To test this, we isolated EVs from iPSC-derived neuronal cultures generated from an fAD patient harboring a A246E mutation to presenilin-1 and stereotactically injected these EVs into the hippocampi of wild-type C57BL/6 mice. Five weeks after injection, mice were euthanized and pathology evaluated. Mice injected with fAD EVs displayed increased tau phosphorylation at multiple sites relative to PBS and non-disease control EV injected groups. Moreover, fAD EV injected hippocampi contained significantly more tau inclusions in the CA1 hippocampal neuronal field than controls. In total, these findings identify EVs as a potential mediator of fAD-associated tau dysregulation and warrant future studies to investigate the therapeutic potential of EV-targeted treatments for fAD.


Subject(s)
Alzheimer Disease/metabolism , Extracellular Vesicles , Neurons/metabolism , tau Proteins/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Cells, Cultured , Humans , Induced Pluripotent Stem Cells , Mice , Mice, Inbred C57BL , Mutation/genetics , Nanoparticles , Phosphorylation , Presenilin-1/genetics , Tauopathies/metabolism , Tauopathies/pathology
SELECTION OF CITATIONS
SEARCH DETAIL