Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
3.
Drugs R D ; 19(2): 149-166, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30919310

ABSTRACT

BACKGROUND AND OBJECTIVES: Inhibition of Abelson (Abl) tyrosine kinase as a therapeutic target has been gaining attention in neurodegeneration. Post-mortem Alzheimer's and Parkinson's disease brains show that the levels of several other tyrosine kinases, including Discoidin Domain Receptors (DDR1/2) are elevated. Knockdown of these tyrosine kinases with shRNA reduces neurotoxic proteins, including alpha-synuclein, beta-amyloid and tau. METHODS: Direct profiling of the pharmacokinetics of multi-kinase inhibitors Nilotinib, Bosutinib, Bafetinib, Radotinib and LCB-03-0110 shows differential levels of brain penetration but the ability of these agents to reduce toxic proteins is independent of brain concentration and selectivity to Abl. RESULTS: Our results indicate that the effective dose of Nilotinib has the lowest plasma:brain ratio (1%) followed by Bosutinib and Radotinib (5%), Bafetinib (12%) and LCB-03-0110 (12%). However, similar doses of multi-kinase Abl/DDR inhibitor Nilotinib, DDR/Src inhibitor LCB-03-0110 and Abl/Src inhibitor Bosutinib were much more effective than the more selective Abl inhibitors Radotinib and Bafetinib. Taken together, these data suggest that a multi-kinase target that includes Abl and other tyrosine kinases (DDRs, and Src) may offer more advantages alleviating neurodegenerative pathologies than the absolute CNS drug concentration and selectivity to Abl. CONCLUSION: DDRs and Src are other potential co-targets with Abl in neurodegeneration.


Subject(s)
Alzheimer Disease/drug therapy , Parkinson Disease/drug therapy , Protein Kinase Inhibitors/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Discoidin Domain Receptors/antagonists & inhibitors , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Hippocampus/pathology , Humans , Male , Mesencephalon/pathology , Mice , Mice, Transgenic , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , RNA, Small Interfering/metabolism , Rats , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics , src-Family Kinases/metabolism
4.
Hum Mol Genet ; 28(4): 548-560, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30329047

ABSTRACT

Ubiquitin specific proteases (USPs) are de-ubiquitinases (DUBs) that control protein ubiquitination cycle. The role of DUBs is poorly understood in neurodegenerative diseases. We found that USP13 is overexpressed in post-mortem Parkinson's disease (PD) brains. We investigated whether changes in USP13 levels can affect two molecules, parkin and alpha-synuclein, that are implicated in PD pathogenesis. Parkin is an E3 ubiquitin ligase that is regulated by ubiquitination and targets certain proteins for degradation, and alpha-synuclein may be ubiquitinated and recycled in the normal brain. We found that USP13 independently regulates parkin and alpha-synuclein ubiquitination in models of alpha-synucleinopathies. USP13 shRNA knockdown increases alpha-synuclein ubiquitination and clearance, in a parkin-independent manner. Furthermore, USP13 overexpression counteracts the effects of a tyrosine kinase inhibitor, Nilotinib, while USP13 knockdown facilitates Nilotinib effects on alpha-synculein clearance, suggesting that alpha-synuclein ubiquitnation is important for its clearance. These studies provide novel evidence of USP13 effects on parkin and alpha-synuclein metabolism and suggest that USP13 is a potential therapeutic target in the alpha-synucleinopathies.


Subject(s)
Endopeptidases/genetics , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics , alpha-Synuclein/genetics , Autopsy , Brain/metabolism , Brain/pathology , Endopeptidases/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Parkinson Disease/pathology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
5.
J Neurol Sci ; 391: 90-99, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30103978

ABSTRACT

Tau hyperphosphorylation is a critical factor in neurodegenerative diseases, including dementia and Parkinsonism. Existing animal models of tauopathies express tau in neurons within the forebrain and do not often show tau accumulation in the brainstem and astrocytes. This study aims to understand the effects of differential regional expression of tau on neurotransmitter balance in the brain. To obtain an animal model that expresses tau in the brainstem, we bred hemizygous mice that express P301L tau (TauP301L) and detected hyper-phosphorylated tau (p-tau) predominantly in the hippocampus, cortex, brainstem and thalamus. We previously demonstrated that TauP301L mice [26] express tau under the control of a prion promoter in both neurons and astrocytes, reminiscent of human tauopathies. We treated TauP301L mice with tyrosine kinase inhibitors (TKIs) to determine the effects of tau clearance on neurotransmitter balance and astrocytic function. 13C/1H MRS reveals astrocytic dysfunction via reduced glial aspartate and impaired glutamate-glutamine cycle. An increase in glutamate and GABA and decrease in glutamine were observed in homozygous mice compared to hemizygous and control littermates. Daily treatment with TKIs, nilotinib or bosutinib led to p-tau clearance via autophagy and reversal of neurotransmitter imbalance. These data suggest that accumulation of p-tau in the brainstem does not alter dopamine metabolism but may trigger glutamate toxicity and astrocytic dysfunction in the TauP301L mouse. TKIs reverse tau effects via reversal of neurotransmitter imbalance.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Aniline Compounds/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Autophagy/drug effects , Autophagy/physiology , Brain/drug effects , Brain/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Mice, Transgenic , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , Proton Magnetic Resonance Spectroscopy , Pyrimidines/pharmacology , Quinolines/pharmacology , Tauopathies/drug therapy , Tauopathies/pathology , tau Proteins/genetics
7.
CNS Drugs ; 32(1): 1-11, 2018 01.
Article in English | MEDLINE | ID: mdl-29492779

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disease characterized by Lewy body pathology of which the primary constituent is aggregated misfolded alpha-synuclein protein. Currently, there are no clinical therapies for treatment of the underlying alpha-synuclein dysfunction and accumulation, and the standard of care for patients with Parkinson's disease focuses only on symptom management, creating an immense therapeutic gap that needs to be filled. Defects in autophagy have been strongly implicated in Parkinson's disease. Here, we review evidence from human, mouse, and cell culture studies to briefly explain these defects in autophagy in Parkinson's disease and the necessity for autophagy to be carefully and precisely tuned to maintain neuron survival. We summarize recent experimental agents for treating alpha-synuclein accumulation in α-synuclein Parkinson's disease and related synucleinopathies. Most of the efforts for developing experimental agents have focused on immunotherapeutic strategies, but we discuss why those efforts are misplaced. Finally, we emphasize why increasing autophagy flux for alpha-synuclein clearance is the most promising therapeutic strategy. Activating autophagy has been successful in preclinical models of Parkinson's disease and yields promising results in clinical trials.


Subject(s)
Antiparkinson Agents/pharmacology , Autophagy/drug effects , Parkinson Disease/drug therapy , Animals , Drug Development/methods , Drug Evaluation, Preclinical/methods , Humans , Mice , Parkinson Disease/physiopathology , alpha-Synuclein/metabolism
8.
J Neuroimmunol ; 311: 1-9, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28863860

ABSTRACT

The role of cell surface tyrosine kinase collagen-activated receptors known as discoidin domain receptors (DDRs) is unknown in neurodegenerative diseases. We detect up-regulation in DDRs level in post-mortem Alzheimer and Parkinson brains. Lentiviral shRNA knockdown of DDR1 and DDR2 reduces the levels of α-synuclein, tau, and ß-amyloid and prevents cell loss in vivo and in vitro. DDR1 and DDR2 knockdown alters brain immunity and significantly reduces the level of triggering receptor expressed on myeloid cells (TREM)-2 and microglia. These studies suggest that DDR1 and DDR2 inhibition is a potential target to clear neurotoxic proteins and reduce inflammation in neurodegeneration.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Discoidin Domain Receptors/metabolism , Parkinson Disease/complications , Parkinson Disease/pathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Case-Control Studies , Cell Line, Tumor , Cytokines/metabolism , Discoidin Domain Receptors/antagonists & inhibitors , Discoidin Domain Receptors/genetics , Encephalitis/drug therapy , Encephalitis/metabolism , Female , Hippocampus/metabolism , Humans , Male , Mice , Mice, Transgenic , Mutation/genetics , Neuroblastoma/pathology , Parkinson Disease/therapy , Peptide Fragments/metabolism , Rats , Up-Regulation/physiology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
9.
J Alzheimers Dis ; 60(2): 461-481, 2017.
Article in English | MEDLINE | ID: mdl-28869476

ABSTRACT

Hyperphosphorylation and aggregation of tau protein is a critical factor in many neurodegenerative diseases. These diseases are increasing in prevalence, and there are currently no cures. Previous work from our group and others has shown that tyrosine kinase inhibitors (TKIs) can stimulate autophagy, decrease pathological proteins, and improve symptoms in models of neurodegeneration. Here we examined the role of pazopanib in mouse models that express either human mutant P301L tau (TauP301L) or triple mutant amyloid precursor protein (3x-AßPP). The TauP301L mouse expresses P301L tau under the control of a prion promoter in both neurons and astrocytes, reminiscent of some human tauopathies. Pazopanib crosses the blood-brain barrier with no detectable peripheral off-side effects, and decreases p-tau in TauP301L mice. Pazopanib reaches a brain concentration sufficient for inhibition of several tyrosine kinases, including vascular endothelial growth factor receptors (VEGFRs). Further, pazopanib does not affect microglia but reduces astrocyte levels toward nontransgenic controls in TauP301L mice. Pazopanib does not alter amyloid beta levels or astrocytes in 3x-AßPP mice but modulates a number of inflammatory markers (IP-10, MIP-1α, MIP-1ß, and RANTES). These data suggest that pazopanib may be involved in p-tau clearance and modulation of astrocytic activity in models of tauopathies.


Subject(s)
Astrocytes/drug effects , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Tauopathies/drug therapy , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Animals , Astrocytes/pathology , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cognition/drug effects , Cognition/physiology , Collagen Type IV/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Glial Fibrillary Acidic Protein/metabolism , Indazoles , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Motor Activity/drug effects , Mutation/genetics , Neuroblastoma/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Silver Staining , Sulfonamides/pharmacology , Tauopathies/genetics , Transfection , Treatment Outcome , tau Proteins/genetics
10.
Expert Opin Investig Drugs ; 26(10): 1131-1136, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28817311

ABSTRACT

INTRODUCTION: BACE 1 is a protease that cleaves the transmembrane amyloid precursor protein and generates amyloid-ß peptides that accumulate in AD brains. No known mutations are identified in the gene encoding BACE1 in AD. However, enzyme levels are elevated in AD and a single residue mutation in amyloid precursor protein protects against protein cleavage by BACE1, suggesting BACE involvement in disease pathogenesis. Drugs that can inhibit BACE1 would theoretically prevent Aß accumulation and halt AD onset and progression. Areas covered: This review discusses clinical developments of BACE1 inhibitors and focuses on what is learned about these inhibitors as a potential treatment. Expert opinion: BACE1 inhibition as a therapeutic strategy to improve cognition in AD has been challening. Brain-penetrant BACE1 inhibitors have been developed and clinical trials are underway, both safety and efficacy are questionable. Several clinical trials suggest that BACE1 inhibition and other immunotherapies to reduce brain Aß are insufficient to improve cognition in AD. This may be due to the emphasis on the amyloid hypothesis despite big failures. We may have to seriously consider shifting attention to therapeutic strategies other than BACE1 inhibition or reduction of Aß alone and pay more attention to simultaneous clearance of tau and Aß.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Disease Progression , Drug Design , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Enzyme Inhibitors/pharmacology , Humans
11.
Mol Cell Neurosci ; 83: 46-54, 2017 09.
Article in English | MEDLINE | ID: mdl-28687523

ABSTRACT

The relationship between RNA-binding proteins, particularly TAR DNA binding protein 43 (TDP-43), and neurodegeneration is an important area of research. TDP-43 is involved in so many cellular processes that perturbation of protein homeostasis can lead to countless downstream effects. Understanding what leads to this disease-related protein imbalance and the resulting cellular and molecular effects will help to develop targets for disease intervention, whether it be prevention of protein accumulation, or addressing a secondary effect of protein accumulation. Here we review the current literature of TDP-43 and TDP-43 pathologies, the effects of TDP-43 overexpression and disruption of synaptic proteins through its binding of messenger RNA, leading to synaptic dysfunction. This review highlights some of the still-limited knowledge of the protein TDP-43 and how it can contribute to disease.


Subject(s)
DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/metabolism , Motor Neuron Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/genetics , Humans , Motor Neuron Disease/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Synaptic Transmission
12.
J Neurochem ; 139(4): 610-623, 2016 11.
Article in English | MEDLINE | ID: mdl-27507246

ABSTRACT

The trans-activating response of DNA/RNA-binding protein (TDP)-43 pathology is associated with many neurodegenerative diseases via unknown mechanisms. Here, we use a transgenic mouse model over-expressing human wild-type neuronal TDP-43 to study the effects of TDP-43 pathology on glutamate metabolism and synaptic function. We found that neuronal TDP-43 over-expression affects synaptic protein expression, including Synapsin I, and alters surrounding astrocytic function. TDP-43 over-expression is associated with an increase in glutamate and γ-amino butyric acid and reduction of glutamine and aspartate levels, indicating impairment of presynaptic terminal. TDP-43 also decreases tricarboxylic acid cycle metabolism and induces oxidative stress via lactate accumulation. Neuronal TDP-43 does not alter microglia activity or significantly changes systemic and brain inflammatory markers compared to control. We previously demonstrated that brain-penetrant tyrosine kinase inhibitors (TKIs), nilotinib and bosutinib, reduce TDP-43-induced cell death in transgenic mice. Here, we show that TKIs reverse the effects of TDP-43 on synaptic proteins, increase astrocytic function and restore glutamate and neurotransmitter balance in TDP-43 mice. Nilotinib, but not bosutinib, reverses mitochondrial impairment and oxidative metabolism. Taken together, these data suggest that TKIs can attenuate TDP-43 toxicity and improve synaptic and astrocytic function, independent of microglial or other inflammatory effects. In conclusion, our data demonstrate novel mechanisms of the effects of neuronal TDP-43 over-expression on synaptic protein expression and alteration of astrocytic function.


Subject(s)
Astrocytes/physiology , DNA-Binding Proteins/biosynthesis , Homeostasis/physiology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Synapsins/biosynthesis , Animals , Astrocytes/drug effects , Cell Line, Tumor , Female , Gene Expression , Homeostasis/drug effects , Humans , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Synapses/drug effects , Synapses/metabolism , Synapsins/genetics
13.
Neural Regen Res ; 11(12): 1910-1911, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28197178
14.
Neurodegener Dis ; 15(5): 259-70, 2015.
Article in English | MEDLINE | ID: mdl-26160424

ABSTRACT

Parkin biology has emerged as an exciting area of pharmaceutical development for several human diseases, including cancer and neurodegeneration. Parkin's role is multifaceted in human health and disease and its function affecting major cellular quality control mechanisms, including the ubiquitin-proteasome and autophagy-lysosome systems, is critical in the maintenance of cellular homeostasis. Loss of Parkin function due to aging, protein instability and gene mutations is manifest in a number of human diseases, contributing to the validation of this protein as a therapeutic target. Parkin activation to mobilize cellular quality control mechanisms and counteract dyshomeostasis is a highly desirable area for therapeutic development. The elucidation of Parkin's crystal structure and better understanding of possible posttranslational modifications (i.e. phosphorylation, ubiquitination, etc.) that regulate Parkin's enzymatic activity suggest that this protein is a therapeutic drug target in many human diseases. Here we review Parkin's role in health and disease and discuss the effects of self-ubiquitination and deubiquitination on Parkin activity. This review provides further evidence showing the longitudinal effects of Parkin deletion on mitochondrial function, oxidative stress and neurotransmitter balance in vivo using high-frequency (1)H/(13)C NMR spectroscopy.


Subject(s)
Autophagy , Brain Neoplasms/enzymology , Brain/enzymology , Mitochondria/enzymology , Neurodegenerative Diseases/enzymology , Ubiquitin-Protein Ligases/metabolism , Amino Acids/metabolism , Animals , Apoptosis , Homeostasis , Humans , Neurons/enzymology , Oxidative Stress , Parkinson Disease/enzymology , Parkinson Disease/genetics , Ubiquitin-Protein Ligases/genetics
16.
Hum Mol Genet ; 23(18): 4960-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24847002

ABSTRACT

The transactivation DNA-binding protein (TDP)-43 binds to thousands of mRNAs, but the functional outcomes of this binding remain largely unknown. TDP-43 binds to Park2 mRNA, which expresses the E3 ubiquitin ligase parkin. We previously demonstrated that parkin ubiquitinates TDP-43 and facilitates its translocation from the nucleus to the cytoplasm. Here we used brain penetrant tyrosine kinase inhibitors (TKIs), including nilotinib and bosutinib and showed that they reduce the level of nuclear TDP-43, abrogate its effects on neuronal loss, and reverse cognitive and motor decline. Nilotinib decreased soluble and insoluble TDP-43, while bosutinib did not affect the insoluble level. Parkin knockout mice exhibited high levels of endogenous TDP-43, while nilotinib and bosutinib did not alter TDP-43, underscoring an indispensable role for parkin in TDP-43 sub-cellular localization. These data demonstrate a novel functional relationship between parkin and TDP-43 and provide evidence that TKIs are potential therapeutic candidates for TDP-43 pathologies.


Subject(s)
Cognition/drug effects , DNA-Binding Proteins/metabolism , Motor Skills/drug effects , Neurons/metabolism , Protein Kinase Inhibitors/administration & dosage , Ubiquitin-Protein Ligases/metabolism , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Mice , Mice, Transgenic , Neurons/pathology , Nitriles/administration & dosage , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Quinolines/administration & dosage , Quinolines/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
J Mol Med (Berl) ; 92(4): 373-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337465

ABSTRACT

UNLABELLED: Alzheimer's disease (AD) is a neurodegenerative disorder associated with amyloid accumulation and autophagic changes. Parkin is an E3 ubiquitin ligase involved in proteasomal and autophagic clearance. We previously demonstrated decreased parkin solubility and interaction with the key autophagy enzyme beclin-1 in AD, but tyrosine kinase inhibition restored parkin-beclin-1 interaction. In the current studies, we determined the mechanisms of nilotinib-induced parkin-beclin-1 interaction, which leads to amyloid clearance. Nilotinib increased endogenous parkin levels and ubiquitination, which may enhance parkin recycling via the proteasome, leading to increased activity and interaction with beclin-1. Parkin solubility was decreased and autophagy was altered in amyloid expressing mice, suggesting that amyloid stress affects parkin stability, leading to failure of protein clearance via the lysosome. Isolation of autophagic vacuoles revealed amyloid and parkin accumulation in autophagic compartments but nilotinib decreased insoluble parkin levels and facilitated amyloid deposition into lysosomes in wild type, but not parkin(-/-) mice, further underscoring an essential role for endogenous parkin in amyloid clearance. These results suggest that nilotinib boosts the autophagic machinery, leading to increased level of endogenous parkin that undergoes ubiquitination and interacts with beclin-1 to facilitate amyloid clearance. These data suggest that nilotinib-mediated autophagic changes may trigger parkin response via increased protein levels, providing a therapeutic strategy to reduce Aß and Tau in AD. KEY MESSAGE: Parkin solubility (stability) is decreased in AD and APP transgenic mice. Nilotinib-induced autophagic changes increase endogenous parkin level. Increased parkin level leads to ubiquitination and proteasomal recycling. Re-cycling decreases insoluble parkin and increases parkin-beclin-1 interaction. Beclin-1-parkin interaction enhances amyloid clearance.


Subject(s)
Amyloid/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Alzheimer Disease/enzymology , Animals , Autophagy , Cell Line, Tumor , Enzyme Stability , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Rats
18.
Hum Mol Genet ; 23(5): 1365-75, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24154541

ABSTRACT

Intraneuronal accumulation of ß-amyloid (Aß)42 is one of the earliest pathological events in humans and in animal models of Alzheimer's disease (AD). Apolipoprotein E 4 (APOE4) is the major identified genetic risk factor for late-onset AD, with Aß deposition beginning earlier in apoE4-positive subjects. To directly determine the effects of APOE genotype on intraneuronal accumulation of Aß1-42 at the onset of AD pathogenesis, we introduced lentiviral Aß1-42 into the cortex of APOE targeted replacement (TR) mice at the age of 8-9 months. We demonstrated a significant isoform-dependent effect of human APOE, with dramatically enhanced intracellular Aß1-42 deposits in the cerebral cortex of APOE4-TR mice 2 weeks after injection. Double-immunofluorescent staining showed that intracellular accumulation of lentiviral Aß1-42 was mainly present in neurons, localized to late endosomes/lysosomes. This intraneuronal accumulation of Aß1-42 correlated with increased tau phosphorylation and cell death in the ipsilateral cortex around the injection site. Aß1-42 was also observed in microglia, but not in astrocytes. Quantitative analysis revealed more neurons with Aß1-42 while less microglia with Aß1-42 nearest to the injection site of Aß1-42 lentivirus in APOE4-TR mice. Finally, apoE was present in neurons of the ipsilateral cortex of APOE-TR mice at 2 weeks after lentivirus injection, in addition to astrocytes and microglia in both the ipsilateral and contralateral cerebral cortex. Taken together, these results demonstrate that apoE4 tips the balance of the glial and neuronal Aß toward the intraneuronal accumulation of Aß.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Genetic Vectors/genetics , Genotype , Lentivirus/genetics , Neurons/metabolism , Transduction, Genetic , Animals , Apolipoprotein E4/metabolism , Cerebral Cortex/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Humans , Intracellular Space/metabolism , Mice , Mice, Transgenic , Microglia/metabolism , Microinjections , Protein Binding , Protein Transport
19.
J Clin Cell Immunol ; 5: 259, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25635231

ABSTRACT

OBJECTIVES: Neuro-inflammation is common in α-Synucleinopathies and Tauopathies; and evidence suggests a link between the tyrosine kinase Abl and neurodegeneration. Abl upregulates α-Synuclein and promotes Tau hyper-phosphorylation (p-Tau), while Abl inhibitors facilitate autophagic clearance. METHODS: A model of α-Synucleinopathy harboring human mutant A53T α-Synuclein and exhibits concomitant increase in murine p-Tau was used to determine the immunological response to Abl inhibition. RESULTS: Age-dependent alterations of brain immunity, including loss of IL-10 and decreased levels of IL-2 and IL-3 were observed in old A53T mice. Brain CCL2 and CCL5 were decreased, but CX3CL1 remained constantly elevated. Young A53T mice exhibited differential systemic and central immune profiles in parallel with increased blood markers of adaptive immunity, suggesting an early systemic immune response. Tyrosine kinase inhibitors (TKIs), including nilotinib and bosutinib reduced brain and peripheral α-Synuclein and p-Tau and modulated blood immunological responses. TKIs did not affect brain IL-10, but they changed the levels of all measured blood immune markers, except CX3CL1. TKIs altered microglia morphology and reduced the number of astrocyte and dendritic cells, suggesting beneficial regulation of microglia. CONCLUSIONS: These data indicate that tyrosine kinase inhibition affects neuro-inflammation via early changes of the peripheral immune profile, leading to modulation of the neuro-immune response to α-Synuclein and p-Tau.

20.
J Neurochem ; 129(2): 350-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24298989

ABSTRACT

The E3 ubiquitin ligase Parkin plays a central role in the pathogenesis of many neurodegenerative diseases. Parkin promotes specific ubiquitination and affects the localization of transactivation response DNA-binding protein 43 (TDP-43), which controls the translation of thousands of mRNAs. Here we tested the effects of lentiviral Parkin and TDP-43 expression on amino acid metabolism in the rat motor cortex using high frequency ¹³C NMR spectroscopy. TDP-43 expression increased glutamate levels, decreased the levels of other amino acids, including glutamine, aspartate, leucine and isoleucine, and impaired mitochondrial tricarboxylic acid cycle. TDP-43 induced lactate accumulation and altered the balance between excitatory (glutamate) and inhibitory (GABA) neurotransmitters. Parkin restored amino acid levels, neurotransmitter balance and tricarboxylic acid cycle metabolism, rescuing neurons from TDP-43-induced apoptotic death. Furthermore, TDP-43 expression led to an increase in 4E-BP levels, perhaps altering translational control and deregulating amino acid synthesis; while Parkin reversed the effects of TDP-43 on the 4E-BP signaling pathway. Taken together, these data suggest that Parkin may affect TDP-43 localization and mitigate its effects on 4E-BP signaling and loss of amino acid homeostasis.


Subject(s)
Amino Acids/metabolism , Cell Death/drug effects , TDP-43 Proteinopathies/drug therapy , Ubiquitin-Protein Ligases/pharmacology , Animals , Blotting, Western , Carrier Proteins/metabolism , Caspase 3/metabolism , Citric Acid Cycle/drug effects , Fluorometry , Genetic Vectors , Homeostasis/drug effects , Homeostasis/physiology , Intracellular Signaling Peptides and Proteins , Lentivirus/genetics , Magnetic Resonance Spectroscopy , Male , Motor Cortex/drug effects , Motor Cortex/metabolism , Neurotransmitter Agents/metabolism , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , TDP-43 Proteinopathies/pathology , TOR Serine-Threonine Kinases/metabolism , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...