Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
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
3.
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
4.
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
5.
Hum Mol Genet ; 22(16): 3315-28, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23666528

ABSTRACT

Parkinson's disease is a movement disorder characterized by death of dopaminergic substantia nigra (SN) neurons and brain accumulation of α-synuclein. The tyrosine kinase Abl is activated in neurodegeneration. Here, we show that lentiviral expression of α-synuclein in the mouse SN leads to Abl activation (phosphorylation) and lentiviral Abl expression increases α-synuclein levels, in agreement with Abl activation in PD brains. Administration of the tyrosine kinase inhibitor nilotinib decreases Abl activity and ameliorates autophagic clearance of α-synuclein in transgenic and lentiviral gene transfer models. Subcellular fractionation shows accumulation of α-synuclein and hyper-phosphorylated Tau (p-Tau) in autophagic vacuoles in α-synuclein expressing brains, but nilotinib enhances protein deposition into the lysosomes. Nilotinib is used for adult leukemia treatment and it enters the brain within US Food and Drug Administration approved doses, leading to autophagic degradation of α-synuclein, protection of SN neurons and amelioration of motor performance. These data suggest that nilotinib may be a therapeutic strategy to degrade α-synuclein in PD and other α-synucleinopathies.


Subject(s)
Autophagy , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Parkinsonian Disorders/metabolism , Pyrimidines/pharmacology , alpha-Synuclein/metabolism , Adult , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/physiology , Genes, abl , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/genetics , Point Mutation , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Substantia Nigra/metabolism , alpha-Synuclein/blood
6.
J Biol Chem ; 288(6): 4103-15, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23258539

ABSTRACT

The importance of E3 ubiquitin ligases, involved in the degradation of misfolded proteins or promotion of protein-protein interaction, is increasingly recognized in neurodegeneration. TDP-43 is a predominantly nuclear protein, which regulates the transcription of thousands of genes and binds to mRNA of the E3 ubiquitin ligase Parkin to regulate its expression. Wild type and mutated TDP-43 are detected in ubiquitinated forms within the cytosol in several neurodegenerative diseases. We elucidated the mechanisms of TDP-43 interaction with Parkin using transgenic A315T mutant TDP-43 (TDP43-Tg) mice, lentiviral wild type TDP-43, and Parkin gene transfer rat models. TDP-43 expression increased Parkin mRNA and protein levels. Lentiviral TDP-43 increased the levels of nuclear and cytosolic protein, whereas Parkin co-expression mediated Lys-48 and Lys-63-linked ubiquitin to TDP-43 and led to cytosolic co-localization of Parkin with ubiquitinated TDP-43. Parkin and TDP-43 formed a multiprotein complex with HDAC6, perhaps to mediate TDP-43 translocation. In conclusion, Parkin ubiquitinates TDP-43 and facilitates its cytosolic accumulation through a multiprotein complex with HDAC6.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Nerve Tissue Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Substitution , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Mice , Mice, Transgenic , Mutation, Missense , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Transport/genetics , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/genetics
7.
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
8.
J Biol Chem ; 287(17): 14192-200, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22378781

ABSTRACT

Phosphorylation of signal transducer and activator of transcription 3 (STAT3) on a single tyrosine residue in response to growth factors, cytokines, interferons, and oncogenes activates its dimerization, translocation to the nucleus, binding to the interferon γ (gamma)-activated sequence (GAS) DNA-binding site and activation of transcription of target genes. STAT3 is constitutively phosphorylated in various cancers and drives gene expression from GAS-containing promoters to promote tumorigenesis. Recently, roles for unphosphorylated STAT3 (U-STAT3) have been described in response to cytokine stimulation, in cancers, and in maintenance of heterochromatin stability. However, the mechanisms underlying U-STAT3 binding to DNA has not been fully investigated. Here, we explore STAT3-DNA interactions by atomic force microscopy (AFM) imaging. We observed that U-STAT3 molecules bind to the GAS DNA-binding site as dimers and monomers. In addition, we observed that U-STAT3 binds to AT-rich DNA sequence sites and recognizes specific DNA structures, such as 4-way junctions and DNA nodes, within negatively supercoiled plasmid DNA. These structures are important for chromatin organization and our data suggest a role for U-STAT3 as a chromatin/genome organizer. Unexpectedly, we found that a C-terminal truncated 67.5-kDa STAT3 isoform recognizes single-stranded spacers within cruciform structures that also have a role in chromatin organization and gene expression. This isoform appears to be abundant in the nuclei of cancer cells and, therefore, may have a role in regulation of gene expression. Taken together, our data highlight novel mechanisms by which U-STAT3 binds to DNA and supports U-STAT3 function as a transcriptional activator and a chromatin/genomic organizer.


Subject(s)
Chromatin/chemistry , DNA/chemistry , STAT3 Transcription Factor/metabolism , Binding Sites , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Kinetics , Male , Microscopy, Atomic Force/methods , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Subcellular Fractions
9.
J Neurochem ; 118(4): 521-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21166806

ABSTRACT

Methamphetamine (MA) is a highly addictive psychostimulant that, used in excess, may be neurotoxic. Although the mechanisms that underlie its addictive potential are not completely understood, in animal models matrix metalloproteinase (MMP) inhibitors can reduce behavioral correlates of addiction. In addition, evidence from genome-wide association studies suggests that polymorphisms in synaptic cell-adhesion molecules (CAMs), known MMP substrates, are linked to addictive potential in humans. In the present study, we examined the ability of MA to stimulate cleavage of intercellular adhesion molecule-5 (ICAM-5), a synaptic CAM expressed on dendritic spines in the telencephalon. Previous studies have shown that shedding of ICAM-5 is associated with maturation of dendritic spines, and that MMP-dependent shedding occurs with long term potentiation. Herein, we show that MA stimulates ectodomain cleavage of ICAM-5 in vitro, and that this is abrogated by a broad spectrum MMP inhibitor. We also show that an acute dose of MA, administered in vivo, is associated with cleavage of ICAM-5 in murine hippocampus and striatum. This occurs within 6 h and is accompanied by an increase in MMP-9 protein. In related experiments, we examined the potential consequences of ICAM-5 shedding. We demonstrate that the ICAM-5 ectodomain can interact with ß(1) integrins, and that it can stimulate ß(1) integrin-dependent phosphorylation of cofilin, an event that has previously been linked to MMP-dependent spine maturation. Together these data support an emerging appreciation of MMPs as effectors of synaptic plasticity and suggest a mechanism by which MA may influence the same.


Subject(s)
Cell Adhesion Molecules/metabolism , Central Nervous System Stimulants/toxicity , Methamphetamine/toxicity , Nerve Tissue Proteins/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Densitometry , Dipeptides/pharmacology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Immunoprecipitation , Integrin beta1/biosynthesis , Male , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/biosynthesis , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Protease Inhibitors/pharmacology , Rats , Spine/growth & development , Spine/metabolism , Transfection
10.
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
11.
Mol Neurodegener ; 9: 46, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25384392

ABSTRACT

BACKGROUND: Tau is an axonal protein that binds to and regulates microtubule function. Hyper-phosphorylation of Tau reduces its binding to microtubules and it is associated with ß-amyloid deposition in Alzheimer's disease. Paradoxically, Tau reduction may prevent ß-amyloid pathology, raising the possibility that Tau mediates intracellular Aß clearance. The current studies investigated the role of Tau in autophagic and proteasomal intracellular Aß1-42 clearance and the subsequent effect on plaque deposition. RESULTS: Tau deletion impaired Aß clearance via autophagy, but not the proteasome, while introduction of wild type human Tau into Tau-/- mice partially restored autophagic clearance of Aß1-42, suggesting that exogenous Tau expression can support autophagic Aß1-42 clearance. Tau deletion impaired autophagic flux and resulted in Aß1-42 accumulation in pre-lysosomal autophagic vacuoles, affecting Aß1-42 deposition into the lysosome. This autophagic defect was associated with decreased intracellular Aß1-42 and increased plaque load in Tau-/- mice, which displayed less cell death. Nilotinib, an Abl tyrosine kinase inhibitor that promotes autophagic clearance mechanisms, reduced Aß1-42 only when exogenous human Tau was expressed in Tau-/- mice. CONCLUSIONS: These studies demonstrate that Tau deletion affects intracellular Aß1-42 clearance, leading to extracellular plaque.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Extracellular Space/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plaque, Amyloid/metabolism , Proteasome Endopeptidase Complex/metabolism
12.
Exp Neurol ; 251: 127-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23333589

ABSTRACT

Tau hyper-phosphorylation (p-Tau) and neuro-inflammation are hallmarks of neurodegeneration. Previous findings suggest that microglial activation via CX3CL1 promotes p-Tau. We examined inflammation and autophagic p-Tau clearance in lentiviral Tau and mutant P301L expressing rats and used lentiviral Aß1-42 to induce p-Tau. Lentiviral Tau or P301L expression significantly increased caspase-3 activity and TNF-α, but CX3CL1 was significantly higher in animals expressing Tau compared to P301L. Lentiviral Aß1-42 induced p-Tau 4 weeks post-injection, and increased caspase-3 activation (8-fold) and TNF-α levels. Increased levels of ADAM-10/17 were also detected with p-Tau. IL-6 levels were increased but CX3CL1 did not change in the absence of p-Tau (2 weeks); however, p-Tau reversed these effects, which were associated with increased microglial activity. We observed changes in autophagic markers, including accumulation of autophagic vacuoles (AVs) and p-Tau accumulation in autophagosomes but not lysosomes, suggesting alteration of autophagy. Taken together, microglial activation may promote p-Tau independent of total Tau levels via CX3CL1 signaling, which seems to depend on interaction with inflammatory markers, mainly IL-6. The simultaneous change in autophagy and CX3CL1 signaling suggests communication between microglia and neurons, raising the possibility that accumulation of intraneuronal amyloid, due to lack of autophagic clearance, may lead microglia activation to promote p-Tau as a tag for phagocytic degradation.


Subject(s)
Autophagy/physiology , Chemokine CX3CL1/metabolism , Gene Expression Regulation/physiology , Signal Transduction/physiology , tau Proteins/genetics , tau Proteins/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Autophagy/genetics , Caspase 3/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/genetics , Gene Transfer Techniques , Glial Fibrillary Acidic Protein/metabolism , Lentivirus/genetics , Male , Microglia/metabolism , Microglia/ultrastructure , Mutation/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure
13.
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
14.
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.

15.
Autophagy ; 9(8): 1249-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23787811

ABSTRACT

The effects of ABL1/ABL inhibition on clearance of SNCA/α-synuclein were evaluated in animal models of α-synucleinopathies. Parkinson disease (PD) is a movement disorder characterized by death of dopaminergic substantia nigra (SN) neurons and brain accumulation of SNCA. The tyrosine kinase ABL1 is activated in several neurodegenerative diseases. An increase in ABL1 activity is detected in human postmortem PD brains. Lentiviral expression of SNCA in the mouse SN activates ABL1 via phosphorylation, while lentiviral Abl expression increases SNCA levels. Administration of the brain-penetrant tyrosine kinase inhibitor Nilotinib decreases Abl activity and facilitates autophagic clearance of SNCA in transgenic and lentiviral gene transfer models. Subcellular fractionation demonstrates accumulation of SNCA and hyperphosphorylated MAPT/Tau (p-MAPT) in autophagic vacuoles in SNCA-expressing brains, while Nilotinib treatment leads to protein deposition into the lysosomes, suggesting enhanced autophagic clearance. These data suggest that Nilotinib may be a therapeutic strategy to degrade SNCA in PD and other α-synucleinopathies.


Subject(s)
Autophagy , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Parkinsonian Disorders/metabolism , Pyrimidines/pharmacology , alpha-Synuclein/metabolism , Animals , Humans , Male
16.
EMBO Mol Med ; 5(8): 1247-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23737459

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are effective therapies for leukaemia. Alzheimer is a neurodegenerative disease characterized by accumulation of ß-amyloid (plaques) and hyper-phosphorylated Tau (tangles). Here we show that AD animals have high levels of insoluble parkin and decreased parkin-Beclin-1 interaction, while peripheral administration of TKIs, including Nilotinib and Bosutinib, increases soluble parkin leading to amyloid clearance and cognitive improvement. Blocking Beclin-1 expression with shRNA or parkin deletion prevents tyrosine kinase (TK) inhibition-induced amyloid clearance, suggesting that functional parkin-Beclin-1 interaction mediates amyloid degradation. Isolation of autophagic vacuoles (AVs) in AD mouse brain shows accumulation of parkin and amyloid, consistent with previous results in AD brains, while Bosutinib and Nilotinib increase parkin-Beclin-1 interaction and result in protein deposition in the lysosome. These data suggest that decreased parkin solubility impedes parkin-Beclin-1 interaction and amyloid clearance. We identified two FDA-approved anti-cancer drugs as potential treatment for AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Apoptosis Regulatory Proteins/metabolism , Cognition/drug effects , Membrane Proteins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Beclin-1 , Brain/metabolism , Cognition Disorders/metabolism , Humans , Mice , Neurodegenerative Diseases/metabolism , Nitriles/pharmacology , Phosphorylation , Pyrimidines/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/metabolism , tau Proteins/metabolism
17.
PLoS One ; 8(12): e83914, 2013.
Article in English | MEDLINE | ID: mdl-24386307

ABSTRACT

Parkinson's disease (PD) is a movement disorder associated with genetic and age related causes. Although autosomal recessive early onset PD linked to parkin mutations does not exhibit α-Synuclein accumulation, while autosomal dominant and sporadic PD manifest with α-Synuclein inclusions, loss of dopaminergic substantia nigra neurons is a common denominator in PD. Here we show that decreased parkin ubiquitination and loss of parkin stability impair interaction with Beclin-1 and alter α-Synuclein degradation, leading to death of dopaminergic neurons. Tyrosine kinase inhibition increases parkin ubiquitination and interaction with Beclin-1, promoting autophagic α-Synuclein clearance and nigral neuron survival. However, loss of parkin via deletion increases α-Synuclein in the blood compared to the brain, suggesting that functional parkin prevents α-Synuclein release into the blood. These studies demonstrate that parkin ubiquitination affects its protein stability and E3 ligase activity, possibly leading to α-Synuclein sequestration and subsequent clearance.


Subject(s)
Autophagy , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , alpha-Synuclein/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Humans , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Kinase Inhibitors/pharmacology , Rats , Ubiquitination/drug effects , alpha-Synuclein/blood
18.
J Alzheimers Dis ; 33(1): 231-47, 2013.
Article in English | MEDLINE | ID: mdl-22954671

ABSTRACT

Alzheimer's disease (AD) is an aging disorder characterized by amyloid-ß (Aß) accumulation in extracellular plaques and formation of intracellular tangles containing hyperphosphorylated tau (p-Tau). Autophagic defects, leading to accumulation of autophagosomes, are recognized in AD. Parkin is an E3 ubiquitin ligase involved in degradation of proteins via autophagy and the proteasome. We investigated the role of parkin in postmortem brain tissues from 21 AD patients and 15 control subjects. We detected decreased parkin solubility in AD cortex and parkin co-localization with intraneuronal Aß(1-42) in the hippocampus and cortex of AD patients. Parkin accumulation with intraneuronal Aß and p-Tau was detected in autophagosomes in AD brains. To determine the role of parkin in Aß clearance, we generated gene transfer animals expressing lentiviral Aß(1-42)with and without parkin and examined autophagic mechanisms. Lentiviral expression of Aß(1-42) led to p-Tau accumulation and induced autophagic defects, leading to accumulation of autophagic vacuoles. However, co-expression of wild type parkin facilitated autophagic clearance and promoted deposition of Aß(1-42) and p-Tau into the lysosome. Taken together, these data suggest that Aß(1-42) alters normal autophagy and parkin enhances autophagic clearance. In conclusion, decreased parkin solubility may lead to co-localization with intraneuronal Aß(1-42) and compromise the cell autophagic clearance ability. Parkin may clear autophagic defects via autophagosome degradation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Autophagy/physiology , Neurons/metabolism , Peptide Fragments/metabolism , Ubiquitin-Protein Ligases/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Female , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Male , Middle Aged , Neurons/chemistry , Neurons/pathology , Rats , Rats, Sprague-Dawley , Solubility , tau Proteins/metabolism
19.
PLoS One ; 8(7): e69136, 2013.
Article in English | MEDLINE | ID: mdl-23844251

ABSTRACT

Matrix metalloproteinases (MMPs) are zinc dependent endopeptidases that can be released from neurons in an activity dependent manner to play a role in varied forms of learning and memory. MMP inhibitors impair hippocampal long term potentiation (LTP), spatial memory, and behavioral correlates of drug addiction. Since MMPs are thought to influence LTP through a ß1 integrin dependent mechanism, it has been suggested that these enzymes cleave specific substrates to generate integrin binding ligands. In previously published work, we have shown that neuronal activity stimulates rapid MMP dependent shedding of intercellular adhesion molecule-5 (ICAM-5), a synaptic adhesion molecule expressed on dendrites of the telencephalon. We have also shown that the ICAM-5 ectodomain can interact with ß1 integrins to stimulate integrin dependent phosphorylation of cofilin, an event that occurs with dendritic spine maturation and LTP. In the current study, we investigate the potential for the ICAM-5 ectodomain to stimulate changes in α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor (AMPAR) dependent glutamatergic transmission. Single cell recordings show that the ICAM-5 ectodomain stimulates an increase in the frequency, but not the amplitude, of AMPA mini excitatory post synaptic currents (mEPSCs). With biotinylation and precipitation assays, we also show that the ICAM-5 ectodomain stimulates an increase in membrane levels of GluA1, but not GluA2, AMPAR subunits. In addition, we observe an ICAM-5 associated increase in GluA1 phosphorylation at serine 845. Concomitantly, ICAM-5 affects an increase in GluA1 surface staining along dendrites without affecting an increase in dendritic spine number. Together these data are consistent with the possibility that soluble ICAM-5 increases glutamatergic transmission and that post-synaptic changes, including increased phosphorylation and dendritic insertion of GluA1, could contribute. We suggest that future studies are warranted to determine whether ICAM-5 is one of a select group of synaptic CAMs whose shedding contributes to MMP dependent effects on learning and memory.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendrites/metabolism , Excitatory Postsynaptic Potentials/physiology , Nerve Tissue Proteins/metabolism , Receptors, AMPA/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules/pharmacology , Cells, Cultured , Dendrites/drug effects , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Immunohistochemistry , Matrix Metalloproteinases/metabolism , Models, Biological , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Phosphorylation/drug effects , Proteolysis , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Serine/metabolism , Solubility
20.
Int J Alzheimers Dis ; 2012: 345472, 2012.
Article in English | MEDLINE | ID: mdl-22919540

ABSTRACT

The role of inflammation in neurodegenerative diseases has been widely demonstrated. Intraneuronal protein accumulation may regulate microglial activity via the fractalkine (CX3CL1) signaling pathway that provides a mechanism through which neurons communicate with microglia. CX3CL1 levels fluctuate in different stages of neurodegenerative diseases and in various animal models, warranting further investigation of the mechanisms underlying microglial response to pathogenic proteins, including Tau, ß-amyloid (Aß), and α-synuclein. The temporal relationship between microglial activity and localization of pathogenic proteins (intra- versus extracellular) likely determines whether neuroinflammation mitigates or exacerbates disease progression. Evidence in transgenic models suggests a beneficial effect of microglial activity on clearance of proteins like Aß and a detrimental effect on Tau modification, but the role of CX3CL1 signaling in α-synucleinopathies is less clear. Here we review the nature of fractalkine-mediated neuronmicroglia interaction, which has significant implications for the efficacy of anti-inflammatory treatments during different stages of neurodegenerative pathology. Specifically, it is likely that anti-inflammatory treatment in early stages of disease during intraneuronal accumulation of proteins could be beneficial, while anti-inflammatory treatment in later stages when proteins are secreted to the extracellular space could exacerbate disease progression.

SELECTION OF CITATIONS
SEARCH DETAIL