Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Brain Commun ; 4(2): fcac042, 2022.
Article in English | MEDLINE | ID: mdl-35282165

ABSTRACT

The accumulation of α-synuclein inclusions in vulnerable neuronal populations pathologically defines Lewy body diseases including Parkinson's disease. Recent pre-clinical studies suggest poly(ADP-ribose) polymerase-1 activation and the subsequent generation of poly(ADP-ribose) polymer represent key steps in the formation of toxic α-synuclein aggregates and neurodegeneration. Several studies suggest that the inhibition of poly(ADP-ribose) polymerase-1 activity via the poly(ADP-ribose) polymerase-1/2 small molecule inhibitor ABT-888 (Veliparib), a drug in clinical trials for different cancers, may prevent or ameliorate α-synuclein fibril-induced aggregation, inclusion formation and dopaminergic neurodegeneration. Herein, we evaluated the effects of poly(ADP-ribose) polymer on α-synuclein fibrillization in vitro, the effects of ABT-888 on the formation of fibril-seeded α-synuclein inclusions in primary mouse cortical neurons and the effects of an in-diet ABT-888 dosage regimen with the intracranial injection of α-synuclein fibrils into the mouse dorsal striatum. We found that poly(ADP-ribose) polymer minimally but significantly increased the rate of spontaneously formed α-synuclein fibrils in vitro. Machine-learning algorithms that quantitatively assessed α-synuclein inclusion counts in neurons, both in primary cultures and in the brains of fibril-injected mice, did not reveal differences between ABT-888- and vehicle-treated groups. The in-diet administered ABT-888 molecule demonstrated outstanding brain penetration in mice; however, dopaminergic cell loss in the substantia nigra caused by α-synuclein fibril injections in the striatum was similar between ABT-888- and vehicle-treated groups. α-Synuclein fibril-induced loss of dopaminergic fibres in the dorsal striatum was also similar between ABT-888- and vehicle-treated groups. We conclude that additional pre-clinical evaluation of ABT-888 may be warranted to justify further exploration of ABT-888 for disease modification in Lewy body diseases.

2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443159

ABSTRACT

Inositol-1,4,5-triphosphate (IP3) kinase B (ITPKB) is a ubiquitously expressed lipid kinase that inactivates IP3, a secondary messenger that stimulates calcium release from the endoplasmic reticulum (ER). Genome-wide association studies have identified common variants in the ITPKB gene locus associated with reduced risk of sporadic Parkinson's disease (PD). Here, we investigate whether ITPKB activity or expression level impacts PD phenotypes in cellular and animal models. In primary neurons, knockdown or pharmacological inhibition of ITPKB increased levels of phosphorylated, insoluble α-synuclein pathology following treatment with α-synuclein preformed fibrils (PFFs). Conversely, ITPKB overexpression reduced PFF-induced α-synuclein aggregation. We also demonstrate that ITPKB inhibition or knockdown increases intracellular calcium levels in neurons, leading to an accumulation of calcium in mitochondria that increases respiration and inhibits the initiation of autophagy, suggesting that ITPKB regulates α-synuclein pathology by inhibiting ER-to-mitochondria calcium transport. Furthermore, the effects of ITPKB on mitochondrial calcium and respiration were prevented by pretreatment with pharmacological inhibitors of the mitochondrial calcium uniporter complex, which was also sufficient to reduce α-synuclein pathology in PFF-treated neurons. Taken together, these results identify ITPKB as a negative regulator of α-synuclein aggregation and highlight modulation of ER-to-mitochondria calcium flux as a therapeutic strategy for the treatment of sporadic PD.


Subject(s)
Calcium/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , alpha-Synuclein/metabolism , Animals , Autophagy/genetics , Endoplasmic Reticulum/metabolism , Genome-Wide Association Study/methods , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation/genetics , Signal Transduction/genetics , Synucleinopathies/genetics , Synucleinopathies/metabolism
4.
Cell Syst ; 10(4): 333-350.e14, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32325033

ABSTRACT

Connectivity webs mediate the unique biology of the mammalian brain. Yet, while cell circuit maps are increasingly available, knowledge of their underlying molecular networks remains limited. Here, we applied multi-dimensional biochemical fractionation with mass spectrometry and machine learning to survey endogenous macromolecules across the adult mouse brain. We defined a global "interactome" comprising over one thousand multi-protein complexes. These include hundreds of brain-selective assemblies that have distinct physical and functional attributes, show regional and cell-type specificity, and have links to core neurological processes and disorders. Using reciprocal pull-downs and a transgenic model, we validated a putative 28-member RNA-binding protein complex associated with amyotrophic lateral sclerosis, suggesting a coordinated function in alternative splicing in disease progression. This brain interaction map (BraInMap) resource facilitates mechanistic exploration of the unique molecular machinery driving core cellular processes of the central nervous system. It is publicly available and can be explored here https://www.bu.edu/dbin/cnsb/mousebrain/.


Subject(s)
Brain Mapping/methods , Brain/metabolism , Connectome/methods , Amyotrophic Lateral Sclerosis/metabolism , Animals , DNA-Binding Proteins/genetics , Machine Learning , Mammals/physiology , Mass Spectrometry/methods , Mice , Mutation/genetics
5.
J Neurosci ; 40(1): 107-130, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31704785

ABSTRACT

Individual variation in the addiction liability of amphetamines has a heritable genetic component. We previously identified Hnrnph1 (heterogeneous nuclear ribonucleoprotein H1) as a quantitative trait gene underlying decreased methamphetamine-induced locomotor activity in mice. Here, we showed that mice (both females and males) with a heterozygous mutation in the first coding exon of Hnrnph1 (H1+/-) showed reduced methamphetamine reinforcement and intake and dose-dependent changes in methamphetamine reward as measured via conditioned place preference. Furthermore, H1+/- mice showed a robust decrease in methamphetamine-induced dopamine release in the NAc with no change in baseline extracellular dopamine, striatal whole-tissue dopamine, dopamine transporter protein, dopamine uptake, or striatal methamphetamine and amphetamine metabolite levels. Immunohistochemical and immunoblot staining of midbrain dopaminergic neurons and their forebrain projections for TH did not reveal any major changes in staining intensity, cell number, or forebrain puncta counts. Surprisingly, there was a twofold increase in hnRNP H protein in the striatal synaptosome of H1+/- mice with no change in whole-tissue levels. To gain insight into the mechanisms linking increased synaptic hnRNP H with decreased methamphetamine-induced dopamine release and behaviors, synaptosomal proteomic analysis identified an increased baseline abundance of several mitochondrial complex I and V proteins that rapidly decreased at 30 min after methamphetamine administration in H1+/- mice. In contrast, the much lower level of basal synaptosomal mitochondrial proteins in WT mice showed a rapid increase. We conclude that H1+/- decreases methamphetamine-induced dopamine release, reward, and reinforcement and induces dynamic changes in basal and methamphetamine-induced synaptic mitochondrial function.SIGNIFICANCE STATEMENT Methamphetamine dependence is a significant public health concern with no FDA-approved treatment. We discovered a role for the RNA binding protein hnRNP H in methamphetamine reward and reinforcement. Hnrnph1 mutation also blunted methamphetamine-induced dopamine release in the NAc, a key neurochemical event contributing to methamphetamine addiction liability. Finally, Hnrnph1 mutants showed a marked increase in basal level of synaptosomal hnRNP H and mitochondrial proteins that decreased in response to methamphetamine, whereas WT mice showed a methamphetamine-induced increase in synaptosomal mitochondrial proteins. Thus, we identified a potential role for hnRNP H in basal and dynamic mitochondrial function that informs methamphetamine-induced cellular adaptations associated with reduced addiction liability.


Subject(s)
Dopamine/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Methamphetamine/pharmacology , Mitochondria/drug effects , Reinforcement, Psychology , Reward , Synaptosomes/metabolism , Animals , Anxiety/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopaminergic Neurons/drug effects , Exons/genetics , Exploratory Behavior/drug effects , Female , Heterozygote , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Methamphetamine/toxicity , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mutation , Reflex, Startle/drug effects , Rotarod Performance Test , Substance-Related Disorders/physiopathology
6.
Cell Rep ; 29(13): 4377-4388.e4, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875547

ABSTRACT

Pathological aggregation of RNA binding proteins (RBPs) is associated with dysregulation of RNA splicing in PS19 P301S tau transgenic mice and in Alzheimer's disease brain tissues. The dysregulated splicing particularly affects genes involved in synaptic transmission. The effects of neuroprotective TIA1 reduction on PS19 mice are also examined. TIA1 reduction reduces disease-linked alternative splicing events for the major synaptic mRNA transcripts examined, suggesting that normalization of RBP functions is associated with the neuroprotection. Use of the NetDecoder informatics algorithm identifies key upstream biological targets, including MYC and EGFR, underlying the transcriptional and splicing changes in the protected compared to tauopathy mice. Pharmacological inhibition of MYC and EGFR activity in neuronal cultures tau recapitulates the neuroprotective effects of TIA1 reduction. These results demonstrate that dysfunction of RBPs and RNA splicing processes are major elements of the pathophysiology of tauopathies, as well as potential therapeutic targets for tauopathies.


Subject(s)
RNA Splicing/genetics , Tauopathies/genetics , Alzheimer Disease/genetics , Animals , Brain/metabolism , Down-Regulation/genetics , ErbB Receptors/metabolism , Female , Heterozygote , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sex Characteristics , Spliceosomes/metabolism , Synapses/metabolism , T-Cell Intracellular Antigen-1/genetics , T-Cell Intracellular Antigen-1/metabolism
7.
Acta Neuropathol Commun ; 6(1): 71, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068389

ABSTRACT

The development of insoluble, intracellular neurofibrillary tangles composed of the microtubule-associated protein tau is a defining feature of tauopathies, including Alzheimer's disease (AD). Accumulating evidence suggests that tau pathology co-localizes with RNA binding proteins (RBPs) that are known markers for stress granules (SGs). Here we used proteomics to determine how the network of tau binding proteins changes with disease in the rTg4510 mouse, and then followed up with immunohistochemistry to identify RNA binding proteins that co-localize with tau pathology. The tau interactome networks revealed striking disease-related changes in interactions between tau and a multiple RBPs, and biochemical fractionation studies demonstrated that many of these proteins including hnRNPA0, EWSR1, PABP and RPL7 form insoluble aggregates as tau pathology develops. Immunohistochemical analysis of mouse and human brain tissues suggest a model of evolving pathological interaction, in which RBPs co-localize with pathological phospho-tau but occur adjacent to larger pathological tau inclusions. We suggest a model in which tau initially interacts with RBPs in small complexes, but evolves into isolated aggregated inclusions as tau pathology matures.


Subject(s)
Brain/pathology , Inclusion Bodies/metabolism , Protein Aggregation, Pathological/etiology , RNA-Binding Proteins/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Animals , Cell Cycle Proteins/metabolism , Endodeoxyribonucleases/metabolism , Humans , Immunoprecipitation , Male , Mass Spectrometry , Mice , Mice, Transgenic , Middle Aged , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Phosphorylation/physiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Interaction Maps , Proteomics , T-Cell Intracellular Antigen-1/genetics , T-Cell Intracellular Antigen-1/metabolism
8.
Neurosci Lett ; 684: 109-114, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30003938

ABSTRACT

RNA binding proteins are a diverse class of proteins that regulate all aspects of RNA metabolism. Accumulating studies indicate that heterogeneous nuclear ribonucleoproteins are associated with cellular adaptations in response to drugs of abuse. We recently mapped and validated heterogeneous nuclear ribonucleoprotein H1 (Hnrnph1) as a quantitative trait gene underlying differential behavioral sensitivity to methamphetamine. The molecular mechanisms by which hnRNP H1 alters methamphetamine behaviors are unknown but could involve pre- and/or post-synaptic changes in protein localization and function. Methamphetamine initiates post-synaptic D1 dopamine receptor signaling indirectly by binding to pre-synaptic dopamine transporters and vesicular monoamine transporters of midbrain dopaminergic neurons which triggers reverse transport and accumulation of dopamine at the synapse. Here, we examined changes in neuronal localization of hnRNP H in primary rat cortical neurons that express dopamine receptors that can be modulated by the D1 or D2 dopamine receptor agonists SKF38393 and (-)-Quinpirole HCl, respectively. Basal immunostaining of hnRNP H was localized primarily to the nucleus. D1 dopamine receptor activation induced an increase in hnRNP H nuclear immunostaining as detected by immunocytochemistry with a C-domain directed antibody containing epitope near the glycine-rich domain but not with an N-domain specific antibody. Although there was no change in hnRNP H protein in the nucleus or cytoplasm, there was a decrease in Hnrnph1 transcript following D1 receptor stimulation. Taken together, these results suggest that D1 receptor activation increases availability of the hnRNP H C-terminal epitope, which could potentially reflect changes in protein-protein interactions. Thus, D1 receptor signaling could represent a key molecular post-synaptic event linking Hnrnph1 polymorphisms to drug-induced behavior.


Subject(s)
Dopamine Agonists/pharmacology , Dopaminergic Neurons/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Receptors, Dopamine D1/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Animals , Cells, Cultured , Dopaminergic Neurons/chemistry , Dopaminergic Neurons/drug effects , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/analysis , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/analysis
9.
Nat Neurosci ; 21(1): 72-80, 2018 01.
Article in English | MEDLINE | ID: mdl-29273772

ABSTRACT

Emerging studies suggest a role for tau in regulating the biology of RNA binding proteins (RBPs). We now show that reducing the RBP T-cell intracellular antigen 1 (TIA1) in vivo protects against neurodegeneration and prolongs survival in transgenic P301S Tau mice. Biochemical fractionation shows co-enrichment and co-localization of tau oligomers and RBPs in transgenic P301S Tau mice. Reducing TIA1 decreased the number and size of granules co-localizing with stress granule markers. Decreasing TIA1 also inhibited the accumulation of tau oligomers at the expense of increasing neurofibrillary tangles. Despite the increase in neurofibrillary tangles, TIA1 reduction increased neuronal survival and rescued behavioral deficits and lifespan. These data provide in vivo evidence that TIA1 plays a key role in mediating toxicity and further suggest that RBPs direct the pathway of tau aggregation and the resulting neurodegeneration. We propose a model in which dysfunction of the translational stress response leads to tau-mediated pathology.


Subject(s)
Gene Expression Regulation/genetics , RNA-Binding Proteins/metabolism , Tauopathies/metabolism , Tauopathies/prevention & control , tau Proteins/metabolism , Animals , Animals, Newborn , Cognition Disorders/etiology , Cognition Disorders/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , Cytoplasm/ultrastructure , Disease Models, Animal , Endoribonucleases/metabolism , Female , Locomotion/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/ultrastructure , Neurons/pathology , Neurons/ultrastructure , RNA-Binding Proteins/genetics , Synapses/metabolism , Synapses/ultrastructure , Tauopathies/genetics , Tauopathies/pathology , Trans-Activators/metabolism , tau Proteins/genetics
10.
Cell Rep ; 15(7): 1455-1466, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160897

ABSTRACT

Dendritic mislocalization of microtubule associated protein tau is a hallmark of tauopathies, but the role of dendritic tau is unknown. We now report that tau interacts with the RNA-binding protein (RBP) TIA1 in brain tissue, and we present the brain-protein interactome network for TIA1. Analysis of the TIA1 interactome in brain tissue from wild-type (WT) and tau knockout mice demonstrates that tau is required for normal interactions of TIA1 with proteins linked to RNA metabolism, including ribosomal proteins and RBPs. Expression studies show that tau regulates the distribution of TIA1, and tau accelerates stress granule (SG) formation. Conversely, TIA1 knockdown or knockout inhibits tau misfolding and associated toxicity in cultured hippocampal neurons, while overexpressing TIA1 induces tau misfolding and stimulates neurodegeneration. Pharmacological interventions that prevent SG formation also inhibit tau pathophysiology. These studies suggest that the pathophysiology of tauopathy requires an intimate interaction with RNA-binding proteins.


Subject(s)
RNA-Binding Proteins/metabolism , Tauopathies/metabolism , Tauopathies/physiopathology , tau Proteins/metabolism , tau Proteins/toxicity , Animals , Brain/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Dendrites/drug effects , Dendrites/metabolism , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Folding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Protein Synthesis Inhibitors/pharmacology , Protein Transport/drug effects , Proteome/metabolism , Solubility , T-Cell Intracellular Antigen-1
12.
Brain Res ; 1584: 52-8, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25108040

ABSTRACT

A feature of neurodegenerative disease is the accumulation of insoluble protein aggregates in the brain. In some conditions, including Amyotrophic Lateral Sclerosis and Frontotemporal lobar degeneration, the primary aggregating entities are RNA binding proteins. Through regulated prion-like assembly, RNA binding proteins serve many functions in RNA metabolism that are essential for the healthy maintenance of cells of the central nervous system. Those RNA binding proteins that are the core nucleating factors of stress granules (SGs), including TIA-1, TIAR, TTP and G3BP1, are also found in the pathological lesions of other neurological conditions, such as Alzheimer's disease, where the hallmark aggregating protein is not an RNA binding protein. This discovery suggests that the regulated cellular pathway, which utilizes assembly of RNA binding proteins to package and silence mRNAs during stress, may be integral in the aberrant pathological protein aggregation that occurs in numerous neurodegenerative conditions.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/pathology , Protein Aggregation, Pathological/metabolism , Stress, Physiological , Animals , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , DNA Helicases , DNA-Binding Proteins/metabolism , Humans , Mice , Poly(A)-Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , RNA-Binding Proteins/metabolism , Signal Transduction , T-Cell Intracellular Antigen-1 , Tristetraprolin/metabolism
13.
Mol Cell Neurosci ; 48(1): 72-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21740973

ABSTRACT

The sphingosine 1-phosphate (S1P) receptor modulators have emerged as a new therapeutic opportunity paradigm for the treatment of immune-mediated demyelinating diseases such as multiple sclerosis (MS). The S1P analog fingolimod (FTY720) has been shown to alleviate disease burden in immune-mediated animal models of MS, and has been approved for treatment in clinical trials in patients with MS in the United States. While the immunological effects of FTY720 are well established, there is controversy in the literature regarding the contribution of FTY720 on myelin repair. Here, we directly assessed the impact of FTY720 on myelin repair in cuprizone and lysolecithin (LPC) demyelination models that have a minimal immunological component. FTY720 failed to promote remyelination in either animal model. These studies suggest that while FTY720 may be effective at modulating the immunological attack in MS, it may benefit from an add-on therapy to enhance the myelin repair required for long-term functional restoration in MS.


Subject(s)
Immunosuppressive Agents/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Nerve Regeneration/drug effects , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Animals , Chelating Agents/pharmacology , Cuprizone/pharmacology , Demyelinating Diseases/drug therapy , Disease Models, Animal , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/therapeutic use , Mice , Mice, Inbred C57BL , Propylene Glycols/therapeutic use , Rats , Rats, Sprague-Dawley , Sphingosine/pharmacology , Sphingosine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL