Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
RNA Biol ; 21(1): 1-15, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38372062

ABSTRACT

Although Argonaute (AGO) proteins have been the focus of microRNA (miRNA) studies, we observed AGO-free mature miRNAs directly interacting with RNA-binding proteins, implying the sophisticated nature of fine-tuning gene regulation by miRNAs. To investigate microRNA-binding proteins (miRBPs) globally, we analyzed PAR-CLIP data sets to identify RBP quaking (QKI) as a novel miRBP for let-7b. Potential existence of AGO-free miRNAs were further verified by measuring miRNA levels in genetically engineered AGO-depleted human and mouse cells. We have shown that QKI regulates miRNA-mediated gene silencing at multiple steps, and collectively serves as an auxiliary factor empowering AGO2/let-7b-mediated gene silencing. Depletion of QKI decreases interaction of AGO2 with let-7b and target mRNA, consequently controlling target mRNA decay. This finding indicates that QKI is a complementary factor in miRNA-mediated mRNA decay. QKI, however, also suppresses the dissociation of let-7b from AGO2, and slows the assembly of AGO2/miRNA/target mRNA complexes at the single-molecule level. We also revealed that QKI overexpression suppresses cMYC expression at post-transcriptional level, and decreases proliferation and migration of HeLa cells, demonstrating that QKI is a tumour suppressor gene by in part augmenting let-7b activity. Our data show that QKI is a new type of RBP implicated in the versatile regulation of miRNA-mediated gene silencing.


Subject(s)
MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , HeLa Cells , Gene Silencing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , RNA, Messenger/genetics
2.
Proc Natl Acad Sci U S A ; 115(7): 1635-1640, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386392

ABSTRACT

Mutations in LRRK2 are known to be the most common genetic cause of sporadic and familial Parkinson's disease (PD). Multiple lines of LRRK2 transgenic or knockin mice have been developed, yet none exhibit substantial dopamine (DA)-neuron degeneration. Here we develop human tyrosine hydroxylase (TH) promoter-controlled tetracycline-sensitive LRRK2 G2019S (GS) and LRRK2 G2019S kinase-dead (GS/DA) transgenic mice and show that LRRK2 GS expression leads to an age- and kinase-dependent cell-autonomous neurodegeneration of DA and norepinephrine (NE) neurons. Accompanying the loss of DA neurons are DA-dependent behavioral deficits and α-synuclein pathology that are also LRRK2 GS kinase-dependent. Transmission EM reveals that that there is an LRRK2 GS kinase-dependent significant reduction in synaptic vesicle number and a greater abundance of clathrin-coated vesicles in DA neurons. These transgenic mice indicate that LRRK2-induced DA and NE neurodegeneration is kinase-dependent and can occur in a cell-autonomous manner. Moreover, these mice provide a substantial advance in animal model development for LRRK2-associated PD and an important platform to investigate molecular mechanisms for how DA neurons degenerate as a result of expression of mutant LRRK2.


Subject(s)
Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/physiology , Neurodegenerative Diseases/pathology , Norepinephrine/metabolism , Age Factors , Animals , Behavior, Animal , Dopaminergic Neurons/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Activity , Mutation , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism
3.
Proc Natl Acad Sci U S A ; 112(37): 11696-701, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26324925

ABSTRACT

Mutations in parkin lead to early-onset autosomal recessive Parkinson's disease (PD) and inactivation of parkin is thought to contribute to sporadic PD. Adult knockout of parkin in the ventral midbrain of mice leads to an age-dependent loss of dopamine neurons that is dependent on the accumulation of parkin interacting substrate (PARIS), zinc finger protein 746 (ZNF746), and its transcriptional repression of PGC-1α. Here we show that adult knockout of parkin in mouse ventral midbrain leads to decreases in mitochondrial size, number, and protein markers consistent with a defect in mitochondrial biogenesis. This decrease in mitochondrial mass is prevented by short hairpin RNA knockdown of PARIS. PARIS overexpression in mouse ventral midbrain leads to decreases in mitochondrial number and protein markers and PGC-1α-dependent deficits in mitochondrial respiration. Taken together, these results suggest that parkin loss impairs mitochondrial biogenesis, leading to declining function of the mitochondrial pool and cell death.


Subject(s)
Mitochondria/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Brain/embryology , Brain/metabolism , Cell Death , Cell Line, Tumor , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oxygen Consumption , Parkinson Disease/metabolism , Repressor Proteins/physiology , Ubiquitin-Protein Ligases/genetics
4.
Redox Biol ; 73: 103197, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38781730

ABSTRACT

Astrocytes are the major glial cells in the human brain and provide crucial metabolic and trophic support to neurons. The amyloid-ß peptide (Aß) alter the morphological and functional properties of astrocytes and induce inflammation and calcium dysregulation, contributing to Alzheimer's disease (AD) pathology. Recent studies highlight the role of Toll-like receptor (TLR) 4/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling in inflammation. Reactive oxygen species (ROS) generated due to Aß, induce apoptosis in the brain cells worsening AD progression. Astrocytic cell surface receptors, such as purinergic receptors (P2Y1 and P2Y2), metabotropic glutamate receptor (mGLUR)5, α7 nicotinic acetylcholine receptor (α7nAChR), and N-methyl-d-aspartate receptors (NMDARs), have been suggested to interact with inositol trisphosphate receptor (IP3R) on the endoplasmic reticulum (ER) to induce Ca2+ movement from ER to cytoplasm, causing Ca2+ dysregulation. We found that the citrus flavonoid nobiletin (NOB) protected primary astrocytes from Aß42-induced cytotoxicity and inhibited TLR4/NF-κB signaling in Aß42-induced primary rat astrocytes. NOB was found to regulate Aß42-induced ROS levels through Keap1-Nrf2 pathway. The receptors P2Y1, P2Y2, mGLUR5, α7nAChR, and NMDARs induced intracellular Ca2+ levels by activating IP3R and NOB regulated them, thereby regulating intracellular Ca2+ levels. Molecular docking analysis revealed a possible interaction between NOB and IP3R in IP3R regulation. Furthermore, RNA sequencing revealed various NOB-mediated biological signaling pathways, such as the AD-presenilin, AD-amyloid secretase, and Wnt signaling pathway, suggesting possible neuroprotective roles of NOB. To conclude, NOB is a promising therapeutic agent for AD and works by modulating AD pathology at various levels in Aß42-induced primary rat astrocytes.


Subject(s)
Amyloid beta-Peptides , Astrocytes , Calcium , Flavones , Inositol 1,4,5-Trisphosphate Receptors , Astrocytes/metabolism , Astrocytes/drug effects , Amyloid beta-Peptides/metabolism , Animals , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Rats , Calcium/metabolism , Flavones/pharmacology , Reactive Oxygen Species/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Humans , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology
5.
Amino Acids ; 45(4): 1003-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836417

ABSTRACT

Precise in-gel quantification of total protein amount of bands or spots in gels is the basis of subsequent biochemical, molecular biological and immunological analyses. Though several methods have been designed to evaluate relative amounts of proteins, these methods are of limited reliability because (semi-) quantifications depend on the amount of protein migrating into the gel and different proteins may lead to different absorptions/intensities of stained bands or spots. In the present study, we described a method to quantify both, hydrophilic and hydrophobic proteins using in-gel digestion with proteinase K, subsequent extraction and acid hydrolysis followed by the use of the ninhydrin reaction. The protocol is accurate and compatible with mass spectrometric characterization of proteins. Reproducible in-gel protein quantification was performed from SDS-PAGE and IEF/SDS-PAGE gels using bovine serum albumin as a standard protein. Bacteriorhodopsin separated on SDS-PAGE gel was quantified in addition in order to show that the method is also suitable for quantification of hydrophobic protein. This protocol for reliable in-gel protein quantification, which not only provides "arbitrary units of optical density", can also be completed in a minimum of 4 days or maximum 1 week depending on the type of electrophoresis with the disadvantage of being time consuming.


Subject(s)
Bacteriorhodopsins/analysis , Gels , Ninhydrin/chemistry , Serum Albumin, Bovine/analysis , Staining and Labeling/methods , Acids/chemistry , Animals , Bacteriorhodopsins/chemistry , Cattle , Electrophoresis, Polyacrylamide Gel , Endopeptidase K/metabolism , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Mass Spectrometry , Serum Albumin, Bovine/chemistry
6.
J Parkinsons Dis ; 13(2): 145-164, 2023.
Article in English | MEDLINE | ID: mdl-36710687

ABSTRACT

Regulation of gene expression by epigenetic modifications means lasting and heritable changes in the function of genes without alterations in the DNA sequence. Of all epigenetic mechanisms identified thus far, DNA methylation has been of particular interest in both aging and age-related disease research over the last decade given the consistency of site-specific DNA methylation changes during aging that can predict future health and lifespan. An increasing line of evidence has implied the dynamic nature of DNA (de)methylation events that occur throughout the lifespan has a role in the pathophysiology of aging and age-associated neurodegenerative conditions, including Parkinson's disease (PD). In this regard, PD methylome shows, to some extent, similar genome-wide changes observed in the methylome of healthy individuals of matching age. In this review, we start by providing a brief overview of studies outlining global patterns of DNA methylation, then its mechanisms and regulation, within the context of aging and PD. Considering diverging lines of evidence from different experimental and animal models of neurodegeneration and how they combine to shape our current understanding of tissue-specific changes in DNA methylome in health and disease, we report a high-level comparison of the genomic methylation landscapes of brain, with an emphasis on dopaminergic neurons in PD and in natural aging. We believe this will be particularly useful for systematically dissecting overlapping genome-wide alterations in DNA methylation during PD and healthy aging, and for improving our knowledge of PD-specific changes in methylation patterns independent of aging process.


Subject(s)
DNA Methylation , Parkinson Disease , Animals , Parkinson Disease/metabolism , Epigenesis, Genetic , Aging/genetics , Aging/metabolism , Epigenome
7.
Sci Transl Med ; 15(724): eadd0499, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38019930

ABSTRACT

Pathologic α-synuclein plays an important role in the pathogenesis of α-synucleinopathies such as Parkinson's disease (PD). Disruption of proteostasis is thought to be central to pathologic α-synuclein toxicity; however, the molecular mechanism of this deregulation is poorly understood. Complementary proteomic approaches in cellular and animal models of PD were used to identify and characterize the pathologic α-synuclein interactome. We report that the highest biological processes that interacted with pathologic α-synuclein in mice included RNA processing and translation initiation. Regulation of catabolic processes that include autophagy were also identified. Pathologic α-synuclein was found to bind with the tuberous sclerosis protein 2 (TSC2) and to trigger the activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), which augmented mRNA translation and protein synthesis, leading to neurodegeneration. Genetic and pharmacologic inhibition of mTOR and protein synthesis rescued the dopamine neuron loss, behavioral deficits, and aberrant biochemical signaling in the α-synuclein preformed fibril mouse model and Drosophila transgenic models of pathologic α-synuclein-induced degeneration. Pathologic α-synuclein furthermore led to a destabilization of the TSC1-TSC2 complex, which plays an important role in mTORC1 activity. Constitutive overexpression of TSC2 rescued motor deficits and neuropathology in α-synuclein flies. Biochemical examination of PD postmortem brain tissues also suggested deregulated mTORC1 signaling. These findings establish a connection between mRNA translation deregulation and mTORC1 pathway activation that is induced by pathologic α-synuclein in cellular and animal models of PD.


Subject(s)
Parkinson Disease , Animals , Mice , alpha-Synuclein/metabolism , Disease Models, Animal , Mammals/metabolism , Mechanistic Target of Rapamycin Complex 1 , Parkinson Disease/metabolism , Proteomics , TOR Serine-Threonine Kinases
8.
J Neurochem ; 123(4): 515-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22928743

ABSTRACT

2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) is a quantitatively major enzyme in myelin, where it localizes to the non-compact regions and is bound to the membrane surface. Although its catalytic activity in vitro has been characterized, the physiological function and in vivo substrate of CNPase remain unknown. Especially the N-terminal domain has been poorly characterized; previously, we have shown it is involved in CNPase dimerization and RNA binding. Here, we show that purified CNPase binds to the calcium sensor protein calmodulin (CaM) in a calcium-dependent manner; the binding site is in the N-terminal domain of CNPase. CaM does not affect the phosphodiesterase activity of CNPase in vitro, nor does it influence polyadenylic acid binding. The colocalization of CNPase and CaM during Schwann cell myelination in culture was observed, and CaM antagonists induced the colocalization of CNPase with microtubules in differentiated CG-4 oligodendrocytes. An analysis of post-translational modifications of CNPase from rat brain revealed the presence of two novel phosphorylation sites on Tyr110 and Ser169 within the N-terminal domain. The results indicate a role for the N-terminal domain of CNPase in mediating multiple molecular interactions and provide a starting point for detailed structure-function studies on CNPase and its N-terminal domain.


Subject(s)
2',3'-Cyclic Nucleotide 3'-Phosphodiesterase , Calmodulin/metabolism , Protein Structure, Tertiary/physiology , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/chemistry , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Animals , Brain/cytology , Brain/metabolism , Calmodulin/genetics , Chromatography, Affinity , Chromatography, Gel/methods , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Nerve Fibers, Myelinated/metabolism , Oligodendroglia , Organ Culture Techniques , Phosphorylation/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , Protein Structure, Tertiary/genetics , Proteomics , Rats , Schwann Cells/enzymology , Surface Plasmon Resonance
9.
Neuron ; 110(15): 2422-2437.e9, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35654037

ABSTRACT

Parkinson's disease (PD) is mediated, in part, by intraneuronal accumulation of α-synuclein aggregates andsubsequent death of dopamine (DA) neurons in the substantia nigra pars compacta (SNpc). Microglial hyperactivation of the NOD-like receptor protein 3 (NLRP3) inflammasome has been well-documented in various neurodegenerative diseases, including PD. We show here that loss of parkin activity in mouse and human DA neurons results in spontaneous neuronal NLRP3 inflammasome assembly, leading to DA neuron death. Parkin normally inhibits inflammasome priming by ubiquitinating and targeting NLRP3 for proteasomal degradation. Loss of parkin activity also contributes to the assembly of an active NLRP3 inflammasome complex via mitochondrial-derived reactive oxygen species (mitoROS) generation through the accumulation of another parkin ubiquitination substrate, ZNF746/PARIS. Inhibition of neuronal NLRP3 inflammasome assembly prevents degeneration of DA neurons in familial and sporadic PD models. Strategies aimed at limiting neuronal NLRP3 inflammasome activation hold promise as a disease-modifying therapy for PD.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Parkinson Disease , Ubiquitin-Protein Ligases , Animals , Dopaminergic Neurons/metabolism , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Proteomics ; 11(3): 481-4, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268276

ABSTRACT

Until now quantification of proteins in gel-based methods relies on the amount of protein loaded onto the gel. This does not, however, represent the amount of proteins in the gel and therefore determination of proteins within the gel is mandatory. A method to quantify proteins, both hydrophilic and hydrophobic, using in-gel digestion with proteases, subsequent acid hydrolysis and the use of the ninhydrin reaction is herein presented.


Subject(s)
Gels , Indicators and Reagents/analysis , Ninhydrin/analysis , Proteins/analysis , Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Hydrolysis
11.
Proteomics ; 11(11): 2171-81, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538884

ABSTRACT

The brain GABA(A) receptor (GABA(A) R) is a key element of signaling and neural transmission in health and disease. Recently, complete sequence analysis of the recombinant GABA(A) R has been reported, separation and mass spectrometrical (MS) characterisation from tissue, however, has not been published so far. Hippocampi were homogenised, put on a sucrose gradient 10-69% and the layer from 10 to 20% was used for extraction of membrane proteins by a solution of Triton X-100, 1.5 M aminocaproic acid in the presence of 0.3 M Bis-Tris. This mixture was subsequently loaded onto blue native PAGE (BN-PAGE) with subsequent analysis on denaturing gel systems. Spots from the 3-DE electrophoretic run were stained with Colloidal Coomassie Brilliant Blue, and spots with an apparent molecular weight between 40 and 60 kDa were picked and in-gel digested with trypsin, chymotrypsin and subtilisin. The resulting peptides were analysed by nano-LC-ESI-MS/MS (ion trap) and protein identification was carried out using MASCOT searches. In addition, known GABA(A) R-specific MS information taken from own previous studies was used for searches of GABA(A) R subunits. ß-1, ß-2 and ß-3, θ and ρ-1 subunits were detected and six novel phosphorylation sites were observed and verified by phosphatase treatment. The method used herein enables identification of several GABA(A) R subunits from mouse hippocampus along with phosphorylations of ß-1 (T227, Y230), ß-2 (Y215, T439) and ß-3 (T282, S406) subunits. The procedure forms the basis for GABA(A) R studies at the protein chemical rather than at the immunochemical level in health and disease.


Subject(s)
Hippocampus/chemistry , Hippocampus/metabolism , Mass Spectrometry/methods , Protein Processing, Post-Translational , Proteomics/methods , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Animals , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Receptors, GABA-A/isolation & purification
12.
Hippocampus ; 21(7): 714-23, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20740491

ABSTRACT

There is a significant strain-dependent performance in the Morris water maze (MWM), a paradigm for the evaluation of spatial memory. In contrast, information on molecular differences that may be responsible for differences in spatial memory performance is limited. The aim of the study was therefore to investigate differences in hippocampal protein levels in three groups with different performance in the MWM. C57BL/6J (inbred laboratory strain), PWD/PhJ (inbred strain derived from wild animals of Mus musculus), and Apodemus sylvaticus (AS, genus Apodemus) mice were used for the experiments. Proteins from hippocampi, obtained from a behavioral study on these animals, were extracted and run on two-dimensional gel electrophoresis. Proteins spots were quantified, and spots with significantly different levels were identified by mass spectrometry using an ion trap. A series of 49 proteins from different pathways and cascades (signaling, neuronal network, protein synthesis, secretion and degradation, and antioxidant system; intermediary, fat, and carbohydrate metabolism) were significantly different among hippocampi at the stringent statistical level of P ≤ 0.001. These findings are paralleled by differences in the spatial navigation abilities between the strains within the species Mus musculus (C57BL/6 vs. PWD/PhJ) and between the genera Mus and Apodemus. As shown previously, AS learned the task in the MWM and showed good memory retention when tested at the probe trial (day 12), whereas C57BL/6J learned the task, but failed at the probe trial at day 12 as well as PWD/PhJ that failed to learn the task and failed at the probe trial at day 12. A list of above-mentioned proteins were different between PWD/PhJ with bad and AS with good memory retention and may therefore be related to performance in the MWM and thus to spatial memory formation. The experimental approach, however, does not allow discriminating between differences in protein levels a priori and different protein levels induced by the MWM testing. © 2010 Wiley-Liss, Inc.


Subject(s)
Hippocampus/metabolism , Maze Learning/physiology , Memory/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Space Perception/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Murinae , Species Specificity
13.
Amino Acids ; 40(2): 641-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20640460

ABSTRACT

Astrocytes are glial cells in the central nervous system (CNS) that play key roles in brain physiology, controlling processes, such as neurogenesis, brain energy metabolism and synaptic transmission. Recently, immune functions have also been demonstrated in astrocytes, influencing neuronal survival in the course of neuroinflammatory pathologies. In this regard, PKCepsilon (PKCε) is a protein kinase with an outstanding role in inflammation. Our previous findings indicating that PKCε regulates voltage-dependent calcium channels as well as morphological stellation imply that this kinase controls multiple signalling pathways within astrocytes, including those implicated in activation of immune functions. The present study applies proteomics to investigate new protein targets of PKCε in astrocytes. Primary astrocyte cultures infected with an adenovirus that expresses constitutively active PKCε were compared with infection controls. Two-dimensional gel electrophoresis clearly detected 549 spots in cultured astrocytes, and analysis of differential protein expression revealed 18 spots regulated by PKCε. Protein identification by mass spectrometry (nano-LC-ESI-MS/MS) showed that PKCε targets molecules with heterogeneous functions, including chaperones, cytoskeletal components and proteins implicated in metabolism and signalling. These results support the notion that PKCε is involved in astrocyte activation; also suggesting that multiple astrocyte-dependent processes are regulated by PKCε, including those associated to neuroinflammation.


Subject(s)
Astrocytes/chemistry , Astrocytes/enzymology , Cell Proliferation , Protein Kinase C-epsilon/metabolism , Proteomics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Kinase C-epsilon/genetics , Signal Transduction
14.
Front Integr Neurosci ; 15: 769142, 2021.
Article in English | MEDLINE | ID: mdl-35153687

ABSTRACT

Music therapy (MT) and other rhythmic-based interventions for the treatment of neurodegeneration (ND) have been successful in improving the quality of life of affected individuals. Music therapy and rhythm-based stimuli affect patients with Alzheimer's disease (AD) and Parkinson's disease (PD) respectively not only through cognitive channels and subjective qualifications but also through altered brain structures and neural systems. Often implicated in the pathogenesis and resulting symptoms of these diseases is the role of aberrant circadian rhythmicity (CR), namely disrupted sleep. Recent literature suggests that proper maintenance of this timekeeping framework may be beneficial for patients with neurodegenerative disorders and serve a neuroprotective role. While music therapy can improve the quality of life for neurodegenerative patients, longitudinal studies analyzing sleep patterns of affected individuals and possible mechanisms of intervention remain sparse. Furthermore, the role of music therapy in the context of circadian rhythmicity has not been adequately explored. By analyzing the links between circadian rhythmicity, neurodegeneration, and music therapy, a more comprehensive picture emerges, suggesting that possible uses of non-pharmacological circadian-based music therapy to target mechanisms involved in the pathogenesis of Alzheimer's disease and Parkinson's disease may enhance clinical treatment and potentially indicate neuroprotection as a preventative measure.

15.
Sci Rep ; 11(1): 21500, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728675

ABSTRACT

The transcriptional repressor called parkin interacting substrate (PARIS; ZNF746) was initially identified as a novel co-substrate of parkin and PINK1 that leads to Parkinson's disease (PD) by disrupting mitochondrial biogenesis through peroxisome proliferator-activated receptor gamma (PPARγ) coactivator -1α (PGC-1α) suppression. Since its initial discovery, growing evidence has linked PARIS to defective mitochondrial biogenesis observed in PD pathogenesis. Yet, dopaminergic (DA) neuron-specific mechanistic underpinnings and genome-wide PARIS binding landscape has not been explored. We employed conditional translating ribosome affinity purification (TRAP) followed by RNA sequencing (TRAP-seq) for transcriptome profiling of DA neurons in transgenic Drosophila lines expressing human PARIS wild type (WT) or mutant (C571A). We also generated genome-wide maps of PARIS occupancy using ChIP-seq in human SH-SY5Y cells. The results demonstrated that PPARγ functions as a master regulator of PARIS-induced molecular changes at the transcriptome level, confirming that PARIS acts primarily on PGC-1α to lead to neurodegeneration in PD. Moreover, we identified that PARIS actively modulates expression of PPARγ target genes by physically binding to the promoter regions. Together, our work revealed how PARIS drives adverse effects on modulation of PPAR-γ associated gene clusters in DA neurons.


Subject(s)
Dopaminergic Neurons/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation , Neuroblastoma/metabolism , PPAR gamma/metabolism , Repressor Proteins/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Genome-Wide Association Study , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , PPAR gamma/genetics , RNA-Seq , Repressor Proteins/genetics , Tumor Cells, Cultured
16.
Nat Neurosci ; 24(12): 1673-1685, 2021 12.
Article in English | MEDLINE | ID: mdl-34782793

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating disorder in which motor neurons degenerate, the causes of which remain unclear. In particular, the basis for selective vulnerability of spinal motor neurons (sMNs) and resistance of ocular motor neurons to degeneration in ALS has yet to be elucidated. Here, we applied comparative multi-omics analysis of human induced pluripotent stem cell-derived sMNs and ocular motor neurons to identify shared metabolic perturbations in inherited and sporadic ALS sMNs, revealing dysregulation in lipid metabolism and its related genes. Targeted metabolomics studies confirmed such findings in sMNs of 17 ALS (SOD1, C9ORF72, TDP43 (TARDBP) and sporadic) human induced pluripotent stem cell lines, identifying elevated levels of arachidonic acid. Pharmacological reduction of arachidonic acid levels was sufficient to reverse ALS-related phenotypes in both human sMNs and in vivo in Drosophila and SOD1G93A mouse models. Collectively, these findings pinpoint a catalytic step of lipid metabolism as a potential therapeutic target for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism , Lipid Metabolism/genetics , Mice , Mice, Transgenic , Motor Neurons/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics
17.
Neuron ; 109(23): 3758-3774.e11, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34644545

ABSTRACT

Impairment in glucocerebrosidase (GCase) is strongly associated with the development of Parkinson's disease (PD), yet the regulators responsible for its impairment remain elusive. In this paper, we identify the E3 ligase Thyroid Hormone Receptor Interacting Protein 12 (TRIP12) as a key regulator of GCase. TRIP12 interacts with and ubiquitinates GCase at lysine 293 to control its degradation via ubiquitin proteasomal degradation. Ubiquitinated GCase by TRIP12 leads to its functional impairment through premature degradation and subsequent accumulation of α-synuclein. TRIP12 overexpression causes mitochondrial dysfunction, which is ameliorated by GCase overexpression. Further, conditional TRIP12 knockout in vitro and knockdown in vivo promotes the expression of GCase, which blocks α-synuclein preformed fibrils (α-syn PFFs)-provoked dopaminergic neurodegeneration. Moreover, TRIP12 accumulates in human PD brain and α-synuclein-based mouse models. The identification of TRIP12 as a regulator of GCase provides a new perspective on the molecular mechanisms underlying dysfunctional GCase-driven neurodegeneration in PD.


Subject(s)
Carrier Proteins/metabolism , Glucosylceramidase , Parkinson Disease , Ubiquitin-Protein Ligases/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Glucosylceramidase/genetics , Glucosylceramidase/metabolism , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Ubiquitination , alpha-Synuclein/metabolism
18.
Sci Transl Med ; 13(604)2021 07 28.
Article in English | MEDLINE | ID: mdl-34321320

ABSTRACT

Accumulation of the parkin-interacting substrate (PARIS; ZNF746), due to inactivation of parkin, contributes to Parkinson's disease (PD) through repression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α; PPARGC1A) activity. Here, we identify farnesol as an inhibitor of PARIS. Farnesol promoted the farnesylation of PARIS, preventing its repression of PGC-1α via decreasing PARIS occupancy on the PPARGC1A promoter. Farnesol prevented dopaminergic neuronal loss and behavioral deficits via farnesylation of PARIS in PARIS transgenic mice, ventral midbrain transduction of AAV-PARIS, adult conditional parkin KO mice, and the α-synuclein preformed fibril model of sporadic PD. PARIS farnesylation is decreased in the substantia nigra of patients with PD, suggesting that reduced farnesylation of PARIS may play a role in PD. Thus, farnesol may be beneficial in the treatment of PD by enhancing the farnesylation of PARIS and restoring PGC-1α activity.


Subject(s)
Parkinson Disease , Animals , Dopamine , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Prenylation , Repressor Proteins/metabolism , Substantia Nigra/metabolism
19.
J Proteome Res ; 9(3): 1591-7, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20141154

ABSTRACT

Although changes in protein expression in spinal cord injury (SCI) would be of pivotal interest, information so far is limited. It was therefore the aim of the study to determine protein levels and post-translational modifications in the early phase following SCI in the rat. SCI was induced in Sprague-Dawley rats and sham operated rats served as controls. A gel-based proteomic approach using two-dimensional gel electrophoresis followed by quantification with specific software and subsequent identification of differentially expressed proteins by nano-ESI-LC-MS/MS was applied. Proteins of several pathways and cascades were dysregulated in SCI: 14-3-3 epsilon protein, dynein light chain 1, and tubulin beta-5 chain showed higher levels in SCI, whereas adenylyl cyclase associated protein 1, dihydropyrimidinase-related protein 2, F-actin capping protein subunit beta, glyceraldehyde-3-phosphate dehydrogenase, stress-induced phosphoprotein 1 and transthyretin showed lower levels in the injured tissue. Post-translational modifications indicated free oxygen radical attack on proteins in SCI. The occurrence of stress is indicated by deranged stress-induced phosphoprotein 1 and signaling abnormalities are reflected by adenylyl cyclase-associated protein 1 and 14-3-3 epsilon protein. The findings propose the involvement of the corresponding cascades and challenge further work into aberrant signaling and oxidative stress in SCI, which may form the basis for experimental intervention for spinal cord trauma.


Subject(s)
Protein Processing, Post-Translational , Proteins/analysis , Proteomics/methods , Spinal Cord Injuries/metabolism , 14-3-3 Proteins/analysis , 14-3-3 Proteins/metabolism , Animals , Blotting, Western , Cytoskeletal Proteins/analysis , Cytoskeletal Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Male , Mass Spectrometry , Phosphorylation , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Tubulin/analysis , Tubulin/metabolism
20.
Electrophoresis ; 31(11): 1813-21, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20506418

ABSTRACT

Aralar1 is a mitochondrial aspartate/glutamate carrier and a key component of the malate-aspartate NADH shuttle system. An analytical approach to obtain high sequence coverage is important to predict conformation, identify splice variants and binding partners or generate specific antibodies. Moreover, a method allowing determination of Aralar1 from brain samples is a prerequisite for evaluating a biological role. Sucrose gradient ultracentrifugation was applied to enrich native membrane protein fractions and these were run on blue-native PAGE, followed by multidimensional gel electrophoresis. Spots from the third-dimensional gel electrophoresis were in-gel digested with trypsin, chymotrypsin and subtilisin. Subsequently, peptides were analyzed by nano-ESI-LC-MS/MS using collision-induced dissociation and electron transfer dissociation modes. Modiro v1.1 along with Mascot v2.2 software was used for data handling. Aralar1 could be clearly separated, unambiguously identified and characterized from protein extracts of mouse hippocampus by the use of the multidimensional gel electrophoretic steps. The combined sequence coverage of Aralar1 from trypsin, chymotrypsin and subtilisin digestions was 99.85%. The results provide the basis for future studies of Aralar1 at the protein chemical rather than at the immunochemical level in the brain and thus challenge and enable determination of Aralar1 levels required for understanding biological functions in health and disease.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Hippocampus/chemistry , Mass Spectrometry/methods , Membrane Transport Proteins/analysis , Mitochondrial Proteins/analysis , Proteomics/methods , Animals , Cell Membrane/chemistry , Hydrophobic and Hydrophilic Interactions , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/metabolism , Peptide Fragments/analysis , Peptide Fragments/metabolism , Sequence Analysis, Protein , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL