Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38798570

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder and lacks disease-modifying therapies. We developed a Drosophila model for identifying novel glial-based therapeutic targets for PD. Human alpha-synuclein is expressed in neurons and individual genes are independently knocked down in glia. We performed a forward genetic screen, knocking down the entire Drosophila kinome in glia in alpha-synuclein expressing flies. Among the top hits were five genes (Ak1, Ak6, Adk1, Adk2, and awd) involved in adenosine metabolism. Knockdown of each gene improved locomotor dysfunction, rescued neurodegeneration, and increased brain adenosine levels. We determined that the mechanism of neuroprotection involves adenosine itself, as opposed to a downstream metabolite. We dove deeper into the mechanism for one gene, Ak1, finding rescue of dopaminergic neuron loss, alpha-synuclein aggregation, and bioenergetic dysfunction after glial Ak1 knockdown. We performed metabolomics in Drosophila and in human PD patients, allowing us to comprehensively characterize changes in purine metabolism and identify potential biomarkers of dysfunctional adenosine metabolism in people. These experiments support glial adenosine as a novel therapeutic target in PD.

2.
Genome Res ; 34(4): 590-605, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38599684

ABSTRACT

Missense mutations in the gene encoding the microtubule-associated protein TAU (current and approved symbol is MAPT) cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human TAU in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas-mediated gene editing to model frontotemporal dementia caused by the TAU P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for Tau P251L display age-dependent neurodegeneration, display metabolic defects, and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies, we performed single-cell RNA sequencing on approximately 130,000 cells from brains of Tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified. Gene expression was also altered in glial cells, suggestive of non-cell-autonomous regulation. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell type-specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy that faithfully recapitulates the genetic context and phenotypic features of the human disease, and use the results of comprehensive single-cell sequencing analysis to outline pathways of neurotoxicity and highlight the potential role of non-cell-autonomous changes in glia.


Subject(s)
Disease Models, Animal , Drosophila Proteins , Neuroglia , Neurons , Tauopathies , tau Proteins , Animals , Neuroglia/metabolism , tau Proteins/metabolism , tau Proteins/genetics , Neurons/metabolism , Neurons/pathology , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Signal Transduction , Drosophila melanogaster/genetics , Gene Knock-In Techniques , Drosophila/genetics , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Animals, Genetically Modified , Gene Editing , CRISPR-Cas Systems
3.
bioRxiv ; 2024 Feb 04.
Article in English | MEDLINE | ID: mdl-38352559

ABSTRACT

Missense mutations in the gene encoding the microtubule-associated protein tau cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human tau in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease, but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas mediated gene editing to model frontotemporal dementia caused by the tau P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for tau P251L display age-dependent neurodegeneration, metabolic defects and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies we performed single-cell RNA sequencing on approximately 130,000 cells from brains of tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified and non-cell autonomously in glial cells. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell-type specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy, which faithfully recapitulates the genetic context and phenotypic features of the human disease and use the results of comprehensive single cell sequencing analysis to outline pathways of neurotoxicity and highlight the role of non-cell autonomous changes in glia.

4.
bioRxiv ; 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38328119

ABSTRACT

As the most abundant glial cells in the CNS, astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress have remained elusive. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stresser, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, neurotoxic stress induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechnistically, YTHDF2 RIP-sequencing identified MAP2K4 ( MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed Mn-exposed astrocytes mediates proinflammatory response by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves a key upstream 'molecular switch' controlling SEK1( MAP2K4 )-JNK-cJUN proinflammatory signaling in astrocytes.

5.
Hum Mol Genet ; 33(10): 860-871, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38324746

ABSTRACT

Neuromuscular disorders caused by dysfunction of the mitochondrial respiratory chain are common, severe and untreatable. We recovered a number of mitochondrial genes, including electron transport chain components, in a large forward genetic screen for mutations causing age-related neurodegeneration in the context of proteostasis dysfunction. We created a model of complex I deficiency in the Drosophila retina to probe the role of protein degradation abnormalities in mitochondrial encephalomyopathies. Using our genetic model, we found that complex I deficiency regulates both the ubiquitin/proteasome and autophagy/lysosome arms of the proteostasis machinery. We further performed an in vivo kinome screen to uncover new and potentially druggable mechanisms contributing to complex I related neurodegeneration and proteostasis failure. Reduction of RIOK kinases and the innate immune signaling kinase pelle prevented neurodegeneration in complex I deficiency animals. Genetically targeting oxidative stress, but not RIOK1 or pelle knockdown, normalized proteostasis markers. Our findings outline distinct pathways controlling neurodegeneration and protein degradation in complex I deficiency and introduce an experimentally facile model in which to study these debilitating and currently treatment-refractory disorders.


Subject(s)
Disease Models, Animal , Drosophila Proteins , Electron Transport Complex I , Electron Transport Complex I/deficiency , Mitochondria , Mitochondrial Diseases , Proteostasis , Animals , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/pathology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/deficiency , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Autophagy/genetics , Oxidative Stress/genetics , Drosophila melanogaster/genetics , Mutation , Lysosomes/metabolism , Lysosomes/genetics , Drosophila/genetics , Drosophila/metabolism , Signal Transduction
6.
bioRxiv ; 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38045419

ABSTRACT

Occupational exposure to manganese (Mn) induces manganism and has been widely linked as a contributing environmental factor to Parkinson's disease (PD), featuring dramatic signature overlaps between the two in motor symptoms and clinical hallmarks. However, the molecular mechanism underlying such link remains elusive, and for combating PD, effective mechanism-based therapies are lacking. Here, we developed an adult Drosophila model of Mn toxicity to recapitulate key parkinsonian features, spanning behavioral deficits, neuronal loss, and dysfunctions in lysosome and mitochondria. We performed global metabolomics on flies at an early stage of toxicity and identified metabolism of the B vitamin, biotin (vitamin B 7 ), as a master pathway underpinning Mn toxicity with systemic, body-brain increases in Mn-treated groups compared to the controls. Using Btnd RNAi mutant flies, we show that biotin depletion exacerbates Mn-induced neurotoxicity, parkinsonism, and mitochondrial dysfunction; while in Mn-exposed wild-type flies, biotin feeding dramatically ameliorates these pathophenotypes. We further show in human induced stem cells (iPSCs)- differentiated midbrain dopaminergic neurons that the supplemented biotin protects against Mn-induced neuronal loss, cytotoxicity, and mitochondrial dysregulation. Finally, human data profiling biotin-related proteins show for PD cases elevated circulating levels of biotin transporters but not of metabolic enzymes compared to healthy controls, suggesting humoral biotin transport as a key event involved in PD. Taken together, our findings identified compensatory biotin pathway as a convergent, systemic driver of Mn toxicity and parkinsonian pathology, providing new basis for devising effective countermeasures against manganism and PD. Significance Statement: Environmental exposure to manganese (Mn) may increase the risk for Parkinson's disease (PD); however, the mechanistic basis linking the two remains unclear. Our adult fruit fly ( Drosophila ) model of Mn toxicity recapitulated key Parkinson's hallmarks in vivo spanning behavioral deficits, neuronal loss, and mitochondrial dysfunction. Metabolomics identified the biotin (vitamin B 7 ) pathway as a key mediator, featuring systemic biotin increases in the flies. Rescue trials leveraging biotin-deficient flies, wild-type flies, and human iPSC-derived dopaminergic neurons determined biotin as a driver of manganism, with the parkinsonian phenotypes dramatically reversed through biotin supplementation. Our findings, in line with overexpressed circulating biotin transporters observed in PD patients, suggest compensatory biotin pathway as a key to untangle the Mn-PD link for combating neurodegenerative disease.

8.
Neurobiol Dis ; 174: 105861, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36115552

ABSTRACT

Neuroinflammation is a critical pathophysiological hallmark of neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and traumatic brain injury (TBI). Microglia, the first responders of the brain, are the drivers of this neuroinflammation. Microglial activation, leading to induction of pro-inflammatory factors, like Interleukin 1-ß (IL-1ß), Tumor necrosis factor-α (TNFα), nitrites, and others, have been shown to induce neurodegeneration. Non-steroidal anti-inflammatory drugs (NSAIDs) have been shown to reduce the risk of developing PD, but the mechanism underlying the microglial activation is still under active research. Recently, microglial ion channels have come to the forefront as potential drug targets in multiple neurodegenerative disorders, including AD and PD. Microglia expresses a variety of ion channels, including potassium channels, calcium channels, chloride channels, sodium channels, and proton channels. The diversity of channels present on microglia is responsible for the dynamic nature of these immune cells of the brain. These ion channels regulate microglial proliferation, chemotaxis, phagocytosis, antigen recognition and presentation, apoptosis, and cell signaling leading to inflammation, among other critical critical functions. Understanding the role of these ion channels and the signaling mechanism these channels regulate under pathological conditions is an active area of research. This review will be focusing on the roles of different microglial ion channels, and their potential role in regulating microglial functions in neurodegenerative disorders.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Parkinson Disease , Humans , Microglia/metabolism , Inflammation/pathology , Ion Channels , Neurodegenerative Diseases/pathology , Tumor Necrosis Factor-alpha/metabolism , Parkinson Disease/pathology , Alzheimer Disease/pathology
10.
Cell ; 185(12): 2035-2056.e33, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35688132

ABSTRACT

Alpha-synuclein (αS) is a conformationally plastic protein that reversibly binds to cellular membranes. It aggregates and is genetically linked to Parkinson's disease (PD). Here, we show that αS directly modulates processing bodies (P-bodies), membraneless organelles that function in mRNA turnover and storage. The N terminus of αS, but not other synucleins, dictates mutually exclusive binding either to cellular membranes or to P-bodies in the cytosol. αS associates with multiple decapping proteins in close proximity on the Edc4 scaffold. As αS pathologically accumulates, aberrant interaction with Edc4 occurs at the expense of physiologic decapping-module interactions. mRNA decay kinetics within PD-relevant pathways are correspondingly disrupted in PD patient neurons and brain. Genetic modulation of P-body components alters αS toxicity, and human genetic analysis lends support to the disease-relevance of these interactions. Beyond revealing an unexpected aspect of αS function and pathology, our data highlight the versatility of conformationally plastic proteins with high intrinsic disorder.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , Parkinson Disease/metabolism , Processing Bodies , RNA Stability , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
11.
Int J Biochem Cell Biol ; 147: 106225, 2022 06.
Article in English | MEDLINE | ID: mdl-35550926

ABSTRACT

Despite the growing recognition that gastrointestinal (GI) dysfunction is prevalent in Parkinson's disease (PD) and occurs as a major prodromal symptom of PD, its cellular and molecular mechanisms remain largely unknown. Among the various types of GI cells, enteric glial cells (EGCs), which resemble astrocytes in structure and function, play a critical role in the pathophysiology of many GI diseases including PD. Thus, we investigated how EGCs respond to the environmental pesticides rotenone (Rot) and tebufenpyrad (Tebu) in cell and animal models to better understand the mechanism underlying GI abnormalities. Both Rot and Tebu induce dopaminergic neuronal cell death through complex 1 inhibition of the mitochondrial respiratory chain. We report that exposing a rat enteric glial cell model (CRL-2690 cells) to these pesticides increased mitochondrial fission and reduced mitochondrial fusion by impairing MFN2 function. Furthermore, they also increased mitochondrial superoxide generation and impaired mitochondrial ATP levels and basal respiratory rate. Measurement of LC3, p62 and lysosomal assays revealed impaired autolysosomal function in ECGs during mitochondrial stress. Consistent with our recent findings that mitochondrial dysfunction augments inflammation in astrocytes and microglia, we found that neurotoxic pesticide exposure also enhanced the production of pro-inflammatory factors in EGCs in direct correlation with the loss in mitochondrial mass. Finally, we show that pesticide-induced mitochondrial defects functionally impaired smooth muscle velocity, acceleration, and total kinetic energy in a mixed primary culture of the enteric nervous system (ENS). Collectively, our studies demonstrate for the first time that exposure to environmental neurotoxic pesticides impairs mitochondrial bioenergetics and activates inflammatory pathways in EGCs, further augmenting mitochondrial dysfunction and pro-inflammatory events to induce gut dysfunction. Our findings have major implications in understanding the GI-related pathogenesis and progression of environmentally linked PD.


Subject(s)
Parkinson Disease , Pesticides , Animals , Brain-Gut Axis , Inflammation/chemically induced , Mitochondria , Neuroglia , Parkinson Disease/etiology , Pesticides/toxicity , Rats , Rotenone/toxicity
12.
Environ Health Perspect ; 129(8): 87005, 2021 08.
Article in English | MEDLINE | ID: mdl-34410835

ABSTRACT

BACKGROUND: Chronic environmental exposure to manganese (Mn) can cause debilitating damage to the central nervous system. However, its potential toxic effects on the enteric nervous system (ENS) have yet to be assessed. OBJECTIVE: We examined the effect of Mn on the ENS using both cell and animal models. METHOD: Rat enteric glial cells (EGCs) and mouse primary enteric cultures were exposed to increasing concentrations of Mn and cell viability and mitochondrial health were assessed using various morphological and functional assays. C57BL/6 mice were exposed daily to a sublethal dose of Mn (15mg/kg/d) for 30 d. Gut peristalsis, enteric inflammation, gut microbiome profile, and fecal metabolite composition were assessed at the end of exposure. RESULTS: EGC mitochondria were highly susceptible to Mn neurotoxicity, as evidenced by lower mitochondrial mass, adenosine triphosphate-linked respiration, and aconitase activity as well as higher mitochondrial superoxide, upon Mn exposure. Minor differences were seen in the mouse model: specifically, longer intestinal transit times and higher levels of colonic inflammation. CONCLUSION: Based on our findings from this study, Mn preferentially induced mitochondrial dysfunction in a rat EGC line and in vivo resulted in inflammation in the ENS. https://doi.org/10.1289/EHP7877.


Subject(s)
Enteric Nervous System , Gastrointestinal Microbiome , Animals , Manganese/toxicity , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Rats
13.
Toxicol Sci ; 182(2): 159-167, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34076689

ABSTRACT

Big data approaches have profoundly influenced state-of-the-art in many fields of research, with toxicology being no exception. Here, we use Parkinson's disease as a window through which to explore the challenges of a dual explosion of metabolomic data addressing the myriad environmental exposures individuals experience and genetic analyses implicating many different loci as risk factors for disease. We argue that new experimental approaches are needed to convert the growing body of omics data into molecular mechanisms of disease that can be therapeutically targeted in specific patients. We outline one attractive strategy, which capitalizes on the rapid generation time and advanced molecular tools available in the fruit fly, Drosophila, to provide a platform for mechanistic dissection and drug discovery.


Subject(s)
Parkinson Disease , Animals , Drosophila/genetics , Environmental Exposure , Gene-Environment Interaction , Humans , Parkinson Disease/genetics , Precision Medicine
14.
Mol Neurodegener ; 16(1): 33, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34030727

ABSTRACT

BACKGROUND: Mutations in LRRK2 are the most common cause of familial Parkinson's disease and typically cause disease in the context of abnormal aggregation and deposition of α-synuclein within affected brain tissue. METHODS: We combine genetic analysis of Lrrk-associated toxicity in a penetrant Drosophila model of wild type human α-synuclein neurotoxicity with biochemical analyses and modeling of LRRK2 toxicity in human neurons and transgenic mouse models. RESULTS: We demonstrate that Lrrk and α-synuclein interact to promote neuronal degeneration through convergent effects on the actin cytoskeleton and downstream dysregulation of mitochondrial dynamics and function. We find specifically that monomers and dimers of Lrrk efficiently sever actin and promote normal actin dynamics in vivo. Oligomerization of Lrrk, which is promoted by dominant Parkinson's disease-causing mutations, reduces actin severing activity in vitro and promotes excess stabilization of F-actin in vivo. Importantly, a clinically protective Lrrk mutant reduces oligomerization and α-synuclein neurotoxicity. CONCLUSIONS: Our findings provide a specific mechanistic link between two key molecules in the pathogenesis of Parkinson's disease, α-synuclein and LRRK2, and suggest potential new approaches for therapy development.


Subject(s)
Actins/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/toxicity , Animals , Drosophila , Gene Knock-In Techniques , Humans , Mice , Mice, Inbred C57BL , Neurons/pathology , Parkinson Disease/pathology
15.
Curr Environ Health Rep ; 8(3): 203-211, 2021 09.
Article in English | MEDLINE | ID: mdl-34043217

ABSTRACT

PURPOSE OF REVIEW: Parkinson's disease (PD) is the most prevalent motor disorder and is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the brain. Though the pathology of PD is well established, the cause of this neuronal loss is not well understood. Approximately 90% of PD cases are sporadic, and the environment plays a significant role in disease pathogenesis. The etiology of PD is highly complex, with neuroinflammation being one of the most critical factors implicated in PD. However, the signaling mechanisms underlying neuroinflammation and its interaction with environmental factors are unclear. RECENT FINDINGS: Astroglia and microglia are the two principal cells that play an essential role in maintaining neuronal health in many ways, including through immunological means. Exposure to environmental stressors from various sources affects these glial cells leading to chronic and sustained inflammation. Recent epidemiological studies have identified an interaction among environmental factors and glial genes in Parkinson's disease. Mechanistic studies have shown that exposure to pesticides like rotenone and paraquat, neurotoxic metals like manganese and lead, and even diesel exhaust fumes induce glial activation by regulating various key inflammatory pathways, including the inflammasomes, NOX pathways, and others. This review aims to discuss the recent advances in understanding the mechanism of glial induction in response to environmental stressors and discuss the potential role of gene-environment interaction in driving glial activation.


Subject(s)
Parkinson Disease , Dopaminergic Neurons , Gene-Environment Interaction , Humans , Inflammasomes , Microglia
16.
PLoS Genet ; 17(2): e1009359, 2021 02.
Article in English | MEDLINE | ID: mdl-33556113

ABSTRACT

Vesicular trafficking defects, particularly those in the autophagolysosomal system, have been strongly implicated in the pathogenesis of Parkinson's disease and related α-synucleinopathies. However, mechanisms mediating dysfunction of membrane trafficking remain incompletely understood. Using a Drosophila model of α-synuclein neurotoxicity with widespread and robust pathology, we find that human α-synuclein expression impairs autophagic flux in aging adult neurons. Genetic destabilization of the actin cytoskeleton rescues F-actin accumulation, promotes autophagosome clearance, normalizes the autophagolysosomal system, and rescues neurotoxicity in α-synuclein transgenic animals through an Arp2/3 dependent mechanism. Similarly, mitophagosomes accumulate in human α-synuclein-expressing neurons, and reversal of excessive actin stabilization promotes both clearance of these abnormal mitochondria-containing organelles and rescue of mitochondrial dysfunction. These results suggest that Arp2/3 dependent actin cytoskeleton stabilization mediates autophagic and mitophagic dysfunction and implicate failure of autophagosome maturation as a pathological mechanism in Parkinson's disease and related α-synucleinopathies.


Subject(s)
Actins/metabolism , Autophagosomes/metabolism , Drosophila melanogaster/metabolism , Mitochondria/metabolism , alpha-Synuclein/metabolism , Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/genetics , Actin-Related Protein 2-3 Complex/metabolism , Aging , Animals , Animals, Genetically Modified , Autophagosomes/genetics , Autophagy/genetics , Disease Models, Animal , Drosophila melanogaster/genetics , Humans , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Neurons/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , alpha-Synuclein/genetics
17.
NPJ Parkinsons Dis ; 6(1): 40, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33311497

ABSTRACT

The synaptic protein α-synuclein is linked through genetics and neuropathology to the pathogenesis of Parkinson's disease and related disorders. However, the mechanisms by which α-synuclein influences disease onset and progression are incompletely understood. To identify pathogenic pathways and therapeutic targets we performed proteomic analysis in a highly penetrant new Drosophila model of α-synucleinopathy. We identified 476 significantly upregulated and 563 significantly downregulated proteins in heads from α-synucleinopathy model flies compared to controls. We then used multiple complementary analyses to identify and prioritize genes and pathways within the large set of differentially expressed proteins for functional studies. We performed Gene Ontology enrichment analysis, integrated our proteomic changes with human Parkinson's disease genetic studies, and compared the α-synucleinopathy proteome with that of tauopathy model flies, which are relevant to Alzheimer's disease and related disorders. These approaches identified GTP cyclohydrolase (GCH1) and folate metabolism as candidate mediators of α-synuclein neurotoxicity. In functional validation studies, we found that the knockdown of Drosophila Gch1 enhanced locomotor deficits in α-synuclein transgenic flies, while folate supplementation protected from α-synuclein toxicity. Our integrative analysis suggested that mitochondrial dysfunction was a common downstream mediator of neurodegeneration. Accordingly, Gch1 knockdown enhanced metabolic dysfunction in α-synuclein transgenic fly brains while folate supplementation partially normalized brain bioenergetics. Here we outline and implement an integrative approach to identify and validate potential therapeutic pathways using comparative proteomics and genetics and capitalizing on the facile genetic and pharmacological tools available in Drosophila.

18.
J Clin Invest ; 130(8): 4195-4212, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32597830

ABSTRACT

Characterization of the key cellular targets contributing to sustained microglial activation in neurodegenerative diseases, including Parkinson's disease (PD), and optimal modulation of these targets can provide potential treatments to halt disease progression. Here, we demonstrated that microglial Kv1.3, a voltage-gated potassium channel, was transcriptionally upregulated in response to aggregated α-synuclein (αSynAgg) stimulation in primary microglial cultures and animal models of PD, as well as in postmortem human PD brains. Patch-clamp electrophysiological studies confirmed that the observed Kv1.3 upregulation translated to increased Kv1.3 channel activity. The kinase Fyn, a risk factor for PD, modulated transcriptional upregulation and posttranslational modification of microglial Kv1.3. Multiple state-of-the-art analyses, including Duolink proximity ligation assay imaging, revealed that Fyn directly bound to Kv1.3 and posttranslationally modified its channel activity. Furthermore, we demonstrated the functional relevance of Kv1.3 in augmenting the neuroinflammatory response by using Kv1.3-KO primary microglia and the Kv1.3-specific small-molecule inhibitor PAP-1, thus highlighting the importance of Kv1.3 in neuroinflammation. Administration of PAP-1 significantly inhibited neurodegeneration and neuroinflammation in multiple animal models of PD. Collectively, our results imply that Fyn-dependent regulation of Kv1.3 channels plays an obligatory role in accentuating the neuroinflammatory response in PD and identify Kv1.3 as a potential therapeutic target for PD.


Subject(s)
Kv1.3 Potassium Channel/metabolism , Microglia/metabolism , Parkinson Disease/metabolism , Protein Processing, Post-Translational , Animals , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Mice , Mice, Knockout , Microglia/pathology , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/pathology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
19.
Front Immunol ; 11: 33, 2020.
Article in English | MEDLINE | ID: mdl-32082315

ABSTRACT

Alpha-synuclein (αSynAgg) are pathological hallmarks of Parkinson's disease (PD) and other synucleinopathies that induce microglial activation and immune-mediated neurotoxicity, but the molecular mechanisms of αSynAgg-induced immune activation are poorly defined. We performed quantitative proteomics by mass spectrometry coupled with PCR, immunohistochemical and functional validations studies to define the molecular characteristics of alpha synuclein mediated microglial activation. In mouse microglia, αSynAgg induced robust pro-inflammatory activation (increased expression of 864 genes including Irg1, Ifit1, and Pyhin) and increased nuclear proteins involved in RNA synthesis, splicing, and anti-viral defense mechanisms. Conversely, αSynAgg decreased expression several proteins (including Cdc123, Sod1, and Grn), which were predominantly cytosolic and involved in metabolic, proteasomal and lysosomal mechanisms. Pathway analyses and confirmatory in vitro studies suggested that αSynAgg partly mediates its effects via Stat3 activation. As predicted by our proteomic findings, we verified that αSynAgg induces mitochondrial dysfunction in microglia. Twenty-six proteins differentially expressed by αSynAgg were also identified as PD risk genes in genome-wide association studies (upregulated: Brd2, Clk1, Siglec1; down-regulated: Memo1, Arhgap18, Fyn, and Pgrn/Grn). We validated progranulin (PGRN) as a lysosomal PD-associated protein that is downregulated by αSynAgg in microglia in-vivo and is expressed by microglia in post-mortem PD brain, congruent with our in vitro findings. Conclusion: Together, proteomics approach both reveals novel molecular insights into αSyn-mediated neuroinflammation in PD and other synucleinopathies.


Subject(s)
Microglia/drug effects , Microglia/metabolism , Progranulins/metabolism , Protein Aggregates , Proteome , alpha-Synuclein/pharmacology , Animals , Brain/metabolism , Cell Line , Down-Regulation/drug effects , Genome-Wide Association Study , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/pathology , Progranulins/immunology , Proteomics/methods , Recombinant Proteins/pharmacology
20.
Neurotoxicology ; 75: 186-199, 2019 12.
Article in English | MEDLINE | ID: mdl-31505196

ABSTRACT

Gastrointestinal (GI) disturbances are one of the earliest symptoms affecting most patients with Parkinson's disease (PD). In many cases, these symptoms are observed years before motor impairments become apparent. Hence, the molecular and cellular underpinnings that contribute to this early GI dysfunction in PD have actively been explored using a relevant animal model. The MitoPark model is a chronic, progressive mouse model recapitulating several key pathophysiological aspects of PD. However, GI dysfunction and gut microbiome changes have not been categorized in this model. Herein, we show that decreased GI motility was one of the first non-motor symptoms to develop, evident as early as 8 weeks with significantly different transit times from 12 weeks onwards. These symptoms were observed well before motor symptoms developed, thereby paralleling PD progression in humans. At age 24 weeks, we observed increased colon transit time and reduced fecal water content, indicative of constipation. Intestinal inflammation was evidenced with increased expression of iNOS and TNFα in the small and large intestine. Specifically, iNOS was observed mainly in the enteric plexi, indicating enteric glial cell activation. A pronounced loss of tyrosine hydroxylase-positive neurons occurred at 24 weeks both in the mid-brain region as well as the gut, leading to a corresponding decrease in dopamine (DA) production. We also observed decreased DARPP-32 expression in the colon, validating the loss of DAergic neurons in the gut. However, the total number of enteric neurons did not significantly differ between the two groups. Metabolomic gas chromatography-mass spectrometry analysis of fecal samples showed increased sterol, glycerol, and tocopherol production in MitoPark mice compared to age-matched littermate controls at 20 weeks of age while 16 s microbiome sequencing showed a transient temporal increase in the genus Prevotella. Altogether, the data shed more light on the role of the gut dopaminergic system in maintaining intestinal health. Importantly, this model recapitulates the chronology and development of GI dysfunction along with other non-motor symptoms and can become an attractive translational animal model for pre-clinical assessment of the efficacy of new anti-Parkinsonian drugs that can alleviate GI dysfunction in PD.


Subject(s)
Gastrointestinal Diseases/complications , Gastrointestinal Microbiome , Parkinsonian Disorders/complications , Animals , Blotting, Western , Chromatography, High Pressure Liquid , Colon/chemistry , Disease Models, Animal , Gastric Emptying , Gastrointestinal Diseases/microbiology , Gastrointestinal Transit , Mice, Inbred C57BL , Mice, Transgenic , Neurotransmitter Agents/analysis , Neurotransmitter Agents/metabolism , Parkinsonian Disorders/microbiology , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL