Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 354
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(11): 1943-1959.e21, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35545089

ABSTRACT

Parthanatos-associated apoptosis-inducing factor (AIF) nuclease (PAAN), also known as macrophage migration inhibitor factor (MIF), is a member of the PD-D/E(X)K nucleases that acts as a final executioner in parthanatos. PAAN's role in Parkinson's disease (PD) and whether it is amenable to chemical inhibition is not known. Here, we show that neurodegeneration induced by pathologic α-synuclein (α-syn) occurs via PAAN/MIF nuclease activity. Genetic depletion of PAAN/MIF and a mutant lacking nuclease activity prevent the loss of dopaminergic neurons and behavioral deficits in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Compound screening led to the identification of PAANIB-1, a brain-penetrant PAAN/MIF nuclease inhibitor that prevents neurodegeneration induced by α-syn PFF, AAV-α-syn overexpression, or MPTP intoxication in vivo. Our findings could have broad relevance in human pathologies where parthanatos plays a role in the development of cell death inhibitors targeting the druggable PAAN/MIF nuclease.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Parkinson Disease , Animals , Brain/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Endonucleases/metabolism , Mice , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism
2.
Cell ; 157(2): 472-485, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725412

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinson's disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.


Subject(s)
Neurons/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Proteins/metabolism , Amino Acid Sequence , Animals , Drosophila melanogaster , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Molecular Sequence Data , Neurons/pathology , Parkinson Disease/pathology , Ribosomal Proteins/chemistry
3.
Am J Hum Genet ; 110(3): 499-515, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36724785

ABSTRACT

Telomere maintenance 2 (TELO2), Tel2 interacting protein 2 (TTI2), and Tel2 interacting protein 1 (TTI1) are the three components of the conserved Triple T (TTT) complex that modulates activity of phosphatidylinositol 3-kinase-related protein kinases (PIKKs), including mTOR, ATM, and ATR, by regulating the assembly of mTOR complex 1 (mTORC1). The TTT complex is essential for the expression, maturation, and stability of ATM and ATR in response to DNA damage. TELO2- and TTI2-related bi-allelic autosomal-recessive (AR) encephalopathies have been described in individuals with moderate to severe intellectual disability (ID), short stature, postnatal microcephaly, and a movement disorder (in the case of variants within TELO2). We present clinical, genomic, and functional data from 11 individuals in 9 unrelated families with bi-allelic variants in TTI1. All present with ID, and most with microcephaly, short stature, and a movement disorder. Functional studies performed in HEK293T cell lines and fibroblasts and lymphoblastoid cells derived from 4 unrelated individuals showed impairment of the TTT complex and of mTOR pathway activity which is improved by treatment with Rapamycin. Our data delineate a TTI1-related neurodevelopmental disorder and expand the group of disorders related to the TTT complex.


Subject(s)
Microcephaly , Movement Disorders , Neurodevelopmental Disorders , Humans , Intracellular Signaling Peptides and Proteins , HEK293 Cells , TOR Serine-Threonine Kinases
4.
Nat Chem Biol ; 20(7): 894-905, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38658655

ABSTRACT

Calcium ions serve as key intracellular signals. Local, transient increases in calcium concentrations can activate calcium sensor proteins that in turn trigger downstream effectors. In neurons, calcium transients play a central role in regulating neurotransmitter release and synaptic plasticity. However, it is challenging to capture the molecular events associated with these localized and ephemeral calcium signals. Here we present an engineered biotin ligase that generates permanent molecular traces in a calcium-dependent manner. The enzyme, calcium-dependent BioID (Cal-ID), biotinylates nearby proteins within minutes in response to elevated local calcium levels. The biotinylated proteins can be identified via mass spectrometry and visualized using microscopy. In neurons, Cal-ID labeling is triggered by neuronal activity, leading to prominent protein biotinylation that enables transcription-independent activity labeling in the brain. In summary, Cal-ID produces a biochemical record of calcium signals and neuronal activity with high spatial resolution and molecular specificity.


Subject(s)
Biotinylation , Calcium Signaling , Calcium , Neurons , Calcium/metabolism , Neurons/metabolism , Animals , Carbon-Nitrogen Ligases/metabolism , Carbon-Nitrogen Ligases/chemistry , Humans , Mice , HEK293 Cells , Repressor Proteins , Escherichia coli Proteins
5.
Cell ; 144(5): 689-702, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376232

ABSTRACT

A hallmark of Parkinson's disease (PD) is the preferential loss of substantia nigra dopamine neurons. Here, we identify a new parkin interacting substrate, PARIS (ZNF746), whose levels are regulated by the ubiquitin proteasome system via binding to and ubiquitination by the E3 ubiquitin ligase, parkin. PARIS is a KRAB and zinc finger protein that accumulates in models of parkin inactivation and in human PD brain. PARIS represses the expression of the transcriptional coactivator, PGC-1α and the PGC-1α target gene, NRF-1 by binding to insulin response sequences in the PGC-1α promoter. Conditional knockout of parkin in adult animals leads to progressive loss of dopamine (DA) neurons in a PARIS-dependent manner. Moreover, overexpression of PARIS leads to the selective loss of DA neurons in the substantia nigra, and this is reversed by either parkin or PGC-1α coexpression. The identification of PARIS provides a molecular mechanism for neurodegeneration due to parkin inactivation.


Subject(s)
Parkinson Disease/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Brain/metabolism , Brain/pathology , Dopamine/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 1/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Nuclear Respiratory Factor 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Transcription Factors , Ubiquitin-Protein Ligases/metabolism
6.
Cell ; 145(2): 284-99, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21496646

ABSTRACT

The synaptic insertion or removal of AMPA receptors (AMPAR) plays critical roles in the regulation of synaptic activity reflected in the expression of long-term potentiation (LTP) and long-term depression (LTD). The cellular events underlying this important process in learning and memory are still being revealed. Here we describe and characterize the AAA+ ATPase Thorase, which regulates the expression of surface AMPAR. In an ATPase-dependent manner Thorase mediates the internalization of AMPAR by disassembling the AMPAR-GRIP1 complex. Following genetic deletion of Thorase, the internalization of AMPAR is substantially reduced, leading to increased amplitudes of miniature excitatory postsynaptic currents, enhancement of LTP, and elimination of LTD. These molecular events are expressed as deficits in learning and memory in Thorase null mice. This study identifies an AAA+ ATPase that plays a critical role in regulating the surface expression of AMPAR and thereby regulates synaptic plasticity and learning and memory.


Subject(s)
Adenosine Triphosphatases/metabolism , Neuronal Plasticity , Receptors, AMPA/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/chemistry , Amino Acid Sequence , Animals , Brain/metabolism , Cells, Cultured , Female , Gene Expression Profiling , Humans , Learning , Male , Memory , Mice , Molecular Sequence Data , Rats , Sequence Alignment , Synapses
7.
PLoS Genet ; 19(1): e1010558, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36626371

ABSTRACT

Copper (Cu) has a multifaceted role in brain development, function, and metabolism. Two homologous Cu transporters, Atp7a (Menkes disease protein) and Atp7b (Wilson disease protein), maintain Cu homeostasis in the tissue. Atp7a mediates Cu entry into the brain and activates Cu-dependent enzymes, whereas the role of Atp7b is less clear. We show that during postnatal development Atp7b is necessary for normal morphology and function of choroid plexus (ChPl). Inactivation of Atp7b causes reorganization of ChPl' cytoskeleton and cell-cell contacts, loss of Slc31a1 from the apical membrane, and a decrease in the length and number of microvilli and cilia. In ChPl lacking Atp7b, Atp7a is upregulated but remains intracellular, which limits Cu transport into the brain and results in significant Cu deficit, which is reversed only in older animals. Cu deficiency is associated with down-regulation of Atp7a in locus coeruleus and catecholamine imbalance, despite normal expression of dopamine-ß-hydroxylase. In addition, there are notable changes in the brain lipidome, which can be attributed to inhibition of diacylglyceride-to-phosphatidylethanolamine conversion. These results identify the new role for Atp7b in developing brain and identify metabolic changes that could be exacerbated by Cu chelation therapy.


Subject(s)
Copper , Menkes Kinky Hair Syndrome , Mice , Animals , Copper-Transporting ATPases , Copper/metabolism , Choroid Plexus/metabolism , Menkes Kinky Hair Syndrome/metabolism , Brain/metabolism
8.
FASEB J ; 38(6): e23556, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38498348

ABSTRACT

PARP-1 over-activation results in cell death via excessive PAR generation in different cell types, including neurons following brain ischemia. Glycolysis, mitochondrial function, and redox balance are key cellular processes altered in brain ischemia. Studies show that PAR generated after PARP-1 over-activation can bind hexokinase-1 (HK-1) and result in glycolytic defects and subsequent mitochondrial dysfunction. HK-1 is the neuronal hexokinase and catalyzes the first reaction of glycolysis, converting glucose to glucose-6-phosphate (G6P), a common substrate for glycolysis, and the pentose phosphate pathway (PPP). PPP is critical in maintaining NADPH and GSH levels via G6P dehydrogenase activity. Therefore, defects in HK-1 will not only decrease cellular bioenergetics but will also cause redox imbalance due to the depletion of GSH. In brain ischemia, whether PAR-mediated inhibition of HK-1 results in bioenergetics defects and redox imbalance is not known. We used oxygen-glucose deprivation (OGD) in mouse cortical neurons to mimic brain ischemia in neuronal cultures and observed that PARP-1 activation via PAR formation alters glycolysis, mitochondrial function, and redox homeostasis in neurons. We used pharmacological inhibition of PARP-1 and adenoviral-mediated overexpression of wild-type HK-1 (wtHK-1) and PAR-binding mutant HK-1 (pbmHK-1). Our data show that PAR inhibition or overexpression of HK-1 significantly improves glycolysis, mitochondrial function, redox homeostasis, and cell survival in mouse cortical neurons exposed to OGD. These results suggest that PAR binding and inhibition of HK-1 during OGD drive bioenergetic defects in neurons due to inhibition of glycolysis and impairment of mitochondrial function.


Subject(s)
Brain Ischemia , Oxygen , Mice , Animals , Oxygen/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Hexokinase/genetics , Hexokinase/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/metabolism , Glucose/metabolism , Brain Ischemia/metabolism , Glycolysis , Neurons/metabolism , Oxidation-Reduction
9.
EMBO Rep ; 24(11): e56166, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37870275

ABSTRACT

ZNF746 was identified as parkin-interacting substrate (PARIS). Investigating its pathophysiological properties, we find that PARIS undergoes liquid-liquid phase separation (LLPS) and amorphous solid formation. The N-terminal low complexity domain 1 (LCD1) of PARIS is required for LLPS, whereas the C-terminal prion-like domain (PrLD) drives the transition from liquid to solid phase. In addition, we observe that poly(ADP-ribose) (PAR) strongly binds to the C-terminus of PARIS near the PrLD, accelerating its LLPS and solidification. N-Methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced PAR formation leads to PARIS oligomerization in human iPSC-derived dopaminergic neurons that is prevented by the PARP inhibitor, ABT-888. Furthermore, SDS-resistant PARIS species are observed in the substantia nigra (SN) of aged mice overexpressing wild-type PARIS, but not with a PAR binding-deficient PARIS mutant. PARIS solidification is also found in the SN of mice injected with preformed fibrils of α-synuclein (α-syn PFF) and adult mice with a conditional knockout (KO) of parkin, but not if α-syn PFF is injected into mice deficient for PARP1. Herein, we demonstrate that PARIS undergoes LLPS and PAR-mediated solidification in models of Parkinson's disease.


Subject(s)
Parkinson Disease , Poly Adenosine Diphosphate Ribose , Animals , Humans , Mice , Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
10.
Mol Cell Proteomics ; 22(1): 100452, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36423813

ABSTRACT

Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra (SN) of the brain. Despite decades of studies, the precise pathogenic mechanism of PD is still elusive. An unbiased proteomic analysis of PD patient's brain allows the identification of critical proteins and molecular pathways that lead to dopamine cell death and α-synuclein deposition and the resulting devastating clinical symptoms. In this study, we conducted an in-depth proteome analysis of human SN tissues from 15 PD patients and 15 healthy control individuals combining Orbitrap mass spectrometry with the isobaric tandem mass tag-based multiplexing technology. We identified 10,040 proteins with 1140 differentially expressed proteins in the SN of PD patients. Pathway analysis showed that the ribosome pathway was the most enriched one, followed by gamma-aminobutyric acidergic synapse, retrograde endocannabinoid signaling, cell adhesion molecules, morphine addiction, Prion disease, and PD pathways. Strikingly, the majority of the proteins enriched in the ribosome pathway were mitochondrial ribosomal proteins (mitoribosomes). The subsequent protein-protein interaction analysis and the weighted gene coexpression network analysis confirmed that the mitoribosome is the most enriched protein cluster. Furthermore, the mitoribosome was also identified in our analysis of a replication set of ten PD and nine healthy control SN tissues. This study provides potential disease pathways involved in PD and paves the way to study further the pathogenic mechanism of PD.


Subject(s)
Neurodegenerative Diseases , Parkinson Disease , Humans , Parkinson Disease/metabolism , Proteomics/methods , Substantia Nigra/metabolism , Brain/metabolism , Neurodegenerative Diseases/metabolism
11.
Proc Natl Acad Sci U S A ; 119(15): e2118819119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394877

ABSTRACT

In idiopathic Parkinson's disease (PD), pathologic αSyn aggregates drive oxidative and nitrative stress that may cause genomic and mitochondrial DNA damage. These events are associated with activation of the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) immune pathway, but it is not known whether STING is activated in or contributes to α-synucleinopathies. Herein, we used primary cell cultures and the intrastriatal αSyn preformed fibril (αSyn-PFF) mouse model of PD to demonstrate that αSyn pathology causes STING-dependent neuroinflammation and dopaminergic neurodegeneration. In microglia-astrocyte cultures, αSyn-PFFs induced DNA double-strand break (DSB) damage response signaling (γH2A.X), as well as TBK1 activation that was blocked by STING inhibition. In the αSyn-PFF mouse model, we similarly observed TBK1 activation and increased γH2A.X within striatal microglia prior to the onset of dopaminergic neurodegeneration. Using STING-deficient (Stinggt) mice, we demonstrated that striatal interferon activation in the α-Syn PFF model is STING-dependent. Furthermore, Stinggt mice were protected from α-Syn PFF-induced motor deficits, pathologic αSyn accumulation, and dopaminergic neuron loss. We also observed upregulation of STING protein in the substantia nigra pars compacta (SNpc) of human PD patients that correlated significantly with pathologic αSyn accumulation. STING was similarly upregulated in microglia cultures treated with αSyn-PFFs, which primed the pathway to mount stronger interferon responses when exposed to a STING agonist. Our results suggest that microglial STING activation contributes to both the neuroinflammation and neurodegeneration arising from α-synucleinopathies, including PD.


Subject(s)
Interferon Type I , Membrane Proteins , Parkinson Disease , Synucleinopathies , Animals , Dopaminergic Neurons , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neurodegenerative Diseases , Neuroinflammatory Diseases , Nucleotidyltransferases/metabolism , Signal Transduction , Synucleinopathies/genetics
12.
Proc Natl Acad Sci U S A ; 119(16): e2200545119, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35412917

ABSTRACT

Cocaine exerts its stimulant effect by inhibiting dopamine (DA) reuptake, leading to increased dopamine signaling. This action is thought to reflect the binding of cocaine to the dopamine transporter (DAT) to inhibit its function. However, cocaine is a relatively weak inhibitor of DAT, and many DAT inhibitors do not share cocaine's behavioral actions. Further, recent reports show more potent actions of the drug, implying the existence of a high-affinity receptor for cocaine. We now report high-affinity binding of cocaine associated with the brain acid soluble protein 1 (BASP1) with a dissociation constant (Kd) of 7 nM. Knocking down BASP1 in the striatum inhibits [3H]cocaine binding to striatal synaptosomes. Depleting BASP1 in the nucleus accumbens but not the dorsal striatum diminishes locomotor stimulation in mice. Our findings imply that BASP1 is a pharmacologically relevant receptor for cocaine.


Subject(s)
Calmodulin-Binding Proteins , Carrier Proteins , Cocaine , Cytoskeletal Proteins , Nerve Tissue Proteins , Receptors, Drug , Animals , Binding Sites , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cocaine/metabolism , Cocaine/pharmacology , Corpus Striatum/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Gene Knock-In Techniques , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Receptors, Drug/genetics , Receptors, Drug/metabolism
13.
Proc Natl Acad Sci U S A ; 119(36): e2204835119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36044549

ABSTRACT

Physical activity provides clinical benefit in Parkinson's disease (PD). Irisin is an exercise-induced polypeptide secreted by skeletal muscle that crosses the blood-brain barrier and mediates certain effects of exercise. Here, we show that irisin prevents pathologic α-synuclein (α-syn)-induced neurodegeneration in the α-syn preformed fibril (PFF) mouse model of sporadic PD. Intravenous delivery of irisin via viral vectors following the stereotaxic intrastriatal injection of α-syn PFF cause a reduction in the formation of pathologic α-syn and prevented the loss of dopamine neurons and lowering of striatal dopamine. Irisin also substantially reduced the α-syn PFF-induced motor deficits as assessed behaviorally by the pole and grip strength test. Recombinant sustained irisin treatment of primary cortical neurons attenuated α-syn PFF toxicity by reducing the formation of phosphorylated serine 129 of α-syn and neuronal cell death. Tandem mass spectrometry and biochemical analysis revealed that irisin reduced pathologic α-syn by enhancing endolysosomal degradation of pathologic α-syn. Our findings highlight the potential for therapeutic disease modification of irisin in PD.


Subject(s)
Corpus Striatum , Fibronectins , Parkinson Disease , alpha-Synuclein , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Fibronectins/administration & dosage , Fibronectins/genetics , Fibronectins/metabolism , Mice , Parkinson Disease/metabolism , Parkinson Disease/therapy , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
14.
Mov Disord ; 39(4): 644-650, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38396375

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative condition that pathognomonically involves the death of dopaminergic neurons in the substantia nigra pars compacta, resulting in a myriad of motor and non-motor symptoms. Given the insurmountable burden of this disease on the population and healthcare system, significant efforts have been put forth toward generating disease modifying therapies. This class of treatments characteristically alters disease course, as opposed to current strategies that focus on managing symptoms. Previous literature has implicated the cell death pathway known as parthanatos in PD progression. Inhibition of this pathway by targeting poly (ADP)-ribose polymerase 1 (PARP1) prevents neurodegeneration in a model of idiopathic PD. However, PARP1 has a vast repertoire of functions within the body, increasing the probability of side effects with the long-term treatment likely necessary for clinically significant neuroprotection. Recent work culminated in the development of a novel agent targeting the macrophage migration inhibitory factor (MIF) nuclease domain, also named parthanatos-associated apoptosis-inducing factor nuclease (PAAN). This nuclease activity specifically executes the terminal step in parthanatos. Parthanatos-associated apoptosis-inducing factor nuclease inhibitor-1 was neuroprotective in multiple preclinical mouse models of PD. This piece will focus on contextualizing this discovery, emphasizing its significance, and discussing its potential implications for parthanatos-directed treatment. © 2024 International Parkinson and Movement Disorder Society.


Subject(s)
Dopaminergic Neurons , Macrophage Migration-Inhibitory Factors , Parkinson Disease , Humans , Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Animals , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Parthanatos/drug effects
15.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Article in English | MEDLINE | ID: mdl-34172566

ABSTRACT

The spread of pathological α-synuclein (α-syn) is a crucial event in the progression of Parkinson's disease (PD). Cell surface receptors such as lymphocyte activation gene 3 (LAG3) and amyloid precursor-like protein 1 (APLP1) can preferentially bind α-syn in the amyloid over monomeric state to initiate cell-to-cell transmission. However, the molecular mechanism underlying this selective binding is unknown. Here, we perform an array of biophysical experiments and reveal that LAG3 D1 and APLP1 E1 domains commonly use an alkaline surface to bind the acidic C terminus, especially residues 118 to 140, of α-syn. The formation of amyloid fibrils not only can disrupt the intramolecular interactions between the C terminus and the amyloid-forming core of α-syn but can also condense the C terminus on fibril surface, which remarkably increase the binding affinity of α-syn to the receptors. Based on this mechanism, we find that phosphorylation at serine 129 (pS129), a hallmark modification of pathological α-syn, can further enhance the interaction between α-syn fibrils and the receptors. This finding is further confirmed by the higher efficiency of pS129 fibrils in cellular internalization, seeding, and inducing PD-like α-syn pathology in transgenic mice. Our work illuminates the mechanistic understanding on the spread of pathological α-syn and provides structural information for therapeutic targeting on the interaction of α-syn fibrils and receptors as a potential treatment for PD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloid/metabolism , Antigens, CD/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Animals , Cell Line, Tumor , Endocytosis , Humans , Mice , Nerve Degeneration/pathology , Neurons/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Static Electricity , alpha-Synuclein/chemistry , alpha-Synuclein/toxicity , Lymphocyte Activation Gene 3 Protein
16.
Pharmacol Rev ; 73(4): 33-97, 2021 10.
Article in English | MEDLINE | ID: mdl-34663684

ABSTRACT

Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.


Subject(s)
Parkinson Disease , Humans , Mutation , Neuroprotection , Parkinson Disease/drug therapy , Parkinson Disease/genetics
17.
Int J Mol Sci ; 25(4)2024 Feb 18.
Article in English | MEDLINE | ID: mdl-38397070

ABSTRACT

In recent years, research into Parkinson's disease and similar neurodegenerative disorders has increasingly suggested that these conditions are synonymous with failures in proteostasis. However, the spotlight of this research has remained firmly focused on the tail end of proteostasis, primarily aggregation, misfolding, and degradation, with protein translation being comparatively overlooked. Now, there is an increasing body of evidence supporting a potential role for translation in the pathogenesis of PD, and its dysregulation is already established in other similar neurodegenerative conditions. In this paper, we consider how altered protein translation fits into the broader picture of PD pathogenesis, working hand in hand to compound the stress placed on neurons, until this becomes irrecoverable. We will also consider molecular players of interest, recent evidence that suggests that aggregates may directly influence translation in PD progression, and the implications for the role of protein translation in our development of clinically useful diagnostics and therapeutics.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/etiology , Parkinson Disease/metabolism , Neurons/metabolism , Proteostasis , Protein Biosynthesis , alpha-Synuclein/metabolism
18.
J Neurosci ; 42(49): 9263-9277, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36280265

ABSTRACT

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). In this study, we generated a transgenic model by crossing germline Parkin-/- mice with PolgAD257A mice, an established model of premature aging and mitochondrial stress. We hypothesized that loss of Parkin-/- in PolgAD257A/D257A mice would exacerbate mitochondrial dysfunction, leading to loss of dopamine neurons and nigral-striatal specific neurobehavioral motor dysfunction. We found that aged Parkin-/-/PolgAD257A/D257A male and female mice exhibited severe behavioral deficits, nonspecific to the nigral-striatal pathway, with neither dopaminergic neurodegeneration nor reductions in striatal dopamine. We saw no difference in expression levels of nuclear-encoded subunits of mitochondrial markers and mitochondrial Complex I and IV activities, although we did observe substantial reductions in mitochondrial-encoded COX41I, indicating mitochondrial dysfunction as a result of PolgAD257A/D257A mtDNA mutations. Expression levels of mitophagy markers LC3I/LC3II remained unchanged between cohorts, suggesting no overt mitophagy defects. Expression levels of the parkin substrates, VDAC, NLRP3, and AIMP2 remained unchanged, suggesting no parkin dysfunction. In summary, we were unable to observe dopaminergic neurodegeneration with corresponding nigral-striatal neurobehavioral deficits, nor Parkin or mitochondrial dysfunction in Parkin-/-/PolgAD257A/D257A mice. These findings support a lack of synergism of Parkin loss on mitochondrial dysfunction in mouse models of mitochondrial deficits.SIGNIFICANCE STATEMENT Producing a mouse model of Parkinson's disease (PD) that is etiologically relevant, recapitulates clinical hallmarks, and exhibits reproducible results is crucial to understanding the underlying pathology and in developing disease-modifying therapies. Here, we show that Parkin-/-/PolgAD257A/D257A mice, a previously reported PD mouse model, fails to reproduce a Parkinsonian phenotype. We show that these mice do not display dopaminergic neurodegeneration nor nigral-striatal-dependent motor deficits. Furthermore, we report that Parkin loss does not synergize with mitochondrial dysfunction. Our results demonstrate that Parkin-/-/PolgAD257A/D257A mice are not a reliable model for PD and adds to a growing body of work demonstrating that Parkin loss does not synergize with mitochondrial dysfunction in mouse models of mitochondrial deficits.


Subject(s)
Disease Models, Animal , Dopamine , Mitochondria , Parkinson Disease , Ubiquitin-Protein Ligases , Animals , Female , Male , Mice , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA Polymerase gamma/genetics , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
20.
Nature ; 546(7660): 656-661, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28636593

ABSTRACT

Genetic studies have shown the association of Parkinson's disease with alleles of the major histocompatibility complex. Here we show that a defined set of peptides that are derived from α-synuclein, a protein aggregated in Parkinson's disease, act as antigenic epitopes displayed by these alleles and drive helper and cytotoxic T cell responses in patients with Parkinson's disease. These responses may explain the association of Parkinson's disease with specific major histocompatibility complex alleles.


Subject(s)
Parkinson Disease/immunology , T-Lymphocytes/immunology , alpha-Synuclein/immunology , Aged , Aged, 80 and over , Alleles , Amino Acid Sequence , Autoimmunity , Epitopes, T-Lymphocyte/immunology , Female , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Male , Middle Aged , Parkinson Disease/genetics , Parkinson Disease/pathology , Peptide Fragments/chemistry , Peptide Fragments/immunology , T-Lymphocytes/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , alpha-Synuclein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL