Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Mol Cell ; 81(17): 3481-3495.e7, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34358446

ABSTRACT

PRMT5 is an essential arginine methyltransferase and a therapeutic target in MTAP-null cancers. PRMT5 uses adaptor proteins for substrate recruitment through a previously undefined mechanism. Here, we identify an evolutionarily conserved peptide sequence shared among the three known substrate adaptors (CLNS1A, RIOK1, and COPR5) and show that it is necessary and sufficient for interaction with PRMT5. We demonstrate that PRMT5 uses modular adaptor proteins containing a common binding motif for substrate recruitment, comparable with other enzyme classes such as kinases and E3 ligases. We structurally resolve the interface with PRMT5 and show via genetic perturbation that it is required for methylation of adaptor-recruited substrates including the spliceosome, histones, and ribosomal complexes. Furthermore, disruption of this site affects Sm spliceosome activity, leading to intron retention. Genetic disruption of the PRMT5-substrate adaptor interface impairs growth of MTAP-null tumor cells and is thus a site for development of therapeutic inhibitors of PRMT5.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/physiology , Animals , Cell Line, Tumor , Cytoplasm/metabolism , Female , HCT116 Cells , HEK293 Cells , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ion Channels/metabolism , Male , Methylation , Mice , Mice, Nude , Nuclear Proteins/metabolism , Peptides/genetics , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Spliceosomes/metabolism
2.
Mol Cell ; 73(5): 1001-1014.e8, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30527540

ABSTRACT

In Parkinson's disease (PD), α-synuclein (αS) pathologically impacts the brain, a highly lipid-rich organ. We investigated how alterations in αS or lipid/fatty acid homeostasis affect each other. Lipidomic profiling of human αS-expressing yeast revealed increases in oleic acid (OA, 18:1), diglycerides, and triglycerides. These findings were recapitulated in rodent and human neuronal models of αS dyshomeostasis (overexpression; patient-derived triplication or E46K mutation; E46K mice). Preventing lipid droplet formation or augmenting OA increased αS yeast toxicity; suppressing the OA-generating enzyme stearoyl-CoA-desaturase (SCD) was protective. Genetic or pharmacological SCD inhibition ameliorated toxicity in αS-overexpressing rat neurons. In a C. elegans model, SCD knockout prevented αS-induced dopaminergic degeneration. Conversely, we observed detrimental effects of OA on αS homeostasis: in human neural cells, excess OA caused αS inclusion formation, which was reversed by SCD inhibition. Thus, monounsaturated fatty acid metabolism is pivotal for αS-induced neurotoxicity, and inhibiting SCD represents a novel PD therapeutic approach.


Subject(s)
Antiparkinson Agents/pharmacology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Lipid Metabolism/drug effects , Metabolomics/methods , Neurons/drug effects , Parkinson Disease/drug therapy , Stearoyl-CoA Desaturase/antagonists & inhibitors , alpha-Synuclein/toxicity , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Cell Line , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Diglycerides/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/enzymology , Induced Pluripotent Stem Cells/pathology , Lipid Droplets/drug effects , Lipid Droplets/enzymology , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Nerve Degeneration , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neurons/enzymology , Neurons/pathology , Oleic Acid/metabolism , Parkinson Disease/enzymology , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats, Sprague-Dawley , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Stearoyl-CoA Desaturase/metabolism , Triglycerides/metabolism , alpha-Synuclein/genetics
3.
FEMS Yeast Res ; 18(3)2018 05 01.
Article in English | MEDLINE | ID: mdl-29546391

ABSTRACT

Tyrosine phosphorylation is a key biochemical signal that controls growth and differentiation in multicellular organisms. Saccharomyces cerevisiae and nearly all other unicellular eukaryotes lack intact phosphotyrosine signaling pathways. However, many of these organisms have primitive phosphotyrosine-binding proteins and tyrosine phosphatases, leading to the assumption that the major barrier for emergence of phosphotyrosine signaling was the negative consequences of promiscuous tyrosine kinase activity. In this work, we reveal that the classic oncogene v-Src, which phosphorylates many dozens of proteins in yeast, is toxic because it disrupts a specific spore wall remodeling pathway. Using genetic selections, we find that expression of a specific cyclic peptide, or overexpression of SMK1, a MAP kinase that controls spore wall assembly, both lead to robust growth despite a continuous high level of phosphotyrosine in the yeast proteome. Thus, minimal genetic manipulations allow yeast to tolerate high levels of phosphotyrosine. These results indicate that the introduction of tyrosine kinases within single-celled organisms may not have been a major obstacle to the evolution of phosphotyrosine signaling.


Subject(s)
Genes, src , Phosphotyrosine/metabolism , Saccharomyces cerevisiae/genetics , Mitogen-Activated Protein Kinases/genetics , Peptides, Cyclic/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Tyrosine/metabolism
4.
Cell Rep ; 33(1): 108224, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33027662

ABSTRACT

The ε4 allele of apolipoprotein E (APOE4) is a genetic risk factor for many diseases, including late-onset Alzheimer's disease (AD). We investigate the cellular consequences of APOE4 in human iPSC-derived astrocytes, observing an endocytic defect in APOE4 astrocytes compared with their isogenic APOE3 counterparts. Given the evolutionarily conserved nature of endocytosis, we built a yeast model to identify genetic modifiers of the endocytic defect associated with APOE4. In yeast, only the expression of APOE4 results in dose-dependent defects in both endocytosis and growth. We discover that increasing expression of the early endocytic adaptor protein Yap1802p, a homolog of the human AD risk factor PICALM, rescues the APOE4-induced endocytic defect. In iPSC-derived human astrocytes, increasing expression of PICALM similarly reverses endocytic disruptions. Our work identifies a functional interaction between two AD genetic risk factors-APOE4 and PICALM-centered on the conserved biological process of endocytosis.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein E4/metabolism , Endocytosis/physiology , Alzheimer Disease/pathology , Humans , Risk Factors
5.
Cell Syst ; 4(2): 242-250.e4, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28131823

ABSTRACT

Synucleinopathies, including Parkinson's disease (PD), are associated with the misfolding and mistrafficking of alpha-synuclein (α-syn). Here, using an ascorbate peroxidase (APEX)-based labeling method combined with mass spectrometry, we defined a network of proteins in the immediate vicinity of α-syn in living neurons to shed light on α-syn function. This approach identified 225 proteins, including synaptic proteins, proteins involved in endocytic vesicle trafficking, the retromer complex, phosphatases and mRNA binding proteins. Many were in complexes with α-syn, and some were encoded by genes known to be risk factors for PD and other neurodegenerative diseases. Endocytic trafficking and mRNA translation proteins within this spatial α-syn map overlapped with genetic modifiers of α-syn toxicity, developed in an accompanying study (Khurana et al., this issue of Cell Systems). Our data suggest that perturbation of these particular pathways is directly related to the spatial localization of α-syn within the cell. These approaches provide new avenues to systematically examine protein function and pathology in living cells.


Subject(s)
Ascorbate Peroxidases/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , alpha-Synuclein/metabolism , Animals , Ascorbate Peroxidases/chemistry , Cells, Cultured , HEK293 Cells , Humans , Hydrogen Peroxide/chemistry , Mass Spectrometry , Neurons/cytology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Transport , Rats , alpha-Synuclein/chemistry
6.
Cell Syst ; 4(2): 157-170.e14, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28131822

ABSTRACT

Numerous genes and molecular pathways are implicated in neurodegenerative proteinopathies, but their inter-relationships are poorly understood. We systematically mapped molecular pathways underlying the toxicity of alpha-synuclein (α-syn), a protein central to Parkinson's disease. Genome-wide screens in yeast identified 332 genes that impact α-syn toxicity. To "humanize" this molecular network, we developed a computational method, TransposeNet. This integrates a Steiner prize-collecting approach with homology assignment through sequence, structure, and interaction topology. TransposeNet linked α-syn to multiple parkinsonism genes and druggable targets through perturbed protein trafficking and ER quality control as well as mRNA metabolism and translation. A calcium signaling hub linked these processes to perturbed mitochondrial quality control and function, metal ion transport, transcriptional regulation, and signal transduction. Parkinsonism gene interaction profiles spatially opposed in the network (ATP13A2/PARK9 and VPS35/PARK17) were highly distinct, and network relationships for specific genes (LRRK2/PARK8, ATXN2, and EIF4G1/PARK18) were confirmed in patient induced pluripotent stem cell (iPSC)-derived neurons. This cross-species platform connected diverse neurodegenerative genes to proteinopathy through specific mechanisms and may facilitate patient stratification for targeted therapy.


Subject(s)
Neurodegenerative Diseases/pathology , alpha-Synuclein/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Ataxin-2/chemistry , Ataxin-2/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Susceptibility , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Gene Regulatory Networks/genetics , Genome, Fungal , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neurodegenerative Diseases/genetics , Neurons/cytology , Neurons/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , alpha-Synuclein/genetics
7.
Biomol Eng ; 21(2): 67-72, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15113560

ABSTRACT

We engineered green fluorescent protein (GFP) into affinity fluorescent proteins (aFPs) biosensors. The aFPs detect protein-protein interactions by enhanced fluorescence intensity. In a proof of principle demonstration, aFPs containing haemagglutinin (HA) tag bind specifically to the anti-HA antibody. The sensitivity and specificity is enhanced 28-fold by incorporation of aFPs into solid-phase surface.


Subject(s)
Antibodies, Monoclonal/chemistry , Biosensing Techniques , Green Fluorescent Proteins/chemistry , Hemagglutinins/chemistry , Antibodies, Monoclonal/metabolism , Binding Sites/genetics , Fluorescence , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hemagglutinins/genetics , Hemagglutinins/metabolism , Protein Binding/genetics , Protein Engineering , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity , Spectrometry, Fluorescence , Surface Properties
8.
Science ; 342(6161): 983-7, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24158904

ABSTRACT

The induced pluripotent stem (iPS) cell field holds promise for in vitro disease modeling. However, identifying innate cellular pathologies, particularly for age-related neurodegenerative diseases, has been challenging. Here, we exploited mutation correction of iPS cells and conserved proteotoxic mechanisms from yeast to humans to discover and reverse phenotypic responses to α-synuclein (αsyn), a key protein involved in Parkinson's disease (PD). We generated cortical neurons from iPS cells of patients harboring αsyn mutations, who are at high risk of developing PD dementia. Genetic modifiers from unbiased screens in a yeast model of αsyn toxicity led to identification of early pathogenic phenotypes in patient neurons. These included nitrosative stress, accumulation of endoplasmic reticulum (ER)-associated degradation substrates, and ER stress. A small molecule identified in a yeast screen (NAB2), and the ubiquitin ligase Nedd4 it affects, reversed pathologic phenotypes in these neurons.


Subject(s)
Benzimidazoles/pharmacology , Neurons/drug effects , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Benzimidazoles/chemistry , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation , Neurogenesis , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Rats , alpha-Synuclein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL