Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 213
Filter
1.
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
2.
Cell ; 173(1): 62-73.e9, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29526462

ABSTRACT

Aggregates of human islet amyloid polypeptide (IAPP) in the pancreas of patients with type 2 diabetes (T2D) are thought to contribute to ß cell dysfunction and death. To understand how IAPP harms cells and how this might be overcome, we created a yeast model of IAPP toxicity. Ste24, an evolutionarily conserved protease that was recently reported to degrade peptides stuck within the translocon between the cytoplasm and the endoplasmic reticulum, was the strongest suppressor of IAPP toxicity. By testing variants of the human homolog, ZMPSTE24, with varying activity levels, the rescue of IAPP toxicity proved to be directly proportional to the declogging efficiency. Clinically relevant ZMPSTE24 variants identified in the largest database of exomes sequences derived from T2D patients were characterized using the yeast model, revealing 14 partial loss-of-function variants, which were enriched among diabetes patients over 2-fold. Thus, clogging of the translocon by IAPP oligomers may contribute to ß cell failure.


Subject(s)
Islet Amyloid Polypeptide/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Endoplasmic Reticulum Stress/drug effects , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/toxicity , Membrane Proteins/chemistry , Membrane Proteins/genetics , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Models, Biological , Mutagenesis , Protein Aggregates/physiology , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Unfolded Protein Response/drug effects
3.
Cell ; 168(5): 856-866.e12, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28215707

ABSTRACT

HSP90 acts as a protein-folding buffer that shapes the manifestations of genetic variation in model organisms. Whether HSP90 influences the consequences of mutations in humans, potentially modifying the clinical course of genetic diseases, remains unknown. By mining data for >1,500 disease-causing mutants, we found a strong correlation between reduced phenotypic severity and a dominant (HSP90 ≥ HSP70) increase in mutant engagement by HSP90. Examining the cancer predisposition syndrome Fanconi anemia in depth revealed that mutant FANCA proteins engaged predominantly by HSP70 had severely compromised function. In contrast, the function of less severe mutants was preserved by a dominant increase in HSP90 binding. Reducing HSP90's buffering capacity with inhibitors or febrile temperatures destabilized HSP90-buffered mutants, exacerbating FA-related chemosensitivities. Strikingly, a compensatory FANCA somatic mutation from an "experiment of nature" in monozygotic twins both prevented anemia and reduced HSP90 binding. These findings provide one plausible mechanism for the variable expressivity and environmental sensitivity of genetic diseases.


Subject(s)
Fanconi Anemia/genetics , Fanconi Anemia/pathology , HSP90 Heat-Shock Proteins/genetics , Protein Folding , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/chemistry , Fanconi Anemia Complementation Group A Protein/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Mutation, Missense , Protein Interaction Domains and Motifs , Stress, Physiological , Twins, Monozygotic
4.
Cell ; 171(4): 966-979.e18, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29056345

ABSTRACT

Protein aggregation is a hallmark of many diseases but also underlies a wide range of positive cellular functions. This phenomenon has been difficult to study because of a lack of quantitative and high-throughput cellular tools. Here, we develop a synthetic genetic tool to sense and control protein aggregation. We apply the technology to yeast prions, developing sensors to track their aggregation states and employing prion fusions to encode synthetic memories in yeast cells. Utilizing high-throughput screens, we identify prion-curing mutants and engineer "anti-prion drives" that reverse the non-Mendelian inheritance pattern of prions and eliminate them from yeast populations. We extend our technology to yeast RNA-binding proteins (RBPs) by tracking their propensity to aggregate, searching for co-occurring aggregates, and uncovering a group of coalescing RBPs through screens enabled by our platform. Our work establishes a quantitative, high-throughput, and generalizable technology to study and control diverse protein aggregation processes in cells.


Subject(s)
Genetic Techniques , Prions/genetics , Genetic Engineering , Genetic Techniques/economics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Synthetic Biology/methods , mRNA Cleavage and Polyadenylation Factors/metabolism
5.
Cell ; 167(2): 369-381.e12, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693355

ABSTRACT

Prions are a paradigm-shifting mechanism of inheritance in which phenotypes are encoded by self-templating protein conformations rather than nucleic acids. Here, we examine the breadth of protein-based inheritance across the yeast proteome by assessing the ability of nearly every open reading frame (ORF; ∼5,300 ORFs) to induce heritable traits. Transient overexpression of nearly 50 proteins created traits that remained heritable long after their expression returned to normal. These traits were beneficial, had prion-like patterns of inheritance, were common in wild yeasts, and could be transmitted to naive cells with protein alone. Most inducing proteins were not known prions and did not form amyloid. Instead, they are highly enriched in nucleic acid binding proteins with large intrinsically disordered domains that have been widely conserved across evolution. Thus, our data establish a common type of protein-based inheritance through which intrinsically disordered proteins can drive the emergence of new traits and adaptive opportunities.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Quantitative Trait, Heritable , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Amyloid/metabolism , Evolution, Molecular , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Open Reading Frames , Prions/chemistry , Prions/metabolism , Proteome , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
6.
Cell ; 163(3): 620-8, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26456111

ABSTRACT

Biological processes occur in complex environments containing a myriad of potential interactors. Unfortunately, limitations on the sensitivity of biophysical techniques normally restrict structural investigations to purified systems, at concentrations that are orders of magnitude above endogenous levels. Dynamic nuclear polarization (DNP) can dramatically enhance the sensitivity of nuclear magnetic resonance (NMR) spectroscopy and enable structural studies in biologically complex environments. Here, we applied DNP NMR to investigate the structure of a protein containing both an environmentally sensitive folding pathway and an intrinsically disordered region, the yeast prion protein Sup35. We added an exogenously prepared isotopically labeled protein to deuterated lysates, rendering the biological environment "invisible" and enabling highly efficient polarization transfer for DNP. In this environment, structural changes occurred in a region known to influence biological activity but intrinsically disordered in purified samples. Thus, DNP makes structural studies of proteins at endogenous levels in biological contexts possible, and such contexts can influence protein structure.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Peptide Termination Factors/chemistry , Prions/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Molecular Sequence Data , Peptide Termination Factors/metabolism , Prions/metabolism , Protein Folding , Protein Structure, Secondary , Saccharomyces cerevisiae Proteins/metabolism
7.
Cell ; 161(3): 647-660, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25910212

ABSTRACT

How disease-associated mutations impair protein activities in the context of biological networks remains mostly undetermined. Although a few renowned alleles are well characterized, functional information is missing for over 100,000 disease-associated variants. Here we functionally profile several thousand missense mutations across a spectrum of Mendelian disorders using various interaction assays. The majority of disease-associated alleles exhibit wild-type chaperone binding profiles, suggesting they preserve protein folding or stability. While common variants from healthy individuals rarely affect interactions, two-thirds of disease-associated alleles perturb protein-protein interactions, with half corresponding to "edgetic" alleles affecting only a subset of interactions while leaving most other interactions unperturbed. With transcription factors, many alleles that leave protein-protein interactions intact affect DNA binding. Different mutations in the same gene leading to different interaction profiles often result in distinct disease phenotypes. Thus disease-associated alleles that perturb distinct protein activities rather than grossly affecting folding and stability are relatively widespread.


Subject(s)
Disease/genetics , Mutation, Missense , Protein Interaction Maps , Proteins/genetics , Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Humans , Open Reading Frames , Protein Folding , Protein Stability
8.
Cell ; 158(5): 1072-1082, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171408

ABSTRACT

[GAR(+)] is a protein-based element of inheritance that allows yeast (Saccharomyces cerevisiae) to circumvent a hallmark of their biology: extreme metabolic specialization for glucose fermentation. When glucose is present, yeast will not use other carbon sources. [GAR(+)] allows cells to circumvent this "glucose repression." [GAR(+)] is induced in yeast by a factor secreted by bacteria inhabiting their environment. We report that de novo rates of [GAR(+)] appearance correlate with the yeast's ecological niche. Evolutionarily distant fungi possess similar epigenetic elements that are also induced by bacteria. As expected for a mechanism whose adaptive value originates from the selective pressures of life in biological communities, the ability of bacteria to induce [GAR(+)] and the ability of yeast to respond to bacterial signals have been extinguished repeatedly during the extended monoculture of domestication. Thus, [GAR(+)] is a broadly conserved adaptive strategy that links environmental and social cues to heritable changes in metabolism.


Subject(s)
Epigenesis, Genetic , Glucose/metabolism , Prions/metabolism , Saccharomyces cerevisiae/metabolism , Ascomycota/genetics , Ascomycota/metabolism , Bacteria/chemistry , Bacteria/genetics , Dekkera/genetics , Dekkera/metabolism , Phenotype , Saccharomyces cerevisiae/genetics
9.
Cell ; 158(2): 434-448, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25036637

ABSTRACT

Chaperones are abundant cellular proteins that promote the folding and function of their substrate proteins (clients). In vivo, chaperones also associate with a large and diverse set of cofactors (cochaperones) that regulate their specificity and function. However, how these cochaperones regulate protein folding and whether they have chaperone-independent biological functions is largely unknown. We combined mass spectrometry and quantitative high-throughput LUMIER assays to systematically characterize the chaperone-cochaperone-client interaction network in human cells. We uncover hundreds of chaperone clients, delineate their participation in specific cochaperone complexes, and establish a surprisingly distinct network of protein-protein interactions for cochaperones. As a salient example of the power of such analysis, we establish that NUDC family cochaperones specifically associate with structurally related but evolutionarily distinct ß-propeller folds. We provide a framework for deciphering the proteostasis network and its regulation in development and disease and expand the use of chaperones as sensors for drug-target engagement.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Protein Interaction Maps , Humans , Protein Folding , Tacrolimus Binding Proteins/metabolism
10.
Cell ; 158(3): 564-78, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083868

ABSTRACT

Stromal cells within the tumor microenvironment are essential for tumor progression and metastasis. Surprisingly little is known about the factors that drive the transcriptional reprogramming of stromal cells within tumors. We report that the transcriptional regulator heat shock factor 1 (HSF1) is frequently activated in cancer-associated fibroblasts (CAFs), where it is a potent enabler of malignancy. HSF1 drives a transcriptional program in CAFs that complements, yet is completely different from, the program it drives in adjacent cancer cells. This CAF program is uniquely structured to support malignancy in a non-cell-autonomous way. Two central stromal signaling molecules-TGF-ß and SDF1-play a critical role. In early-stage breast and lung cancer, high stromal HSF1 activation is strongly associated with poor patient outcome. Thus, tumors co-opt the ancient survival functions of HSF1 to orchestrate malignancy in both cell-autonomous and non-cell-autonomous ways, with far-reaching therapeutic implications.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Chemokine CXCL12/metabolism , Fibroblasts/metabolism , Heat Shock Transcription Factors , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Transforming Growth Factor beta/metabolism
11.
Cell ; 158(5): 1083-1093, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171409

ABSTRACT

In experimental science, organisms are usually studied in isolation, but in the wild, they compete and cooperate in complex communities. We report a system for cross-kingdom communication by which bacteria heritably transform yeast metabolism. An ancient biological circuit blocks yeast from using other carbon sources in the presence of glucose. [GAR(+)], a protein-based epigenetic element, allows yeast to circumvent this "glucose repression" and use multiple carbon sources in the presence of glucose. Some bacteria secrete a chemical factor that induces [GAR(+)]. [GAR(+)] is advantageous to bacteria because yeast cells make less ethanol and is advantageous to yeast because their growth and long-term viability is improved in complex carbon sources. This cross-kingdom communication is broadly conserved, providing a compelling argument for its adaptive value. By heritably transforming growth and survival strategies in response to the selective pressures of life in a biological community, [GAR(+)] presents a unique example of Lamarckian inheritance.


Subject(s)
Epigenesis, Genetic , Prions/metabolism , Saccharomyces cerevisiae/metabolism , Staphylococcus hominis/metabolism , Fermentation , Glucose/metabolism , Saccharomyces cerevisiae/genetics , Staphylococcus hominis/genetics , Wine/microbiology , Yeasts/genetics , Yeasts/metabolism
12.
Cell ; 153(1): 153-65, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540696

ABSTRACT

Prion proteins undergo self-sustaining conformational conversions that heritably alter their activities. Many of these proteins operate at pivotal positions in determining how genotype is translated into phenotype. But the breadth of prion influences on biology and their evolutionary significance are just beginning to be explored. We report that a prion formed by the Mot3 transcription factor, [MOT3(+)], governs the acquisition of facultative multicellularity in the budding yeast Saccharomyces cerevisiae. The traits governed by [MOT3(+)] involved both gains and losses of Mot3 regulatory activity. [MOT3(+)]-dependent expression of FLO11, a major determinant of cell-cell adhesion, produced diverse lineage-specific multicellular phenotypes in response to nutrient deprivation. The prions themselves were induced by ethanol and eliminated by hypoxia-conditions that occur sequentially in the natural respiro-fermentative cycles of yeast populations. These data demonstrate that prions can act as environmentally responsive molecular determinants of multicellularity and contribute to the natural morphological diversity of budding yeast.


Subject(s)
Prions/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Transcription Factors/metabolism , Carbon/metabolism , Ethanol/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Oxygen/metabolism , Phenotype , Prions/chemistry , Prions/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics
13.
Cell ; 150(5): 987-1001, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22939624

ABSTRACT

HSP90 is a molecular chaperone that associates with numerous substrate proteins called clients. It plays many important roles in human biology and medicine, but determinants of client recognition by HSP90 have remained frustratingly elusive. We systematically and quantitatively surveyed most human kinases, transcription factors, and E3 ligases for interaction with HSP90 and its cochaperone CDC37. Unexpectedly, many more kinases than transcription factors bound HSP90. CDC37 interacted with kinases, but not with transcription factors or E3 ligases. HSP90::kinase interactions varied continuously over a 100-fold range and provided a platform to study client protein recognition. In wild-type clients, HSP90 did not bind particular sequence motifs, but rather associated with intrinsically unstable kinases. Stabilization of the kinase in either its active or inactive conformation with diverse small molecules decreased HSP90 association. Our results establish HSP90 client recognition as a combinatorial process: CDC37 provides recognition of the kinase family, whereas thermodynamic parameters determine client binding within the family.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Protein Interaction Mapping , Amino Acid Sequence , Animals , Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Humans , Luciferases, Renilla/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein Stability , Proteome/analysis , Receptors, Steroid/metabolism , Sequence Alignment , Thermodynamics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
14.
Cell ; 150(3): 549-62, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863008

ABSTRACT

Heat-Shock Factor 1 (HSF1), master regulator of the heat-shock response, facilitates malignant transformation, cancer cell survival, and proliferation in model systems. The common assumption is that these effects are mediated through regulation of heat-shock protein (HSP) expression. However, the transcriptional network that HSF1 coordinates directly in malignancy and its relationship to the heat-shock response have never been defined. By comparing cells with high and low malignant potential alongside their nontransformed counterparts, we identify an HSF1-regulated transcriptional program specific to highly malignant cells and distinct from heat shock. Cancer-specific genes in this program support oncogenic processes: cell-cycle regulation, signaling, metabolism, adhesion and translation. HSP genes are integral to this program, however, many are uniquely regulated in malignancy. This HSF1 cancer program is active in breast, colon and lung tumors isolated directly from human patients and is strongly associated with metastasis and death. Thus, HSF1 rewires the transcriptome in tumorigenesis, with prognostic and therapeutic implications.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasms/metabolism , Transcription Factors/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome, Human , Heat Shock Transcription Factors , Humans , Neoplasms/pathology , Transcription Factors/analysis , Transcription Factors/genetics
15.
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
16.
Proc Natl Acad Sci U S A ; 121(29): e2313370121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985769

ABSTRACT

Heat Shock Factor 1 (HSF1) is best known as the master transcriptional regulator of the heat-shock response (HSR), a conserved adaptive mechanism critical for protein homeostasis (proteostasis). Combining a genome-wide RNAi library with an HSR reporter, we identified Jumonji domain-containing protein 6 (JMJD6) as an essential mediator of HSF1 activity. In follow-up studies, we found that JMJD6 is itself a noncanonical transcriptional target of HSF1 which acts as a critical regulator of proteostasis. In a positive feedback circuit, HSF1 binds and promotes JMJD6 expression, which in turn reduces heat shock protein 70 (HSP70) R469 monomethylation to disrupt HSP70-HSF1 repressive complexes resulting in enhanced HSF1 activation. Thus, JMJD6 is intricately wired into the proteostasis network where it plays a critical role in cellular adaptation to proteotoxic stress.


Subject(s)
HSP70 Heat-Shock Proteins , Heat Shock Transcription Factors , Heat-Shock Response , Jumonji Domain-Containing Histone Demethylases , Proteostasis , Humans , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Heat-Shock Response/physiology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Proteostasis/physiology , Feedback, Physiological , Adaptation, Physiological , HEK293 Cells , Proteotoxic Stress
17.
Cell ; 146(2): 318-31, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21757228

ABSTRACT

Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinson's disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies.


Subject(s)
Parkinson Disease/pathology , Pluripotent Stem Cells , Point Mutation , Cell Line , Embryonic Stem Cells , Genetic Engineering , Genome-Wide Association Study , Humans , Mutagenesis , Oligonucleotides/metabolism , alpha-Synuclein/genetics
18.
Cell ; 137(1): 146-58, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19345193

ABSTRACT

Prions are proteins that convert between structurally and functionally distinct states, one or more of which is transmissible. In yeast, this ability allows them to act as non-Mendelian elements of phenotypic inheritance. To further our understanding of prion biology, we conducted a bioinformatic proteome-wide survey for prionogenic proteins in S. cerevisiae, followed by experimental investigations of 100 prion candidates. We found an unexpected amino acid bias in aggregation-prone candidates and discovered that 19 of these could also form prions. At least one of these prion proteins, Mot3, produces a bona fide prion in its natural context that increases population-level phenotypic heterogeneity. The self-perpetuating states of these proteins present a vast source of heritable phenotypic variation that increases the adaptability of yeast populations to diverse environments.


Subject(s)
Prions/analysis , Proteome/analysis , Saccharomyces cerevisiae Proteins/analysis , Amino Acid Sequence , Amyloid/metabolism , Asparagine/metabolism , Cytosol/metabolism , Genome, Fungal , Glutamine/metabolism , Heat-Shock Proteins/metabolism , Molecular Sequence Data , Peptide Termination Factors , Phenotype , Prions/chemistry , Prions/genetics , Prions/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
19.
Annu Rev Cell Dev Biol ; 26: 211-33, 2010.
Article in English | MEDLINE | ID: mdl-20500090

ABSTRACT

In the late 1990s, mutations in the synaptic protein α-synuclein (α-syn) were identified in families with hereditary Parkinson's disease (PD). Rapidly, α-syn became the target of numerous investigations that have transformed our understanding of the pathogenesis underlying this disorder. α-Syn is the major component of Lewy bodies (LBs), cytoplasmic protein aggregates that form in the neurons of PD patients. α-Syn interacts with lipid membranes and adopts amyloid conformations that deposit within LBs. Work in yeast and other model systems has revealed that α-syn-associated toxicity might be the consequence of abnormal membrane interactions and alterations in vesicle trafficking. Here we review evidence regarding α-syn's normal interactions with membranes and regulation of synaptic vesicles as well as how overexpression of α-syn yields global cellular dysfunction. Finally, we present a model linking vesicle dynamics to toxicity with the sincere hope that understanding these disease mechanisms will lead to the development of novel, potent therapeutics.


Subject(s)
Parkinson Disease/pathology , alpha-Synuclein/metabolism , Animals , Humans , Lewy Bodies/metabolism , Parkinson Disease/metabolism , Transport Vesicles/metabolism
20.
Nat Rev Mol Cell Biol ; 11(7): 515-28, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20531426

ABSTRACT

Heat shock protein 90 (HSP90) is a highly conserved molecular chaperone that facilitates the maturation of a wide range of proteins (known as clients). Clients are enriched in signal transducers, including kinases and transcription factors. Therefore, HSP90 regulates diverse cellular functions and exerts marked effects on normal biology, disease and evolutionary processes. Recent structural and functional analyses have provided new insights on the transcriptional and biochemical regulation of HSP90 and the structural dynamics it uses to act on a diverse client repertoire. Comprehensive understanding of how HSP90 functions promises not only to provide new avenues for therapeutic intervention, but to shed light on fundamental biological questions.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Proteins/metabolism , Animals , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , Humans , Models, Biological , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Folding , Proteins/chemistry , Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL