Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.358
Filter
Add more filters

Publication year range
1.
Cell ; 187(5): 1238-1254.e14, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38367616

ABSTRACT

CD4+ T cells with latent HIV-1 infection persist despite treatment with antiretroviral agents and represent the main barrier to a cure of HIV-1 infection. Pharmacological disruption of viral latency may expose HIV-1-infected cells to host immune activity, but the clinical efficacy of latency-reversing agents for reducing HIV-1 persistence remains to be proven. Here, we show in a randomized-controlled human clinical trial that the histone deacetylase inhibitor panobinostat, when administered in combination with pegylated interferon-α2a, induces a structural transformation of the HIV-1 reservoir cell pool, characterized by a disproportionate overrepresentation of HIV-1 proviruses integrated in ZNF genes and in chromatin regions with reduced H3K27ac marks, the molecular target sites for panobinostat. By contrast, proviruses near H3K27ac marks were actively selected against, likely due to increased susceptibility to panobinostat. These data suggest that latency-reversing treatment can increase the immunological vulnerability of HIV-1 reservoir cells and accelerate the selection of epigenetically privileged HIV-1 proviruses.


Subject(s)
HIV Infections , HIV-1 , Histone Deacetylase Inhibitors , Interferon-alpha , Panobinostat , Proviruses , Humans , HIV Infections/drug therapy , HIV-1/genetics , Panobinostat/therapeutic use , Proviruses/drug effects , Virus Latency , Histone Deacetylase Inhibitors/therapeutic use , Interferon-alpha/therapeutic use
2.
Annu Rev Biochem ; 89: 529-555, 2020 06 20.
Article in English | MEDLINE | ID: mdl-32097570

ABSTRACT

Protein folding in the cell is mediated by an extensive network of >1,000 chaperones, quality control factors, and trafficking mechanisms collectively termed the proteostasis network. While the components and organization of this network are generally well established, our understanding of how protein-folding problems are identified, how the network components integrate to successfully address challenges, and what types of biophysical issues each proteostasis network component is capable of addressing remains immature. We describe a chemical biology-informed framework for studying cellular proteostasis that relies on selection of interesting protein-folding problems and precise researcher control of proteostasis network composition and activities. By combining these methods with multifaceted strategies to monitor protein folding, degradation, trafficking, and aggregation in cells, researchers continue to rapidly generate new insights into cellular proteostasis.


Subject(s)
Molecular Chaperones/genetics , Molecular Probe Techniques , Proteome/genetics , Proteostasis Deficiencies/genetics , Proteostasis/genetics , Animals , CRISPR-Cas Systems , Gene Expression Regulation , Half-Life , Heat-Shock Response/drug effects , Humans , Molecular Chaperones/metabolism , Protein Aggregates , Protein Engineering/methods , Protein Folding/drug effects , Protein Transport/drug effects , Proteome/chemistry , Proteome/metabolism , Proteostasis/drug effects , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , Signal Transduction , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Unfolded Protein Response/drug effects
3.
Cell ; 183(6): 1572-1585.e16, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33157040

ABSTRACT

Cellular functioning requires the orchestration of thousands of molecular interactions in time and space. Yet most molecules in a cell move by diffusion, which is sensitive to external factors like temperature. How cells sustain complex, diffusion-based systems across wide temperature ranges is unknown. Here, we uncover a mechanism by which budding yeast modulate viscosity in response to temperature and energy availability. This "viscoadaptation" uses regulated synthesis of glycogen and trehalose to vary the viscosity of the cytosol. Viscoadaptation functions as a stress response and a homeostatic mechanism, allowing cells to maintain invariant diffusion across a 20°C temperature range. Perturbations to viscoadaptation affect solubility and phase separation, suggesting that viscoadaptation may have implications for multiple biophysical processes in the cell. Conditions that lower ATP trigger viscoadaptation, linking energy availability to rate regulation of diffusion-controlled processes. Viscoadaptation reveals viscosity to be a tunable property for regulating diffusion-controlled processes in a changing environment.


Subject(s)
Energy Metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Temperature , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Diffusion , Glycogen/metabolism , Homeostasis , Models, Biological , Solubility , Trehalose , Viscosity
4.
Cell ; 181(4): 818-831.e19, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32359423

ABSTRACT

Cells sense elevated temperatures and mount an adaptive heat shock response that involves changes in gene expression, but the underlying mechanisms, particularly on the level of translation, remain unknown. Here we report that, in budding yeast, the essential translation initiation factor Ded1p undergoes heat-induced phase separation into gel-like condensates. Using ribosome profiling and an in vitro translation assay, we reveal that condensate formation inactivates Ded1p and represses translation of housekeeping mRNAs while promoting translation of stress mRNAs. Testing a variant of Ded1p with altered phase behavior as well as Ded1p homologs from diverse species, we demonstrate that Ded1p condensation is adaptive and fine-tuned to the maximum growth temperature of the respective organism. We conclude that Ded1p condensation is an integral part of an extended heat shock response that selectively represses translation of housekeeping mRNAs to promote survival under conditions of severe heat stress.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Fungal/genetics , Protein Biosynthesis/genetics , Saccharomyces cerevisiae Proteins/metabolism , DEAD-box RNA Helicases/physiology , Gene Expression/genetics , Genes, Essential/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/physiology
5.
Cell ; 179(4): 880-894.e10, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31668804

ABSTRACT

Current approaches to reducing the latent HIV reservoir entail first reactivating virus-containing cells to become visible to the immune system. A critical second step is killing these cells to reduce reservoir size. Endogenous cytotoxic T-lymphocytes (CTLs) may not be adequate because of cellular exhaustion and the evolution of CTL-resistant viruses. We have designed a universal CAR-T cell platform based on CTLs engineered to bind a variety of broadly neutralizing anti-HIV antibodies. We show that this platform, convertibleCAR-T cells, effectively kills HIV-infected, but not uninfected, CD4 T cells from blood, tonsil, or spleen and only when armed with anti-HIV antibodies. convertibleCAR-T cells also kill within 48 h more than half of the inducible reservoir found in blood of HIV-infected individuals on antiretroviral therapy. The modularity of convertibleCAR-T cell system, which allows multiplexing with several anti-HIV antibodies yielding greater breadth and control, makes it a promising tool for attacking the latent HIV reservoir.


Subject(s)
Antibodies, Anti-Idiotypic/pharmacology , HIV Infections/therapy , Immunotherapy, Adoptive , Virus Replication/genetics , Animals , Antibodies, Anti-Idiotypic/immunology , HEK293 Cells , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Humans , Mice , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Primary Cell Culture , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology , Virus Latency/immunology , Virus Replication/immunology
6.
Cell ; 172(3): 590-604.e13, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29373831

ABSTRACT

Stress granules (SGs) are transient ribonucleoprotein (RNP) aggregates that form during cellular stress and are increasingly implicated in human neurodegeneration. To study the proteome and compositional diversity of SGs in different cell types and in the context of neurodegeneration-linked mutations, we used ascorbate peroxidase (APEX) proximity labeling, mass spectrometry, and immunofluorescence to identify ∼150 previously unknown human SG components. A highly integrated, pre-existing SG protein interaction network in unstressed cells facilitates rapid coalescence into larger SGs. Approximately 20% of SG diversity is stress or cell-type dependent, with neuronal SGs displaying a particularly complex repertoire of proteins enriched in chaperones and autophagy factors. Strengthening the link between SGs and neurodegeneration, we demonstrate aberrant dynamics, composition, and subcellular distribution of SGs in cells from amyotrophic lateral sclerosis (ALS) patients. Using three Drosophila ALS/FTD models, we identify SG-associated modifiers of neurotoxicity in vivo. Altogether, our results highlight SG proteins as central to understanding and ultimately targeting neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cytoplasmic Granules/metabolism , Protein Interaction Maps , Ribonucleoproteins/metabolism , Stress, Physiological , Animals , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Humans , Neurons/metabolism , Protein Transport
7.
Cell ; 171(7): 1625-1637.e13, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29198525

ABSTRACT

When unfolded proteins accumulate in the endoplasmic reticulum (ER), the unfolded protein response (UPR) increases ER-protein-folding capacity to restore protein-folding homeostasis. Unfolded proteins activate UPR signaling across the ER membrane to the nucleus by promoting oligomerization of IRE1, a conserved transmembrane ER stress receptor. However, the coupling of ER stress to IRE1 oligomerization and activation has remained obscure. Here, we report that the ER luminal co-chaperone ERdj4/DNAJB9 is a selective IRE1 repressor that promotes a complex between the luminal Hsp70 BiP and the luminal stress-sensing domain of IRE1α (IRE1LD). In vitro, ERdj4 is required for complex formation between BiP and IRE1LD. ERdj4 associates with IRE1LD and recruits BiP through the stimulation of ATP hydrolysis, forcibly disrupting IRE1 dimers. Unfolded proteins compete for BiP and restore IRE1LD to its default, dimeric, and active state. These observations establish BiP and its J domain co-chaperones as key regulators of the UPR.


Subject(s)
Endoribonucleases/metabolism , HSP40 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response , Animals , Cricetinae , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Humans , Protein Folding
8.
Cell ; 168(6): 1028-1040.e19, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283059

ABSTRACT

In eukaryotic cells, diverse stresses trigger coalescence of RNA-binding proteins into stress granules. In vitro, stress-granule-associated proteins can demix to form liquids, hydrogels, and other assemblies lacking fixed stoichiometry. Observing these phenomena has generally required conditions far removed from physiological stresses. We show that poly(A)-binding protein (Pab1 in yeast), a defining marker of stress granules, phase separates and forms hydrogels in vitro upon exposure to physiological stress conditions. Other RNA-binding proteins depend upon low-complexity regions (LCRs) or RNA for phase separation, whereas Pab1's LCR is not required for demixing, and RNA inhibits it. Based on unique evolutionary patterns, we create LCR mutations, which systematically tune its biophysical properties and Pab1 phase separation in vitro and in vivo. Mutations that impede phase separation reduce organism fitness during prolonged stress. Poly(A)-binding protein thus acts as a physiological stress sensor, exploiting phase separation to precisely mark stress onset, a broadly generalizable mechanism.


Subject(s)
Cytoplasmic Granules/metabolism , Poly(A)-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Amino Acid Sequence , Cytoplasmic Granules/chemistry , Hot Temperature , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Mutagenesis , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/genetics , Proline/analysis , Proline/metabolism , Protein Domains , Ribonucleases/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Stress, Physiological
9.
Mol Cell ; 84(1): 80-93, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38103561

ABSTRACT

Cellular homeostasis is constantly challenged by a myriad of extrinsic and intrinsic stressors. To mitigate the stress-induced damage, cells activate transient survival programs. The heat shock response (HSR) is an evolutionarily well-conserved survival program that is activated in response to proteotoxic stress. The HSR encompasses a dual regulation of transcription, characterized by rapid activation of genes encoding molecular chaperones and concomitant global attenuation of non-chaperone genes. Recent genome-wide approaches have delineated the molecular depth of stress-induced transcriptional reprogramming. The dramatic rewiring of gene and enhancer networks is driven by key transcription factors, including heat shock factors (HSFs), that together with chromatin-modifying enzymes remodel the 3D chromatin architecture, determining the selection of either gene activation or repression. Here, we highlight the current advancements of molecular mechanisms driving transcriptional reprogramming during acute heat stress. We also discuss the emerging implications of HSF-mediated stress signaling in the context of physiological and pathological conditions.


Subject(s)
Proteostasis , Transcription Factors , Proteostasis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Heat-Shock Response/genetics , Molecular Chaperones/genetics , Chromatin/genetics , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism
10.
Mol Cell ; 84(15): 2856-2869.e9, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121843

ABSTRACT

RNA polymerase II (RNA Pol II)-mediated transcription is a critical, highly regulated process aided by protein complexes at distinct steps. Here, to investigate RNA Pol II and transcription-factor-binding and dissociation dynamics, we generated endogenous photoactivatable-GFP (PA-GFP) and HaloTag knockins using CRISPR-Cas9, allowing us to track a population of molecules at the induced Hsp70 loci in Drosophila melanogaster polytene chromosomes. We found that early in the heat-shock response, little RNA Pol II and DRB sensitivity-inducing factor (DSIF) are reused for iterative rounds of transcription. Surprisingly, although PAF1 and Spt6 are found throughout the gene body by chromatin immunoprecipitation (ChIP) assays, they show markedly different binding behaviors. Additionally, we found that PAF1 and Spt6 are only recruited after positive transcription elongation factor (P-TEFb)-mediated phosphorylation and RNA Pol II promoter-proximal pause escape. Finally, we observed that PAF1 may be expendable for transcription of highly expressed genes where nucleosome density is low. Thus, our live-cell imaging data provide key constraints to mechanistic models of transcription regulation.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , RNA Polymerase II , Transcription, Genetic , Transcriptional Elongation Factors , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Positive Transcriptional Elongation Factor B/metabolism , Positive Transcriptional Elongation Factor B/genetics , Promoter Regions, Genetic , CRISPR-Cas Systems , Transcription Factors/metabolism , Transcription Factors/genetics , Polytene Chromosomes/genetics , Polytene Chromosomes/metabolism , Gene Expression Regulation , Phosphorylation , Protein Binding , Heat-Shock Response/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
11.
Mol Cell ; 84(4): 687-701.e7, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38266641

ABSTRACT

Molecular chaperones are critical for protein homeostasis and are implicated in several human pathologies such as neurodegeneration and cancer. While the binding of chaperones to nascent and misfolded proteins has been studied in great detail, the direct interaction between chaperones and RNA has not been systematically investigated. Here, we provide the evidence for widespread interaction between chaperones and RNA in human cells. We show that the major chaperone heat shock protein 70 (HSP70) binds to non-coding RNA transcribed by RNA polymerase III (RNA Pol III) such as tRNA and 5S rRNA. Global chromatin profiling revealed that HSP70 binds genomic sites of transcription by RNA Pol III. Detailed biochemical analyses showed that HSP70 alleviates the inhibitory effect of cognate tRNA transcript on tRNA gene transcription. Thus, our study uncovers an unexpected role of HSP70-RNA interaction in the biogenesis of a specific class of non-coding RNA with wider implications in cancer therapeutics.


Subject(s)
HSP70 Heat-Shock Proteins , Neoplasms , Humans , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , RNA , RNA Polymerase III/genetics , RNA Polymerase III/metabolism , RNA, Transfer/genetics , RNA, Untranslated/genetics
12.
Mol Cell ; 84(9): 1727-1741.e12, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38547866

ABSTRACT

Heat-shocked cells prioritize the translation of heat shock (HS) mRNAs, but the underlying mechanism is unclear. We report that HS in budding yeast induces the disassembly of the eIF4F complex, where eIF4G and eIF4E assemble into translationally arrested mRNA ribonucleoprotein particles (mRNPs) and HS granules (HSGs), whereas eIF4A promotes HS translation. Using in vitro reconstitution biochemistry, we show that a conformational rearrangement of the thermo-sensing eIF4A-binding domain of eIF4G dissociates eIF4A and promotes the assembly with mRNA into HS-mRNPs, which recruit additional translation factors, including Pab1p and eIF4E, to form multi-component condensates. Using extracts and cellular experiments, we demonstrate that HS-mRNPs and condensates repress the translation of associated mRNA and deplete translation factors that are required for housekeeping translation, whereas HS mRNAs can be efficiently translated by eIF4A. We conclude that the eIF4F complex is a thermo-sensing node that regulates translation during HS.


Subject(s)
Eukaryotic Initiation Factor-4F , Eukaryotic Initiation Factor-4G , Heat-Shock Response , Poly(A)-Binding Proteins , Protein Biosynthesis , RNA, Messenger , Ribonucleoproteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Heat-Shock Response/genetics , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4F/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-4G/genetics , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4A/genetics , Gene Expression Regulation, Fungal , Protein Binding , RNA, Fungal/metabolism , RNA, Fungal/genetics
13.
Annu Rev Biochem ; 85: 715-42, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27050154

ABSTRACT

Molecular chaperones control the cellular folding, assembly, unfolding, disassembly, translocation, activation, inactivation, disaggregation, and degradation of proteins. In 1989, groundbreaking experiments demonstrated that a purified chaperone can bind and prevent the aggregation of artificially unfolded polypeptides and use ATP to dissociate and convert them into native proteins. A decade later, other chaperones were shown to use ATP hydrolysis to unfold and solubilize stable protein aggregates, leading to their native refolding. Presently, the main conserved chaperone families Hsp70, Hsp104, Hsp90, Hsp60, and small heat-shock proteins (sHsps) apparently act as unfolding nanomachines capable of converting functional alternatively folded or toxic misfolded polypeptides into harmless protease-degradable or biologically active native proteins. Being unfoldases, the chaperones can proofread three-dimensional protein structures and thus control protein quality in the cell. Understanding the mechanisms of the cellular unfoldases is central to the design of new therapies against aging, degenerative protein conformational diseases, and specific cancers.


Subject(s)
Chaperonin 60/chemistry , HSP110 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/chemistry , Heat-Shock Proteins, Small/chemistry , Mitochondrial Proteins/chemistry , Protein Unfolding , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Gene Expression , HSP110 Heat-Shock Proteins/genetics , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/genetics , Heat-Shock Proteins, Small/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Protein Aggregates , Protein Folding , Protein Structure, Quaternary , Rhodospirillum rubrum/chemistry , Rhodospirillum rubrum/metabolism
14.
Genes Dev ; 38(9-10): 380-392, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38816072

ABSTRACT

The ability to sense and respond to proteotoxic insults declines with age, leaving cells vulnerable to chronic and acute stressors. Reproductive cues modulate this decline in cellular proteostasis to influence organismal stress resilience in Caenorhabditis elegans We previously uncovered a pathway that links the integrity of developing embryos to somatic health in reproductive adults. Here, we show that the nuclear receptor NHR-49, an ortholog of mammalian peroxisome proliferator-activated receptor α (PPARα), regulates stress resilience and proteostasis downstream from embryo integrity and other pathways that influence lipid homeostasis and upstream of HSF-1. Disruption of the vitelline layer of the embryo envelope, which activates a proteostasis-enhancing intertissue pathway in somatic cells, triggers changes in lipid catabolism gene expression that are accompanied by an increase in fat stores. NHR-49, together with its coactivator, MDT-15, contributes to this remodeling of lipid metabolism and is also important for the elevated stress resilience mediated by inhibition of the embryonic vitelline layer. Our findings indicate that NHR-49 also contributes to stress resilience in other pathways known to change lipid homeostasis, including reduced insulin-like signaling and fasting, and that increased NHR-49 activity is sufficient to improve proteostasis and stress resilience in an HSF-1-dependent manner. Together, our results establish NHR-49 as a key regulator that links lipid homeostasis and cellular resilience to proteotoxic stress.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Lipid Metabolism , Proteostasis , Receptors, Cytoplasmic and Nuclear , Reproduction , Signal Transduction , Stress, Physiological , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Lipid Metabolism/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Reproduction/genetics , Reproduction/physiology , Mediator Complex/genetics , Mediator Complex/metabolism
15.
Immunity ; 55(2): 224-236.e5, 2022 02 08.
Article in English | MEDLINE | ID: mdl-34995475

ABSTRACT

During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.


Subject(s)
Blood Platelets/drug effects , NFATC Transcription Factors/antagonists & inhibitors , Platelet Aggregation/drug effects , Sepsis/pathology , Animals , Blood Coagulation/drug effects , Blood Platelets/metabolism , Cell Communication/drug effects , Cytoplasmic Granules/metabolism , Disease Models, Animal , Extracellular Traps/metabolism , Humans , Inflammation , Mice , NFATC Transcription Factors/metabolism , Neutrophils/metabolism , Receptors, Thrombin/metabolism , Sepsis/metabolism
16.
Cell ; 167(7): 1788-1802.e13, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984727

ABSTRACT

More than 98% of the mammalian genome is noncoding, and interspersed transposable elements account for ∼50% of noncoding space. Here, we demonstrate that a specific interaction between the polycomb protein EZH2 and RNA made from B2 SINE retrotransposons controls stress-responsive genes in mouse cells. In the heat-shock model, B2 RNA binds stress genes and suppresses their transcription. Upon stress, EZH2 is recruited and triggers cleavage of B2 RNA. B2 degradation in turn upregulates stress genes. Evidence indicates that B2 RNA operates as a "speed bump" against advancement of RNA polymerase II, and temperature stress releases the brakes on transcriptional elongation. These data attribute a new function to EZH2 that is independent of its histone methyltransferase activity and reconcile how EZH2 can be associated with both gene repression and activation. Our study reveals that EZH2 and B2 together control activation of a large network of genes involved in thermal stress.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation , Heat-Shock Response , RNA, Untranslated/metabolism , Retroelements , Animals , Embryonic Stem Cells/metabolism , Mice , NIH 3T3 Cells , RNA Polymerase II/metabolism , Transcription, Genetic
17.
Mol Cell ; 83(2): 252-265.e13, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36630955

ABSTRACT

The conserved regulon of heat shock factor 1 in budding yeast contains chaperones for general protein folding as well as zinc-finger protein Zpr1, whose essential role in archaea and eukaryotes remains unknown. Here, we show that Zpr1 depletion causes acute proteotoxicity driven by biosynthesis of misfolded eukaryotic translation elongation factor 1A (eEF1A). Prolonged Zpr1 depletion leads to eEF1A insufficiency, thereby inducing the integrated stress response and inhibiting protein synthesis. Strikingly, we show by using two distinct biochemical reconstitution approaches that Zpr1 enables eEF1A to achieve a conformational state resistant to protease digestion. Lastly, we use a ColabFold model of the Zpr1-eEF1A complex to reveal a folding mechanism mediated by the Zpr1's zinc-finger and alpha-helical hairpin structures. Our work uncovers the long-sought-after function of Zpr1 as a bespoke chaperone tailored to the biogenesis of one of the most abundant proteins in the cell.


Subject(s)
Carrier Proteins , Molecular Chaperones , Carrier Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Biosynthesis , Zinc/metabolism , Zinc Fingers , Peptide Elongation Factor 1/metabolism
18.
Mol Cell ; 83(17): 3108-3122.e13, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37597513

ABSTRACT

General protein folding is mediated by chaperones that utilize ATP hydrolysis to regulate client binding and release. Zinc-finger protein 1 (Zpr1) is an essential ATP-independent chaperone dedicated to the biogenesis of eukaryotic translation elongation factor 1A (eEF1A), a highly abundant GTP-binding protein. How Zpr1-mediated folding is regulated to ensure rapid Zpr1 recycling remains an unanswered question. Here, we use yeast genetics and microscopy analysis, biochemical reconstitution, and structural modeling to reveal that folding of eEF1A by Zpr1 requires GTP hydrolysis. Furthermore, we identify the highly conserved altered inheritance of mitochondria 29 (Aim29) protein as a Zpr1 co-chaperone that recognizes eEF1A in the GTP-bound, pre-hydrolysis conformation. This interaction dampens Zpr1⋅eEF1A GTPase activity and facilitates client exit from the folding cycle. Our work reveals that a bespoke ATP-independent chaperone system has mechanistic similarity to ATPase chaperones but unexpectedly relies on client GTP hydrolysis to regulate the chaperone-client interaction.


Subject(s)
Carrier Proteins , GTP Phosphohydrolases , Molecular Chaperones , Peptide Elongation Factors , Saccharomyces cerevisiae Proteins , Humans , Adenosine Triphosphate , GTP Phosphohydrolases/genetics , Guanosine Triphosphate , Molecular Chaperones/genetics , Peptide Elongation Factors/metabolism , Saccharomyces cerevisiae , Carrier Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Protein Folding
19.
Genes Dev ; 37(9-10): 398-417, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37257919

ABSTRACT

Eusocial insect reproductive females show strikingly longer life spans than nonreproductive female workers despite high genetic similarity. In the ant Harpegnathos saltator (Hsal), workers can transition to reproductive "gamergates," acquiring a fivefold prolonged life span by mechanisms that are poorly understood. We found that gamergates have elevated expression of heat shock response (HSR) genes in the absence of heat stress and enhanced survival with heat stress. This HSR gene elevation is driven in part by gamergate-specific up-regulation of the gene encoding a truncated form of a heat shock factor most similar to mammalian HSF2 (hsalHSF2). In workers, hsalHSF2 was bound to DNA only upon heat stress. In gamergates, hsalHSF2 bound to DNA even in the absence of heat stress and was localized to gamergate-biased HSR genes. Expression of hsalHSF2 in Drosophila melanogaster led to enhanced heat shock survival and extended life span in the absence of heat stress. Molecular characterization illuminated multiple parallels between long-lived flies and gamergates, underscoring the centrality of hsalHSF2 to extended ant life span. Hence, ant caste-specific heat stress resilience and extended longevity can be transferred to flies via hsalHSF2. These findings reinforce the critical role of proteostasis in health and aging and reveal novel mechanisms underlying facultative life span extension in ants.


Subject(s)
Ants , Longevity , Animals , Female , Longevity/genetics , Ants/genetics , Drosophila melanogaster/genetics , Aging , Heat-Shock Response/genetics , Mammals
20.
Mol Cell ; 82(4): 741-755.e11, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35148816

ABSTRACT

Stresses such as heat shock trigger the formation of protein aggregates and the induction of a disaggregation system composed of molecular chaperones. Recent work reveals that several cases of apparent heat-induced aggregation, long thought to be the result of toxic misfolding, instead reflect evolved, adaptive biomolecular condensation, with chaperone activity contributing to condensate regulation. Here we show that the yeast disaggregation system directly disperses heat-induced biomolecular condensates of endogenous poly(A)-binding protein (Pab1) orders of magnitude more rapidly than aggregates of the most commonly used misfolded model substrate, firefly luciferase. Beyond its efficiency, heat-induced condensate dispersal differs from heat-induced aggregate dispersal in its molecular requirements and mechanistic behavior. Our work establishes a bona fide endogenous heat-induced substrate for long-studied heat shock proteins, isolates a specific example of chaperone regulation of condensates, and underscores needed expansion of the proteotoxic interpretation of the heat shock response to encompass adaptive, chaperone-mediated regulation.


Subject(s)
Biomolecular Condensates/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Poly(A)-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Binding, Competitive , Biomolecular Condensates/genetics , HSP40 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Poly(A)-Binding Proteins/genetics , Protein Aggregates , Protein Binding , Protein Folding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL