Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
RNA ; 30(6): 644-661, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38423626

ABSTRACT

UV-crosslinking has proven to be an invaluable tool for the identification of RNA-protein interactomes. The paucity of methods for distinguishing background from bona fide RNA-protein interactions, however, makes attribution of RNA-binding function on UV-crosslinking alone challenging. To address this need, we previously reported an RNA-binding protein (RBP) confidence scoring metric (RCS), incorporating both signal-to-noise (S:N) and protein abundance determinations to distinguish high- and low-confidence candidate RBPs. Although RCS has utility, we sought a direct metric for quantification and comparative evaluation of protein-RNA interactions. Here we propose the use of protein-specific UV-crosslinking efficiency (%CL), representing the molar fraction of a protein that is crosslinked to RNA, for functional evaluation of candidate RBPs. Application to the HeLa RNA interactome yielded %CL values for 1097 proteins. Remarkably, %CL values span over five orders of magnitude. For the HeLa RNA interactome, %CL values comprise a range from high efficiency, high specificity interactions, e.g., the Elav protein HuR and the Pumilio homolog Pum2, with %CL values of 45.9 and 24.2, respectively, to very low efficiency and specificity interactions, for example, the metabolic enzymes glyceraldehyde-3-phosphate dehydrogenase, fructose-bisphosphate aldolase, and alpha-enolase, with %CL values of 0.0016, 0.006, and 0.008, respectively. We further extend the utility of %CL through prediction of protein domains and classes with known RNA-binding functions, thus establishing it as a useful metric for RNA interactome analysis. We anticipate that this approach will benefit efforts to establish functional RNA interactomes and support the development of more predictive computational approaches for RBP identification.


Subject(s)
RNA-Binding Proteins , RNA , Ultraviolet Rays , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , RNA/metabolism , RNA/genetics , Humans , HeLa Cells , Protein Binding , Cross-Linking Reagents/chemistry
2.
Semin Cell Dev Biol ; 156: 160-166, 2024 03 15.
Article in English | MEDLINE | ID: mdl-36202692

ABSTRACT

Stress granules (SGs), structurally dynamic, optically resolvable, macromolecular assemblies of mRNAs, RNA binding proteins (RBPs), translation factors, ribosomal subunits, as well as other interacting proteins, assemble in response to cell stress conditions that elicit phosphorylation of eukaryotic initiation factor 2α (eIF2α) and consequently, the inactivation of translation initiation. SG biology is conserved throughout eukaryotes and has recently been linked to the pathological sequelae of neurodegenerative disorders, cancer biology, and viral infection. Substantial insights into mechanisms of SG biogenesis, and more broadly the phenomenon of biological liquid-liquid phase separation (LLPS), have been aided by detailed proteomic and transcriptomic studies as well as in vitro reconstitution approaches. A particularly interesting and largely unexplored element of SG biology is the cell biological context of SG biogenesis, including its subcellular organization and more recently, evidence that the endoplasmic reticulum (ER) membrane may serve important functions in RNA granule biology generally and SG biogenesis specifically. A central role for the ER in SG biogenesis is discussed and a hypothesis linking SG formation on the ER to the trafficking, localization and de novo translation of newly exported mRNAs is presented.


Subject(s)
Proteomics , Stress Granules , Cytoplasmic Granules , Endoplasmic Reticulum/metabolism , RNA-Binding Proteins/metabolism , RNA, Messenger/metabolism
3.
Nat Commun ; 14(1): 5868, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37735163

ABSTRACT

Recent efforts towards the comprehensive identification of RNA-bound proteomes have revealed a large, surprisingly diverse family of candidate RNA-binding proteins (RBPs). Quantitative metrics for characterization and validation of protein-RNA interactions and their dynamic interactions have, however, proven analytically challenging and prone to error. Here we report a method termed LEAP-RBP (Liquid-Emulsion-Assisted-Purification of RNA-Bound Protein) for the selective, quantitative recovery of UV-crosslinked RNA-protein complexes. By virtue of its high specificity and yield, LEAP-RBP distinguishes RNA-bound and RNA-free protein levels and reveals common sources of experimental noise in RNA-centric RBP enrichment methods. We introduce strategies for accurate RBP identification and signal-based metrics for quantifying protein-RNA complex enrichment, relative RNA occupancy, and method specificity. In this work, the utility of our approach is validated by comprehensive identification of RBPs whose association with mRNA is modulated in response to global mRNA translation state changes and through in-depth benchmark comparisons with current methodologies.


Subject(s)
Benchmarking , RNA , RNA, Messenger/genetics , Protein Biosynthesis , RNA-Binding Proteins/genetics
4.
Proc Natl Acad Sci U S A ; 120(38): e2221448120, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37695916

ABSTRACT

Evidence has long suggested that epidermal growth factor receptor (EGFR) may play a prominent role in triple-negative breast cancer (TNBC) pathogenesis, but clinical trials of EGFR inhibitors have yielded disappointing results. Using a candidate drug screen, we identified that inhibition of cyclin-dependent kinases 12 and 13 (CDK12/13) dramatically sensitizes diverse models of TNBC to EGFR blockade. This combination therapy drives cell death through the 4E-BP1-dependent suppression of the translation and translation-linked turnover of driver oncoproteins, including MYC. A genome-wide CRISPR/Cas9 screen identified the CCR4-NOT complex as a major determinant of sensitivity to the combination therapy whose loss renders 4E-BP1 unresponsive to drug-induced dephosphorylation, thereby rescuing MYC translational suppression and promoting MYC stability. The central roles of CCR4-NOT and 4E-BP1 in response to the combination therapy were further underscored by the observation of CNOT1 loss and rescue of 4E-BP1 phosphorylation in TNBC cells that naturally evolved therapy resistance. Thus, pharmacological inhibition of CDK12/13 reveals a long-proposed EGFR dependence in TNBC that functions through the cooperative regulation of translation-coupled oncoprotein stability.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , ErbB Receptors/genetics , Phosphorylation , Cell Death , Oncogene Proteins , Cyclin-Dependent Kinases/genetics , Transcription Factors
5.
RNA ; 29(11): 1703-1724, 2023 11.
Article in English | MEDLINE | ID: mdl-37643813

ABSTRACT

Signal recognition particle (SRP) pathway function in protein translocation across the endoplasmic reticulum (ER) is well established; its role in RNA localization to the ER remains, however, unclear. In current models, mRNAs undergo translation- and SRP-dependent trafficking to the ER, with ER localization mediated via interactions between SRP-bound translating ribosomes and the ER-resident SRP receptor (SR), a heterodimeric complex comprising SRA, the SRP-binding subunit, and SRB, an integral membrane ER protein. To study SRP pathway function in RNA localization, SR knockout (KO) mammalian cell lines were generated and the consequences of SR KO on steady-state and dynamic mRNA localization examined. CRISPR/Cas9-mediated SRPRB KO resulted in profound destabilization of SRA. Pairing siRNA silencing of SRPRA in SRPRB KO cells yielded viable SR KO cells. Steady-state mRNA compositions and ER-localization patterns in parental and SR KO cells were determined by cell fractionation and deep sequencing. Notably, steady-state cytosol and ER mRNA compositions and partitioning patterns were largely unaltered by loss of SR expression. To examine SRP pathway function in RNA localization dynamics, the subcellular trafficking itineraries of newly exported mRNAs were determined by 4-thiouridine (4SU) pulse-labeling/4SU-seq/cell fractionation. Newly exported mRNAs were distinguished by high ER enrichment, with ER localization being SR-independent. Intriguingly, under conditions of translation initiation inhibition, the ER was the default localization site for all newly exported mRNAs. These data demonstrate that mRNA localization to the ER can be uncoupled from the SRP pathway function and reopen questions regarding the mechanism of RNA localization to the ER.


Subject(s)
Endoplasmic Reticulum , Signal Recognition Particle , Animals , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Cytosol/metabolism , Membrane Proteins/genetics , Mammals/genetics
6.
Cell Rep ; 40(9): 111268, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044842

ABSTRACT

Patients with human epidermal growth factor receptor 2-positive (HER2+/ERBB2) breast cancer often present with brain metastasis. HER2-targeted therapies have not been successful to treat brain metastases in part due to poor blood-brain barrier (BBB) penetrance and emergence of resistance. Here, we report that Abelson (ABL) kinase allosteric inhibitors improve overall survival and impair HER2+ brain metastatic outgrowth in vivo. Mechanistically, ABL kinases phosphorylate the RNA-binding protein Y-box-binding protein 1 (YB-1). ABL kinase inhibition disrupts binding of YB-1 to the ERBB2 mRNA and impairs translation, leading to a profound decrease in HER2 protein levels. ABL-dependent tyrosine phosphorylation of YB-1 promotes HER2 translation. Notably, loss of YB-1 inhibits brain metastatic outgrowth and impairs expression of a subset of ABL-dependent brain metastatic targets. These data support a role for ABL kinases in the translational regulation of brain metastatic targets through YB-1 and offer a therapeutic target for HER2+ brain metastasis patients.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Proto-Oncogene Proteins c-abl , Y-Box-Binding Protein 1 , Brain/metabolism , Brain Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Cell Line, Tumor , Female , Humans , Proto-Oncogene Proteins c-abl/metabolism , Receptor, ErbB-2/metabolism , Y-Box-Binding Protein 1/genetics
7.
RNA ; 27(10): 1241-1256, 2021 10.
Article in English | MEDLINE | ID: mdl-34244458

ABSTRACT

Stress granules (SGs) are membraneless organelles composed of mRNAs and RNA binding proteins which undergo assembly in response to stress-induced inactivation of translation initiation. In general, SG recruitment is limited to a subpopulation of a given mRNA species and RNA-seq analyses of purified SGs revealed that signal sequence-encoding (i.e., endoplasmic reticulum [ER]-targeted) transcripts are significantly underrepresented, consistent with prior reports that ER localization can protect mRNAs from SG recruitment. Using translational profiling, cell fractionation, and single molecule mRNA imaging, we examined SG biogenesis following activation of the unfolded protein response (UPR) by 1,4-dithiothreitol (DTT) and report that gene-specific subsets of cytosolic and ER-targeted mRNAs can be recruited into SGs. Furthermore, we demonstrate that SGs form in close proximity to or directly associated with the ER membrane. ER-associated SG assembly was also observed during arsenite stress, suggesting broad roles for the ER in SG biogenesis. Recruitment of a given mRNA into SGs required stress-induced translational repression, though translational inhibition was not solely predictive of an mRNA's propensity for SG recruitment. SG formation was prevented by the transcriptional inhibitors actinomycin D or triptolide, suggesting a functional link between gene transcriptional state and SG biogenesis. Collectively these data demonstrate that ER-targeted and cytosolic mRNAs can be recruited into ER-associated SGs and this recruitment is sensitive to transcriptional inhibition. We propose that newly transcribed mRNAs exported under conditions of suppressed translation initiation are primary SG substrates, with the ER serving as the central subcellular site of SG formation.


Subject(s)
Cytoplasmic Granules/genetics , Endoplasmic Reticulum/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Unfolded Protein Response , Biomarkers/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Cytosol/drug effects , Cytosol/metabolism , Dactinomycin/pharmacology , Diterpenes/pharmacology , Dithiothreitol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Epoxy Compounds/pharmacology , Gene Expression , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Organelle Biogenesis , Peptide Chain Initiation, Translational/drug effects , Phenanthrenes/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Single Molecule Imaging , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism
8.
Mol Cell Proteomics ; 19(11): 1826-1849, 2020 11.
Article in English | MEDLINE | ID: mdl-32788342

ABSTRACT

Protein synthesis on the endoplasmic reticulum (ER) requires the dynamic coordination of numerous cellular components. Together, resident ER membrane proteins, cytoplasmic translation factors, and both integral membrane and cytosolic RNA-binding proteins operate in concert with membrane-associated ribosomes to facilitate ER-localized translation. Little is known, however, regarding the spatial organization of ER-localized translation. This question is of growing significance as it is now known that ER-bound ribosomes contribute to secretory, integral membrane, and cytosolic protein synthesis alike. To explore this question, we utilized quantitative proximity proteomics to identify neighboring protein networks for the candidate ribosome interactors SEC61ß (subunit of the protein translocase), RPN1 (oligosaccharyltransferase subunit), SEC62 (translocation integral membrane protein), and LRRC59 (ribosome binding integral membrane protein). Biotin labeling time course studies of the four BioID reporters revealed distinct labeling patterns that intensified but only modestly diversified as a function of labeling time, suggesting that the ER membrane is organized into discrete protein interaction domains. Whereas SEC61ß and RPN1 reporters identified translocon-associated networks, SEC62 and LRRC59 reporters revealed divergent protein interactomes. Notably, the SEC62 interactome is enriched in redox-linked proteins and ER luminal chaperones, with the latter likely representing proximity to an ER luminal chaperone reflux pathway. In contrast, the LRRC59 interactome is highly enriched in SRP pathway components, translation factors, and ER-localized RNA-binding proteins, uncovering a functional link between LRRC59 and mRNA translation regulation. Importantly, analysis of the LRRC59 interactome by native immunoprecipitation identified similar protein and functional enrichments. Moreover, [35S]-methionine incorporation assays revealed that siRNA silencing of LRRC59 expression reduced steady state translation levels on the ER by ca. 50%, and also impacted steady state translation levels in the cytosol compartment. Collectively, these data reveal a functional domain organization for the ER and identify a key role for LRRC59 in the organization and regulation of local translation.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Protein Biosynthesis , Proteome/metabolism , Ribosomes/metabolism , Cell Line, Tumor , Computational Biology , Cytosol/metabolism , Gene Ontology , Gene Silencing , Humans , Mass Spectrometry , Membrane Proteins/genetics , Oxidation-Reduction , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Maps , Proteomics , RNA, Small Interfering , Recombinant Proteins , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism
9.
CBE Life Sci Educ ; 18(4): ar51, 2019 12.
Article in English | MEDLINE | ID: mdl-31622166

ABSTRACT

Although doctoral students in the biomedical sciences have been recognized as a population at particular risk for mental health problems such as burnout and depression, little research has been conducted to identify candidate targets for intervention. To this end, we used the stress process model to evaluate potential mediators of stress-burnout and stress-depression relationships in biomedical doctoral students. A cross-sectional sample (n = 69) completed validated self-report measures of stress; symptoms of burnout and depression; and perceptions of mastery, social support, and advisor support. In linear regression models, we found that academic stressors were most predictive of burnout, whereas family/monetary stressors were most predictive of depression. In mediation models, we found that the relationship between stress and burnout was partially mediated by mastery and advisor support, while the stress-depression relationship was partially mediated by mastery. These findings represent a first step in identifying interventional targets to improve mental health in this at-risk population. Whereas certain stressors are inherent to the doctoral training environment, psychosocial interventions to enhance one's sense of mastery and/or to improve advisor relationships may mitigate the influence of such stressors on burnout and depression.


Subject(s)
Biomedical Research/education , Burnout, Professional/psychology , Depression/psychology , Education, Graduate , Stress, Psychological/psychology , Students/psychology , Adult , Cross-Sectional Studies , Female , Humans , Male , Pilot Projects , Regression Analysis , Social Support
10.
CBE Life Sci Educ ; 18(2): ar27, 2019 06.
Article in English | MEDLINE | ID: mdl-31150319

ABSTRACT

Although burnout and mental health problems may adversely impact quality of scientific research, academic productivity, and attrition in biomedical doctoral training programs, very little research has been done on this topic. Recent studies have used brief survey methods to begin to explore burnout and mental health problems in biomedical doctoral students. In this pilot study, biomedical doctoral students (N = 69; 12% of enrolled biomedical doctoral students at a large research institution's school of medicine in the United States) were administered standardized psychiatric interviews and self-report questionnaires focused on dimensions of burnout, mental health symptoms, and academic outcomes. We discovered high levels of burnout, depression, and anxiety. Additionally, we identified that burnout was significantly associated with thoughts related to dropping out, subjective appraisal of employment opportunities, functional impairment due to a mental health problem, and having at least one current psychiatric disorder. These findings extend prior research indicating the presence of significant emotional health challenges doctoral students in biomedical graduate programs face involving high burnout and difficulties with the training environment. We outline several recommendations and next steps to programmatically understand and address these emerging emotional wellness concerns in biomedical doctoral students.


Subject(s)
Biomedical Research/education , Burnout, Professional/psychology , Education, Graduate , Mental Health , Adult , Anxiety/psychology , Depression/psychology , Female , Humans , Male , Pilot Projects , Students , United States , Young Adult
11.
J Biol Chem ; 294(22): 8942-8958, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31004035

ABSTRACT

The endoplasmic reticulum (ER) is a nexus for mRNA localization and translation, and recent studies have demonstrated that ER-bound ribosomes also play a transcriptome-wide role in regulating proteome composition. The Sec61 translocon (SEC61) serves as the receptor for ribosomes that translate secretory/integral membrane protein-encoding mRNAs, but whether SEC61 also serves as a translation site for cytosolic protein-encoding mRNAs remains unknown. Here, using a BioID proximity-labeling approach in HEK293T Flp-In cell lines, we examined interactions between ER-resident proteins and ribosomes in vivo Using in vitro analyses, we further focused on bona fide ribosome interactors (i.e. SEC61) and ER proteins (ribophorin I, leucine-rich repeat-containing 59 (LRRC59), and SEC62) previously implicated in associating with ribosomes. We observed labeling of ER-bound ribosomes with the SEC61ß and LRRC59 BioID reporters, comparatively modest labeling with the ribophorin I reporter, and no labeling with the SEC62 reporter. A biotin pulse-chase/subcellular fractionation approach to examine ribosome exchange at the SEC61ß and LRRC59 sites revealed that, at steady state, ribosomes at these sites comprise both rapid- and slow-exchanging pools. Global translational initiation arrest elicited by the inhibitor harringtonine accelerated SEC61ß reporter-labeled ribosome exchange. RNA-Seq analyses of the mRNAs associated with SEC61ß- and LRRC59-labeled ribosomes revealed both site-enriched and shared mRNAs and further established that the ER has a transcriptome-wide role in regulating proteome composition. These results provide evidence that ribosomes interact with the ER membrane via multiple modes and suggest regulatory mechanisms that control global proteome composition via ER membrane-bound ribosomes.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Profiling/methods , Ribosomes/metabolism , Biotin/genetics , Biotin/metabolism , Cytosol/metabolism , Genes, Reporter , HEK293 Cells , Harringtonines/chemistry , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Ribosomes/chemistry , SEC Translocation Channels/chemistry , SEC Translocation Channels/genetics , SEC Translocation Channels/metabolism
12.
RNA ; 24(5): 688-703, 2018 05.
Article in English | MEDLINE | ID: mdl-29438049

ABSTRACT

Astrocyte elevated gene-1 (AEG-1), an oncogene whose overexpression promotes tumor cell proliferation, angiogenesis, invasion, and enhanced chemoresistance, is thought to function primarily as a scaffolding protein, regulating PI3K/Akt and Wnt/ß-catenin signaling pathways. Here we report that AEG-1 is an endoplasmic reticulum (ER) resident integral membrane RNA-binding protein (RBP). Examination of the AEG-1 RNA interactome by HITS-CLIP and PAR-CLIP methodologies revealed a high enrichment for endomembrane organelle-encoding transcripts, most prominently those encoding ER resident proteins, and within this cohort, for integral membrane protein-encoding RNAs. Cluster mapping of the AEG-1/RNA interaction sites demonstrated a normalized rank order interaction of coding sequence >5' untranslated region, with 3' untranslated region interactions only weakly represented. Intriguingly, AEG-1/membrane protein mRNA interaction sites clustered downstream from encoded transmembrane domains, suggestive of a role in membrane protein biogenesis. Secretory and cytosolic protein-encoding mRNAs were also represented in the AEG-1 RNA interactome, with the latter category notably enriched in genes functioning in mRNA localization, translational regulation, and RNA quality control. Bioinformatic analyses of RNA-binding motifs and predicted secondary structure characteristics indicate that AEG-1 lacks established RNA-binding sites though shares the property of high intrinsic disorder commonly seen in RBPs. These data implicate AEG-1 in the localization and regulation of secretory and membrane protein-encoding mRNAs and provide a framework for understanding AEG-1 function in health and disease.


Subject(s)
Cell Adhesion Molecules/metabolism , Endoplasmic Reticulum/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Cell Adhesion Molecules/chemistry , Endoplasmic Reticulum/genetics , High-Throughput Nucleotide Sequencing , Humans , Intrinsically Disordered Proteins , Membrane Proteins/genetics , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry , Sequence Analysis, RNA
13.
J Virol ; 92(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29321322

ABSTRACT

A primary question in dengue virus (DENV) biology is the molecular strategy for recruitment of host cell protein synthesis machinery. Here, we combined cell fractionation, ribosome profiling, and transcriptome sequencing (RNA-seq) to investigate the subcellular organization of viral genome translation and replication as well as host cell translation and its response to DENV infection. We report that throughout the viral life cycle, DENV plus- and minus-strand RNAs were highly partitioned to the endoplasmic reticulum (ER), identifying the ER as the primary site of DENV translation. DENV infection was accompanied by an ER compartment-specific remodeling of translation, where ER translation capacity was subverted from host transcripts to DENV plus-strand RNA, particularly at late stages of infection. Remarkably, translation levels and patterns in the cytosol compartment were only modestly affected throughout the experimental time course of infection. Comparisons of ribosome footprinting densities of the DENV plus-strand RNA and host mRNAs indicated that DENV plus-strand RNA was only sparsely loaded with ribosomes. Combined, these observations suggest a mechanism where ER-localized translation and translational control mechanisms, likely cis encoded, are used to repurpose the ER for DENV virion production. Consistent with this view, we found ER-linked cellular stress response pathways commonly associated with viral infection, namely, the interferon response and unfolded protein response, to be only modestly activated during DENV infection. These data support a model where DENV reprograms the ER protein synthesis and processing environment to promote viral survival and replication while minimizing the activation of antiviral and proteostatic stress response pathways.IMPORTANCE DENV, a prominent human health threat with no broadly effective or specific treatment, depends on host cell translation machinery for viral replication, immune evasion, and virion biogenesis. The molecular mechanism by which DENV commandeers the host cell protein synthesis machinery and the subcellular organization of DENV replication and viral protein synthesis is poorly understood. Here, we report that DENV has an almost exclusively ER-localized life cycle, with viral replication and translation largely restricted to the ER. Surprisingly, DENV infection largely affects only ER-associated translation, with relatively modest effects on host cell translation in the cytosol. DENV RNA translation is very inefficient, likely representing a strategy to minimize disruption of ER proteostasis. Overall these findings demonstrate that DENV has evolved an ER-compartmentalized life cycle; thus, targeting the molecular signatures and regulation of the DENV-ER interaction landscape may reveal strategies for therapeutic intervention.


Subject(s)
Dengue Virus/physiology , Dengue/immunology , Endoplasmic Reticulum/immunology , Immune Evasion , Protein Biosynthesis/immunology , RNA, Messenger/immunology , RNA, Viral/immunology , Virus Replication/immunology , Cell Line, Tumor , Dengue/pathology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/virology , Humans , Interferons/immunology , Unfolded Protein Response/immunology
14.
Mol Cell Biol ; 36(13): 1868-80, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27161320

ABSTRACT

Phosphorylation of eukaryotic initiation factor 2α (eIF2α) controls transcriptome-wide changes in mRNA translation in stressed cells. While phosphorylated eIF2α (P-eIF2α) attenuates global protein synthesis, mRNAs encoding stress proteins are more efficiently translated. Two eIF2α phosphatases, containing GADD34 and CReP, catalyze P-eIF2α dephosphorylation. The current view of GADD34, whose transcription is stress induced, is that it functions in a feedback loop to resolve cell stress. In contrast, CReP, which is constitutively expressed, controls basal P-eIF2α levels in unstressed cells. Our studies show that GADD34 drives substantial changes in mRNA translation in unstressed cells, particularly targeting the secretome. Following activation of the unfolded protein response (UPR), rapid translation of GADD34 mRNA occurs and GADD34 is essential for UPR progression. In the absence of GADD34, eIF2α phosphorylation is persistently enhanced and the UPR translational program is significantly attenuated. This "stalled" UPR is relieved by the subsequent activation of compensatory mechanisms that include AKT-mediated suppression of PKR-like kinase (PERK) and increased expression of CReP mRNA, partially restoring protein synthesis. Our studies highlight the coordinate regulation of UPR by the GADD34- and CReP-containing eIF2α phosphatases to control cell viability.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Protein Phosphatase 1/metabolism , Unfolded Protein Response , Animals , Cell Survival , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Membrane Proteins/metabolism , Mice , Phosphorylation , Protein Biosynthesis , Protein Phosphatase 1/genetics
15.
Methods ; 91: 69-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26164698

ABSTRACT

The development and application of ribosome profiling has markedly advanced our understanding of ribosomes and mRNA translation. The experimental approach, which relies on deep sequencing of ribosome-protected mRNA fragments generated by treatment of polyribosomes with exogenous nucleases, provides a transcriptome-wide assessment of translation. The broad application of ribosome profiling has been slowed by the complexity and expense of the protocol. Here, we provide a simplified ribosome profiling method that uses micrococcal nuclease to generate ribosome footprints in crude cellular extracts, which are then purified simply by size selection via polyacrylamide gel electrophoresis. This simplification removes the laborious or expensive purification of ribosomes that has typically been used. This direct extraction method generates gene-level ribosome profiling data that are similar to a method that includes ribosome purification. This protocol should significantly ease the barrier to entry for research groups interested in employing ribosome profiling.


Subject(s)
Micrococcal Nuclease , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism , Sequence Analysis, RNA/methods , High-Throughput Nucleotide Sequencing , Humans , Lymphocytes/metabolism
16.
Science ; 348(6240): 1217, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-26068841

ABSTRACT

Jan et al. (Research Articles, 7 November 2014, p. 716) propose that ribosomes translating secretome messenger RNAs (mRNAs) traffic from the cytosol to the endoplasmic reticulum (ER) upon emergence of the signal peptide and return to the cytosol after termination. An accounting of controls demonstrates that mRNAs initiate translation on ER-bound ribosomes and that ribosomes are retained on the ER through many cycles of translation.


Subject(s)
Cells/metabolism , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Protein Biosynthesis , Ribosomes/metabolism , Humans
17.
Nat Rev Mol Cell Biol ; 16(4): 221-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25735911

ABSTRACT

Pioneering electron microscopy studies defined two primary populations of ribosomes in eukaryotic cells: one freely dispersed through the cytoplasm and the other bound to the surface of the endoplasmic reticulum (ER). Subsequent investigations revealed a specialized function for each population, with secretory and integral membrane protein-encoding mRNAs translated on ER-bound ribosomes, and cytosolic protein synthesis was widely attributed to free ribosomes. Recent findings have challenged this view, and transcriptome-scale studies of mRNA distribution and translation have now demonstrated that ER-bound ribosomes also function in the translation of a large fraction of mRNAs that encode cytosolic proteins. These studies suggest a far more expansive role for the ER in transcriptome expression, where membrane and secretory protein synthesis represents one element of a multifaceted and dynamic contribution to post-transcriptional gene expression.


Subject(s)
Endoplasmic Reticulum/genetics , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Genetic Variation , Humans , Models, Biological
18.
Genetics ; 198(2): 723-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25316788

ABSTRACT

Neurodevelopmental defects in humans represent a clinically heterogeneous group of disorders. Here, we report the genetic and functional dissection of a multigenerational pedigree with an X-linked syndromic disorder hallmarked by microcephaly, growth retardation, and seizures. Using an X-linked intellectual disability (XLID) next-generation sequencing diagnostic panel, we identified a novel missense mutation in the gene encoding 60S ribosomal protein L10 (RPL10), a locus associated previously with autism spectrum disorders (ASD); the p.K78E change segregated with disease under an X-linked recessive paradigm while, consistent with causality, carrier females exhibited skewed X inactivation. To examine the functional consequences of the p.K78E change, we modeled RPL10 dysfunction in zebrafish. We show that endogenous rpl10 expression is augmented in anterior structures, and that suppression decreases head size in developing morphant embryos, concomitant with reduced bulk translation and increased apoptosis in the brain. Subsequently, using in vivo complementation, we demonstrate that p.K78E is a loss-of-function variant. Together, our findings suggest that a mutation within the conserved N-terminal end of RPL10, a protein in close proximity to the peptidyl transferase active site of the 60S ribosomal subunit, causes severe defects in brain formation and function.


Subject(s)
Microcephaly/genetics , Ribosomal Proteins/genetics , Adult , Animals , Apoptosis , Brain/pathology , Cell Proliferation , Child, Preschool , Female , Genes, X-Linked , Genetic Association Studies , Humans , Male , Mutation, Missense , Pedigree , Ribosomal Protein L10 , Young Adult , Zebrafish
19.
Cell ; 158(6): 1362-1374, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215492

ABSTRACT

The unfolded protein response (UPR) is a stress response program that reprograms cellular translation and gene expression in response to proteotoxic stress in the endoplasmic reticulum (ER). One of the primary means by which the UPR alleviates this stress is by reducing protein flux into the ER via a general suppression of protein synthesis and ER-specific mRNA degradation. We report here an additional UPR-induced mechanism for the reduction of protein flux into the ER, where mRNAs that encode signal sequences are released from the ER to the cytosol. By removing mRNAs from the site of translocation, this mechanism may serve as a potent means to transiently reduce ER protein folding load and restore proteostasis. These findings identify the dynamic subcellular localization of mRNAs and translation as a selective and rapid regulatory feature of the cellular response to protein folding stress.


Subject(s)
Endoplasmic Reticulum/metabolism , RNA, Messenger/metabolism , Unfolded Protein Response , Animals , Cytosol/metabolism , Dithiothreitol/metabolism , Endoplasmic Reticulum/chemistry , Fibroblasts , Kinetics , Mice , Open Reading Frames , Polyribosomes/metabolism , Protein Biosynthesis
20.
RNA ; 20(10): 1489-98, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25142066

ABSTRACT

The specialized protein synthesis functions of the cytosol and endoplasmic reticulum compartments are conferred by the signal recognition particle (SRP) pathway, which directs the cotranslational trafficking of signal sequence-encoding mRNAs from the cytosol to the endoplasmic reticulum (ER). Although subcellular mRNA distributions largely mirror the binary pattern predicted by the SRP pathway model, studies in mammalian cells, yeast, and Drosophila have also demonstrated that cytosolic protein-encoding mRNAs are broadly represented on ER-bound ribosomes. A mechanism for such noncanonical mRNA localization remains, however, to be identified. Here, we examine the hypothesis that de novo translation initiation on ER-bound ribosomes serves as a mechanism for localizing cytosolic protein-encoding mRNAs to the ER. As a test of this hypothesis, we performed single molecule RNA fluorescence in situ hybridization studies of subcellular mRNA distributions and report that a substantial fraction of mRNAs encoding the cytosolic protein GAPDH resides in close proximity to the ER. Consistent with these data, analyses of subcellular mRNA and ribosome distributions in multiple cell lines demonstrated that cytosolic protein mRNA-ribosome distributions were strongly correlated, whereas signal sequence-encoding mRNA-ribosome distributions were divergent. Ribosome footprinting studies of ER-bound polysomes revealed a substantial initiation codon read density enrichment for cytosolic protein-encoding mRNAs. We also demonstrate that eukaryotic initiation factor 2α is bound to the ER via a salt-sensitive, ribosome-independent mechanism. Combined, these data support ER-localized translation initiation as a mechanism for mRNA recruitment to the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomes/metabolism , Blotting, Western , Cells, Cultured , Cytosol/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Oligonucleotide Array Sequence Analysis , Polyribosomes/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes/genetics , Signal Recognition Particle/genetics , Signal Recognition Particle/metabolism , Subcellular Fractions
SELECTION OF CITATIONS
SEARCH DETAIL
...