Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nat Commun ; 14(1): 7630, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37993433

ABSTRACT

Although the genetic basis and pathogenesis of type 1 diabetes have been studied extensively, how host responses to environmental factors might contribute to autoantibody development remains largely unknown. Here, we use longitudinal blood transcriptome sequencing data to characterize host responses in children within 12 months prior to the appearance of type 1 diabetes-linked islet autoantibodies, as well as matched control children. We report that children who present with insulin-specific autoantibodies first have distinct transcriptional profiles from those who develop GADA autoantibodies first. In particular, gene dosage-driven expression of GSTM1 is associated with GADA autoantibody positivity. Moreover, compared with controls, we observe increased monocyte and decreased B cell proportions 9-12 months prior to autoantibody positivity, especially in children who developed antibodies against insulin first. Lastly, we show that control children present transcriptional signatures consistent with robust immune responses to enterovirus infection, whereas children who later developed islet autoimmunity do not. These findings highlight distinct immune-related transcriptomic differences between case and control children prior to case progression to islet autoimmunity and uncover deficient antiviral response in children who later develop islet autoimmunity.


Subject(s)
Diabetes Mellitus, Type 1 , Enterovirus Infections , Islets of Langerhans , Humans , Child , Autoantibodies , Transcriptome , Autoimmunity/genetics , Insulin/metabolism , Enterovirus Infections/genetics , Islets of Langerhans/metabolism
2.
PLoS Pathog ; 18(1): e1010249, 2022 01.
Article in English | MEDLINE | ID: mdl-35085371

ABSTRACT

Stress granules (SGs) are highly dynamic cytoplasmic foci that form in response to activation of the integrated stress response (ISR) that results in eIF2α phosphorylation and global translation shutdown. Stress granules, which are largely nucleated by G3BP1, serve as hubs for mRNA triage, but there is mounting evidence that they also perform cell signaling functions that are vital to cell survival, particularly during viral infection. We previously showed that SG formation leads to NFκB activation and JNK signaling and that this association may be due in part to G3BP1-dependent recruitment of PKR to SGs. Others have reported close associations between G3BP1 and various innate immune PRRs of the type 1 interferon signaling system, including RIG-I. We also reported SG assembly dynamics is dependent on the arginine-methylation status of G3BP1. Another protein that rapidly localizes to SGs, TDRD3, is a methyl reader protein that performs transcriptional activation and adaptor functions within the nucleus, but neither the mechanism nor its function in SGs is clear. Here, we present evidence that TDRD3 localizes to SGs partly based upon methylation potential of G3BP1. We also characterize granules that TDRD3 forms during overexpression and show that these granules can form in the absence of G3BP but also contain translation components found in canonical SGs. We also show for the first time that SGs recruit additional interferon effectors IRF3, IRF7, TBK1, and Sting, and provide evidence that TDRD3 may play a role in recruitment of these factors. We also present evidence that TDRD3 is a novel antiviral protein that is cleaved by enteroviral 2A proteinase. G3BP1 and TDRD3 knockdown in cells results in altered transcriptional regulation of numerous IFN effectors in complex modulatory patterns that are distinctive for G3BP1 and TDRD3. Overall, we describe a novel role of TDRD3 in innate immunity in which G3BP1 and TDRD3 may coordinate to play important roles in regulation of innate antiviral defenses.


Subject(s)
DNA Helicases/immunology , Immunity, Innate/immunology , Poly-ADP-Ribose Binding Proteins/immunology , Proteins/immunology , RNA Helicases/immunology , RNA Recognition Motif Proteins/immunology , Virus Diseases/immunology , Cell Line , Humans , Interferons/immunology , Signal Transduction/immunology , Stress Granules/immunology
3.
Annu Rev Med ; 73: 483-499, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34794324

ABSTRACT

Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by insulin deficiency and resultant hyperglycemia. Complex interactions of genetic and environmental factors trigger the onset of autoimmune mechanisms responsible for development of autoimmunity to ß cell antigens and subsequent development of T1D. A potential role of virus infections has long been hypothesized, and growing evidence continues to implicate enteroviruses as the most probable triggering viruses. Recent studies have strengthened the association between enteroviruses and development of autoimmunity in T1D patients, potentially through persistent infections. Enterovirus infections may contribute to different stages of disease development. We review data from both human cohort studies and experimental research exploring the potential roles and molecular mechanisms by which enterovirus infections can impact disease outcome.


Subject(s)
Diabetes Mellitus, Type 1 , Enterovirus Infections , Enterovirus , Insulin-Secreting Cells , Autoimmunity , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/genetics , Enterovirus Infections/epidemiology , Humans
4.
Hum Mutat ; 41(11): 1918-1930, 2020 11.
Article in English | MEDLINE | ID: mdl-32790018

ABSTRACT

Diamond-Blackfan anemia (DBA) is a ribosomopathy of variable expressivity and penetrance characterized by red cell aplasia, congenital anomalies, and predisposition to certain cancers, including early-onset colorectal cancer (CRC). DBA is primarily caused by a dominant mutation of a ribosomal protein (RP) gene, although approximately 20% of patients remain genetically uncharacterized despite exome sequencing and copy number analysis. Although somatic loss-of-function mutations in RP genes have been reported in sporadic cancers, with the exceptions of 5q-myelodysplastic syndrome (RPS14) and microsatellite unstable CRC (RPL22), these cancers are not enriched in DBA. Conversely, pathogenic variants in RPS20 were previously implicated in familial CRC; however, none of the reported individuals had classical DBA features. We describe two unrelated children with DBA lacking variants in known DBA genes who were found by exome sequencing to have de novo novel missense variants in RPS20. The variants affect the same amino acid but result in different substitutions and reduce the RPS20 protein level. Yeast models with mutation of the cognate residue resulted in defects in growth, ribosome biogenesis, and polysome formation. These findings expand the phenotypic spectrum of RPS20 mutation beyond familial CRC to include DBA, which itself is associated with increased risk of CRC.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Germ-Line Mutation , Ribosomal Proteins/genetics , Adolescent , Amino Acid Sequence , Child , Colorectal Neoplasms/genetics , Female , Humans , Infant, Newborn , Male , Pedigree , Penetrance , Protein Structure, Tertiary , Exome Sequencing
5.
Viruses ; 12(7)2020 07 11.
Article in English | MEDLINE | ID: mdl-32664501

ABSTRACT

Using immunohistochemistry, enterovirus capsid proteins were demonstrated in pancreatic islets of patients with type 1 diabetes. Virus proteins are mainly located in beta cells, supporting the hypothesis that enterovirus infections may contribute to the pathogenesis of type 1 diabetes. In samples of pancreatic tissue, enterovirus RNA was also detected, but in extremely small quantities and in a smaller proportion of cases compared to the enteroviral protein. Difficulties in detecting viral RNA could be due to the very small number of infected cells, the possible activity of PCR inhibitors, and the presence-during persistent infection-of the viral genome in unencapsidated forms. The aim of this study was twofold: (a) to examine if enzymes or other compounds in pancreatic tissue could affect the molecular detection of encapsidated vs. unencapsidated enterovirus forms, and (b) to compare the sensitivity of RT-PCR methods used in different laboratories. Dilutions of encapsidated and unencapsidated virus were spiked into human pancreas homogenate and analyzed by RT-PCR. Incubation of pancreatic homogenate on wet ice for 20 h did not influence the detection of encapsidated virus. In contrast, a 15-min incubation on wet ice dramatically reduced detection of unencapsidated forms of virus. PCR inhibitors could not be found in pancreatic extract. The results show that components in the pancreas homogenate may selectively affect the detection of unencapsidated forms of enterovirus. This may lead to difficulties in diagnosing persisting enterovirus infection in the pancreas of patients with type 1 diabetes.


Subject(s)
Capsid Proteins/metabolism , Diabetes Mellitus, Type 1/virology , Enterovirus Infections/complications , Enterovirus/genetics , RNA, Viral/metabolism , Diabetes Mellitus, Type 1/etiology , Enterovirus B, Human/genetics , Enterovirus Infections/virology , Humans , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/virology , Real-Time Polymerase Chain Reaction
6.
Virology ; 540: 88-96, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31759187

ABSTRACT

HIV-1 is dependent upon cellular proteins to mediate the many processes required for viral replication. One such protein, PACS1, functions to localize Furin to the trans-Golgi network where Furin cleaves HIV-1 gp160 Envelope into gp41 and gp120. We show here that PACS1 also shuttles between the nucleus and cytoplasm, associates with the viral Rev protein and its cofactor CRM1, and contributes to nuclear export of viral transcripts. PACS1 appears specific to the Rev-CRM1 pathway and not other retroviral RNA export pathways. Over-expression of PACS1 increases nuclear export of unspliced viral RNA and significantly increases p24 expression in HIV-1-infected Jurkat CD4+ T cells. SiRNA depletion and over-expression experiments suggest that PACS1 may promote trafficking of HIV-1 GagPol RNA to a pathway distinct from that of translation on polyribosomes.


Subject(s)
HIV Infections/metabolism , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , RNA, Viral/metabolism , Vesicular Transport Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , Karyopherins/metabolism , Protein Binding , Protein Transport , RNA Transport , Receptors, Cytoplasmic and Nuclear/metabolism , Virus Replication , Exportin 1 Protein
7.
Gut ; 69(8): 1416-1422, 2020 08.
Article in English | MEDLINE | ID: mdl-31744911

ABSTRACT

OBJECTIVE: Higher gluten intake, frequent gastrointestinal infections and adenovirus, enterovirus, rotavirus and reovirus have been proposed as environmental triggers for coeliac disease. However, it is not known whether an interaction exists between the ingested gluten amount and viral exposures in the development of coeliac disease. This study investigated whether distinct viral exposures alone or together with gluten increase the risk of coeliac disease autoimmunity (CDA) in genetically predisposed children. DESIGN: The Environmental Determinants of Diabetes in the Young study prospectively followed children carrying the HLA risk haplotypes DQ2 and/or DQ8 and constructed a nested case-control design. From this design, 83 CDA case-control pairs were identified. Median age of CDA was 31 months. Stool samples collected monthly up to the age of 2 years were analysed for virome composition by Illumina next-generation sequencing followed by comprehensive computational virus profiling. RESULTS: The cumulative number of stool enteroviral exposures between 1 and 2 years of age was associated with an increased risk for CDA. In addition, there was a significant interaction between cumulative stool enteroviral exposures and gluten consumption. The risk conferred by stool enteroviruses was increased in cases reporting higher gluten intake. CONCLUSIONS: Frequent exposure to enterovirus between 1 and 2 years of age was associated with increased risk of CDA. The increased risk conferred by the interaction between enteroviruses and higher gluten intake indicate a cumulative effect of these factors in the development of CDA.


Subject(s)
Autoimmune Diseases/etiology , Celiac Disease/etiology , Enterovirus/isolation & purification , Feces/virology , Glutens/administration & dosage , Adenoviridae/isolation & purification , Autoantibodies/blood , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , Autoimmunity , Case-Control Studies , Celiac Disease/blood , Celiac Disease/genetics , Child, Preschool , Diet , Female , GTP-Binding Proteins/immunology , Genetic Predisposition to Disease , HLA-DQ Antigens/genetics , Humans , Infant , Male , Metagenomics , Protein Glutamine gamma Glutamyltransferase 2 , Risk Factors , Transglutaminases/immunology
8.
Nat Med ; 25(12): 1865-1872, 2019 12.
Article in English | MEDLINE | ID: mdl-31792456

ABSTRACT

Viruses are implicated in autoimmune destruction of pancreatic islet ß cells, which results in insulin deficiency and type 1 diabetes (T1D)1-4. Certain enteroviruses can infect ß cells in vitro5, have been detected in the pancreatic islets of patients with T1D6 and have shown an association with T1D in meta-analyses4. However, establishing consistency in findings across studies has proven difficult. Obstacles to convincingly linking RNA viruses to islet autoimmunity may be attributed to rapid viral mutation rates, the cyclical periodicity of viruses7 and the selection of variants with altered pathogenicity and ability to spread in populations. ß cells strongly express cell-surface coxsackie and adenovirus receptor (CXADR) genes, which can facilitate enterovirus infection8. Studies of human pancreata and cultured islets have shown significant variation in enteroviral virulence to ß cells between serotypes and within the same serotype9,10. In this large-scale study of known eukaryotic DNA and RNA viruses in stools from children, we evaluated fecally shed viruses in relation to islet autoimmunity and T1D. This study showed that prolonged enterovirus B rather than independent, short-duration enterovirus B infections may be involved in the development of islet autoimmunity, but not T1D, in some young children. Furthermore, we found that fewer early-life human mastadenovirus C infections, as well as CXADR rs6517774, independently correlated with islet autoimmunity.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Type 1/virology , Enterovirus/isolation & purification , RNA, Viral/isolation & purification , Adolescent , Autoimmunity/genetics , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Enterovirus/immunology , Enterovirus/pathogenicity , Feces/virology , Female , Humans , Infant , Insulin/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/virology , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Islets of Langerhans/virology , Male , Pancreas/immunology , Pancreas/pathology , Pancreas/virology
10.
Diabetologia ; 62(5): 744-753, 2019 05.
Article in English | MEDLINE | ID: mdl-30675626

ABSTRACT

In type 1 diabetes, pancreatic beta cells are destroyed by chronic autoimmune responses. The disease develops in genetically susceptible individuals, but a role for environmental factors has been postulated. Viral infections have long been considered as candidates for environmental triggers but, given the lack of evidence for an acute, widespread, cytopathic effect in the pancreas in type 1 diabetes or for a closely related temporal association of diabetes onset with such infections, a role for viruses in type 1 diabetes remains unproven. Moreover, viruses have rarely been isolated from the pancreas of individuals with type 1 diabetes, mainly (but not solely) due to the inaccessibility of the organ. Here, we review past and recent literature to evaluate the proposals that chronic, recurrent and, possibly, persistent enteroviral infections occur in pancreatic beta cells in type 1 diabetes. We also explore whether these infections may be sustained by different virus strains over time and whether multiple viral hits can occur during the natural history of type 1 diabetes. We emphasise that only a minority of beta cells appear to be infected at any given time and that enteroviruses may become replication defective, which could explain why they have been isolated from the pancreas only rarely. We argue that enteroviral infection of beta cells largely depends on the host innate and adaptive immune responses, including innate responses mounted by beta cells. Thus, we propose that viruses could play a role in type 1 diabetes on multiple levels, including in the triggering and chronic stimulation of autoimmunity and in the generation of inflammation and the promotion of beta cell dysfunction and stress, each of which might then contribute to autoimmunity, as part of a vicious circle. We conclude that studies into the effects of vaccinations and/or antiviral drugs (some of which are currently on-going) is the only means by which the role of viruses in type 1 diabetes can be finally proven or disproven.


Subject(s)
Antiviral Agents/therapeutic use , Diabetes Mellitus, Type 1/virology , Enterovirus Infections/prevention & control , Pancreas/physiopathology , Viral Vaccines/therapeutic use , Adaptive Immunity , Autoimmunity , Biological Specimen Banks , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/epidemiology , Enterovirus Infections/complications , Enterovirus Infections/drug therapy , Humans , Immunity, Innate , Insulin-Secreting Cells/metabolism , Pancreas/virology , Viral Vaccines/economics
11.
Nature ; 562(7728): 583-588, 2018 10.
Article in English | MEDLINE | ID: mdl-30356187

ABSTRACT

The development of the microbiome from infancy to childhood is dependent on a range of factors, with microbial-immune crosstalk during this time thought to be involved in the pathobiology of later life diseases1-9 such as persistent islet autoimmunity and type 1 diabetes10-12. However, to our knowledge, no studies have performed extensive characterization of the microbiome in early life in a large, multi-centre population. Here we analyse longitudinal stool samples from 903 children between 3 and 46 months of age by 16S rRNA gene sequencing (n = 12,005) and metagenomic sequencing (n = 10,867), as part of the The Environmental Determinants of Diabetes in the Young (TEDDY) study. We show that the developing gut microbiome undergoes three distinct phases of microbiome progression: a developmental phase (months 3-14), a transitional phase (months 15-30), and a stable phase (months 31-46). Receipt of breast milk, either exclusive or partial, was the most significant factor associated with the microbiome structure. Breastfeeding was associated with higher levels of Bifidobacterium species (B. breve and B. bifidum), and the cessation of breast milk resulted in faster maturation of the gut microbiome, as marked by the phylum Firmicutes. Birth mode was also significantly associated with the microbiome during the developmental phase, driven by higher levels of Bacteroides species (particularly B. fragilis) in infants delivered vaginally. Bacteroides was also associated with increased gut diversity and faster maturation, regardless of the birth mode. Environmental factors including geographical location and household exposures (such as siblings and furry pets) also represented important covariates. A nested case-control analysis revealed subtle associations between microbial taxonomy and the development of islet autoimmunity or type 1 diabetes. These data determine the structural and functional assembly of the microbiome in early life and provide a foundation for targeted mechanistic investigation into the consequences of microbial-immune crosstalk for long-term health.


Subject(s)
Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Surveys and Questionnaires , Adolescent , Animals , Bifidobacterium/classification , Bifidobacterium/genetics , Bifidobacterium/isolation & purification , Breast Feeding/statistics & numerical data , Case-Control Studies , Child , Child, Preschool , Cluster Analysis , Datasets as Topic , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Female , Firmicutes/classification , Firmicutes/genetics , Firmicutes/isolation & purification , Gastrointestinal Microbiome/genetics , Humans , Infant , Male , Milk, Human/immunology , Milk, Human/microbiology , Pets , RNA, Ribosomal, 16S/genetics , Siblings , Time Factors
12.
Nat Commun ; 9(1): 3205, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097567

ABSTRACT

Accurate classification of the human virome is critical to a full understanding of the role viruses play in health and disease. This implies the need for sensitive, specific, and practical pipelines that return precise outputs while still enabling case-specific post hoc analysis. Viral taxonomic characterization from metagenomic data suffers from high background noise and signal crosstalk that confounds current methods. Here we develop VirMAP that overcomes these limitations using techniques that merge nucleotide and protein information to taxonomically classify viral reconstructions independent of genome coverage or read overlap. We validate VirMAP using published data sets and viral mock communities containing RNA and DNA viruses and bacteriophages. VirMAP offers opportunities to enhance metagenomic studies seeking to define virome-host interactions, improve biosurveillance capabilities, and strengthen molecular epidemiology reporting.


Subject(s)
DNA Viruses/genetics , Information Storage and Retrieval , Sequence Analysis, DNA , Software , Base Sequence , Databases, Genetic , Genome, Viral , Humans , Metagenomics
13.
Cancer Res ; 78(15): 4229-4240, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29844125

ABSTRACT

Cooperativity between WNT and FGF signaling is well documented in embryonic development and cancer progression, but the molecular mechanisms underlying this cross-talk remain elusive. In this study, we interrogated the dynamics of RNA levels, ribosome occupancy, and protein expression as a function of inducible FGF signaling in mouse mammary glands with constitutive WNT hyperactivation. Multiomics correlation analysis revealed a substantial discrepancy between RNA and ribosome occupancy levels versus protein levels. However, this discrepancy decreased as cells became premalignant and dynamically responded to FGF signaling, implicating the importance of stringent gene regulation in nontransformed cells. Analysis of individual genes demonstrated that acute FGF hyperactivation increased translation of many stem cell self-renewal regulators, including WNT signaling components, and decreased translation of genes regulating cellular senescence. WNT pathway components translationally upregulated by FGF signaling had long and structured 5' UTRs with a high frequency of polypurine sequences, several of which harbored (CGG)4 motifs that can fold into either stable G-quadruplexes or other stable secondary structures. The FGF-mediated increase in translation of WNT pathway components was compromised by silvestrol, an inhibitor of EIF4A that clamps EIF4A to polypurine sequences to block 43S scanning and inhibits its RNA-unwinding activity important for translation initiation. Moreover, silvestrol treatment significantly delayed FGF-WNT-driven tumorigenesis. Taken together, these results suggest that FGF signaling selectively enhances translation of structured mRNAs, particularly WNT signaling components, and highlight their vulnerability to inhibitors that target the RNA helicase EIF4A.Significance: The RNA helicase EIF4A may serve as a therapeutic target for breast cancers that require FGF and WNT signaling. Cancer Res; 78(15); 4229-40. ©2018 AACR.


Subject(s)
5' Untranslated Regions/genetics , Eukaryotic Initiation Factor-4A/genetics , Protein Biosynthesis/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Wnt Signaling Pathway/genetics , 5' Untranslated Regions/drug effects , Animals , Mice , Protein Biosynthesis/drug effects , RNA Helicases/genetics , RNA, Messenger/genetics , Ribosomes/drug effects , Ribosomes/genetics , Triterpenes/pharmacology , Wnt Signaling Pathway/drug effects
14.
J Biol Chem ; 292(46): 18886-18896, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28972166

ABSTRACT

Stress granules (SG) are membrane-less organelles that are condensates of stalled translation initiation complexes and mRNAs. SG formation is a cytoprotective response to environmental stress and results from protein interactions involving regions of low amino acid complexity and poorly defined post-translational modifications of SG components. Many RNA-binding proteins are methylated, and we previously demonstrated that the potent SG-nucleating protein G3BP1 is methylated by protein arginine methyltransferase 1 and 5 (PRMT1 and PRMT5). G3BP1 methylation represses SG formation and is reversible. Here we functionally link JMJD6 (Jumonji C domain-containing protein 6) to G3BP1 demethylation. Our findings reveal that JMJD6 is a novel SG component that interacts with G3BP1 complexes, and its expression reduces G3BP1 monomethylation and asymmetric dimethylation at three Arg residues. Knockdown of JMJD6 repressed SG formation and G3BP1 demethylation, but SG formation and G3BP1 demethylation were rescued with catalytically active but not mutant JMJD6. These results suggest that JMJD6 functions directly or indirectly as an arginine demethylase of G3BP1 that promotes SG formation.


Subject(s)
Cytoplasmic Granules/metabolism , DNA Helicases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Arginine/metabolism , Cell Line , Demethylation , Humans , Protein Interaction Maps , Protein Processing, Post-Translational , Stress, Physiological
15.
Mol Cell Biol ; 37(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27920254

ABSTRACT

Stress granules (SGs) are large macromolecular aggregates that contain translation initiation complexes and mRNAs. Stress granule formation coincides with translational repression, and stress granules actively signal to mediate cell fate decisions by signaling to the translation apparatus to (i) maintain translational repression, (ii) mount various transcriptional responses, including innate immunity, and (iii) repress apoptosis. Previous work showed that G3BP1 is phosphorylated at serine 149, which regulates G3BP1 oligomerization, stress granule assembly, and RNase activity intrinsic to G3BP1. However, the kinase that phosphorylates G3BP1 was not identified, leaving a key step in stress granule regulation uncharacterized. Here, using chemical inhibition, genetic depletion, and overexpression experiments, we show that casein kinase 2 (CK2) promotes stress granule dynamics. These results link CK2 activity with SG disassembly. We also show that casein kinase 2 phosphorylates G3BP1 at serine 149 in vitro and in cells. These data support a role for casein kinase 2 in regulation of protein synthesis by downregulating stress granule formation through G3BP1.


Subject(s)
Carrier Proteins/metabolism , Casein Kinase II/metabolism , Cytoplasmic Granules/metabolism , Stress, Physiological , Arsenites/toxicity , Casein Kinase II/antagonists & inhibitors , Cell Line, Tumor , Cytoplasmic Granules/drug effects , DNA Helicases , Genes, Dominant , Humans , Phosphorylation/drug effects , Poly-ADP-Ribose Binding Proteins , Protein Kinase Inhibitors/pharmacology , Protein Subunits/metabolism , RNA Helicases , RNA Recognition Motif Proteins , Stress, Physiological/drug effects
17.
J Biol Chem ; 291(43): 22671-22685, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27601476

ABSTRACT

Stress granules (SGs) are cytoplasmic condensates of stalled messenger ribonucleoprotein complexes (mRNPs) that form when eukaryotic cells encounter environmental stress. RNA-binding proteins are enriched for arginine methylation and facilitate SG assembly through interactions involving regions of low amino acid complexity. How methylation of specific RNA-binding proteins regulates RNA granule assembly has not been characterized. Here, we examined the potent SG-nucleating protein Ras-GAP SH3-binding protein 1 (G3BP1), and found that G3BP1 is differentially methylated on specific arginine residues by protein arginine methyltransferase (PRMT) 1 and PRMT5 in its RGG domain. Several genetic and biochemical interventions that increased methylation repressed SG assembly, whereas interventions that decreased methylation promoted SG assembly. Arsenite stress quickly and reversibly decreased asymmetric arginine methylation on G3BP1. These data indicate that arginine methylation in the RGG domain prevents large SG assembly and rapid demethylation is a novel signal that regulates SG formation.


Subject(s)
Arsenites/pharmacology , Carrier Proteins/metabolism , Cytoplasmic Granules/metabolism , Stress, Physiological/drug effects , Arginine/genetics , Arginine/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cytoplasmic Granules/genetics , DNA Helicases , Humans , Methylation , Poly-ADP-Ribose Binding Proteins , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , RNA Helicases , RNA Recognition Motif Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
Viruses ; 8(4): 93, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27043612

ABSTRACT

Enteroviruses such as poliovirus (PV) and coxsackievirus B3 (CVB3) have evolved several parallel strategies to regulate cellular gene expression and stress responses to ensure efficient expression of the viral genome. Enteroviruses utilize their encoded proteinases to take over the cellular translation apparatus and direct ribosomes to viral mRNAs. In addition, viral proteinases are used to control and repress the two main types of cytoplasmic RNA granules, stress granules (SGs) and processing bodies (P-bodies, PBs), which are stress-responsive dynamic structures involved in repression of gene expression. This review discusses these processes and the current understanding of the underlying mechanisms with respect to enterovirus infections. In addition, the review discusses accumulating data suggesting linkage exists between RNA granule formation and innate immune sensing and activation.


Subject(s)
Enterovirus Infections/genetics , Enterovirus Infections/virology , Enterovirus/physiology , Gene Expression Regulation , Host-Pathogen Interactions , Protein Biosynthesis , RNA, Viral/genetics , Stress, Physiological , Animals , Cytoplasmic Granules/metabolism , Enterovirus Infections/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Proteolysis , RNA Caps/genetics , RNA Caps/metabolism , RNA, Viral/metabolism , Signal Transduction
19.
Viruses ; 7(12): 6127-40, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26610553

ABSTRACT

We have previously shown that poliovirus (PV) infection induces stress granule (SG) formation early in infection and then inhibits the formation of SG and disperses processing bodies (PBs) by the mid-phase of infection. Loss of SG was linked to cleavage of G3BP1 by viral 3C proteinase (3C(pro)), however dispersal of PBs was not strongly linked to cleavage of specific factors by viral proteinases, suggesting other viral proteins may play roles in inhibition of SG or PB formation. Here we have screened all viral proteins for roles in inducing or inhibiting the formation of RNA granules by creating fusions with mCherry and expressing them individually in cells. Expression of viral proteins separately revealed that the capsid region P1, 2A(pro), 3A, 3C(pro), the protease precursor 3CD and 3D polymerase all affect RNA granules to varying extents, whereas 2BC does not. 2A(pro), which cleaves eIF4GI, induced SGs as expected, and entered novel foci containing the SG nucleating protein G3BP1. Of the two forms of G3BP, only G3BP1 is cleaved by a virus proteinase, 3C(pro), whereas G3BP2 is not cleaved by 3C(pro) or 2A(pro). Surprisingly, 3CD, which contains proteinase activity, differentially repressed PBs but not SGs. Further, both 2A(pro) and 3C(pro) expression dispersed PBs, however molecular targets were different since PB dispersal due to 2A(pro) and heat shock protein (Hsp)90 inhibition but not 3C(pro), could be rescued by application of oxidative stress to cells. The data indicate that PV repression of SGs and PBs is multifactorial, though protease function is dominant.


Subject(s)
Cytoplasm/virology , Cytoplasmic Granules/metabolism , Host-Pathogen Interactions , Poliovirus/physiology , RNA/metabolism , Viral Proteins/metabolism , Virus Replication
20.
mBio ; 6(2): e02486, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25784705

ABSTRACT

UNLABELLED: Stress granules (SGs) are dynamic cytoplasmic repositories containing translationally silenced mRNAs that assemble upon cellular stress. We recently reported that the SG nucleating protein G3BP1 promotes antiviral activity and is essential in double-stranded RNA-dependent protein kinase (PKR) recruitment to stress granules, thereby driving phosphorylation of the α subunit of eukaryotic initiation factor 2 (eIF2α). Here, we delineate the mechanism for SG-dependent PKR activation. We show that G3BP1 and inactive PKR directly interact with each other, dependent on both the NTF2-like and PXXP domains of G3BP1. The G3BP1-interacting protein Caprin1 also directly interacts with PKR, regulates efficient PKR activation at the stress granule, and is also integral for the release of active PKR into the cytoplasm to engage in substrate recognition. The G3BP1-Caprin1-PKR complex represents a new mode of PKR activation and is important for antiviral activity of G3BP1 and PKR during infection with mengovirus. Our data links stress responses and their resultant SGs with innate immune activation through PKR without a requirement for foreign double-stranded RNA (dsRNA) pattern recognition. IMPORTANCE: Our previous work indicates that stress granules have antiviral activity and mediate innate immunity through functions of G3BP1; however, the mechanistic details of these functions were not resolved. We show that much of the antiviral activity of stress granules is contingent on the function of PKR in a complex with G3BP1 and Caprin1. The PKR-G3BP1-Caprin1 complex undergoes dynamic transitioning within and outside stress granules to accomplish PKR activation and translational repression. This mechanism appears to function distinctly from canonical pattern recognition of double-stranded RNA by PKR. Therefore, this mechanism bridges the stress response with innate immunity, allowing the cell to respond to many cellular stressors and amplify the pathogen pattern recognition systems of innate immunity.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cytoplasmic Granules/metabolism , Mengovirus/immunology , RNA, Double-Stranded/metabolism , eIF-2 Kinase/metabolism , Animals , Cell Line , DNA Helicases , Host-Pathogen Interactions , Humans , Mengovirus/growth & development , Mice, Knockout , Poly-ADP-Ribose Binding Proteins , Protein Binding , Protein Interaction Mapping , RNA Helicases , RNA Recognition Motif Proteins
SELECTION OF CITATIONS
SEARCH DETAIL