Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
EMBO J ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778156

ABSTRACT

During infection viruses hijack host cell metabolism to promote their replication. Here, analysis of metabolite alterations in macrophages exposed to poly I:C recognises that the antiviral effector Protein Kinase RNA-activated (PKR) suppresses glucose breakdown within the pentose phosphate pathway (PPP). This pathway runs parallel to central glycolysis and is critical to producing NADPH and pentose precursors for nucleotides. Changes in metabolite levels between wild-type and PKR-ablated macrophages show that PKR controls the generation of ribose 5-phosphate, in a manner distinct from its established function in gene expression but dependent on its kinase activity. PKR phosphorylates and inhibits the Ribose 5-Phosphate Isomerase A (RPIA), thereby preventing interconversion of ribulose- to ribose 5-phosphate. This activity preserves redox control but decreases production of ribose 5-phosphate for nucleotide biosynthesis. Accordingly, the PKR-mediated immune response to RNA suppresses nucleic acid production. In line, pharmacological targeting of the PPP during infection decreases the replication of the Herpes simplex virus. These results identify an immune response-mediated control of host cell metabolism and suggest targeting the RPIA as a potential innovative antiviral treatment.

2.
Front Immunol ; 14: 1106737, 2023.
Article in English | MEDLINE | ID: mdl-36875104

ABSTRACT

Here we investigate the function of the innate immune molecule protein kinase R (PKR) in intestinal inflammation. To model a colitogenic role of PKR, we determine the physiological response to dextran sulfate sodium (DSS) of wild-type and two transgenic mice strains mutated to express either a kinase-dead PKR or to ablate expression of the kinase. These experiments recognize kinase-dependent and -independent protection from DSS-induced weight loss and inflammation, against a kinase-dependent increase in the susceptibility to DSS-induced injury. We propose these effects arise through PKR-dependent alteration of gut physiology, evidenced as altered goblet cell function and changes to the gut microbiota at homeostasis that suppresses inflammasome activity by controlling autophagy. These findings establish that PKR functions as both a protein kinase and a signaling molecule in instituting immune homeostasis in the gut.


Subject(s)
Colitis , Animals , Mice , Inflammation , Homeostasis , Autophagy , Mice, Transgenic , Protein Kinases
3.
Nat Commun ; 13(1): 5413, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109526

ABSTRACT

Anti-cancer immunity and response to immune therapy is influenced by the metabolic states of the tumours. Immune checkpoint blockade therapy (ICB) is known to involve metabolic adaptation, however, the mechanism is not fully known. Here we show, by metabolic profiling of plasma samples from melanoma-bearing mice undergoing anti-PD1 and anti-CTLA4 combination therapy, that higher levels of purine metabolites, including inosine, mark ICB sensitivity. Metabolic profiles of ICB-treated human cancers confirm the association between inosine levels and ICB sensitivity. In mouse models, inosine supplementation sensitizes tumours to ICB, even if they are intrinsically ICB resistant, by enhancing T cell-mediated cytotoxicity and hence generating an immunologically hotter microenvironment. We find that inosine directly inhibits UBA6 in tumour cells, and lower level of UBA6 makes the tumour more immunogenic and this is reflected in favourable outcome following ICB therapy in human melanomas. Transplanted mouse melanoma and breast cancer cells with genetic ablation of Uba6 show higher sensitivity to ICB than wild type tumours. Thus, we provide evidence of an inosine-regulated UBA6-dependent pathway governing tumour-intrinsic immunogenicity and hence sensitivity to immune checkpoint inhibition, which might provide targets to overcome ICB resistance.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Animals , Combined Modality Therapy , Humans , Inosine/pharmacology , Melanoma/pathology , Mice , Radioimmunotherapy , Tumor Microenvironment , Ubiquitin-Activating Enzymes
4.
J Virol ; 96(13): e0045422, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35695504

ABSTRACT

In this issue, Gao and colleagues (J Virol 96:e00167-22, https://doi.org/10.1128/JVI.00167-22) dissect innate immune signaling in a microglial cell line infected with severe fever with thrombocytopenia syndrome virus (SFTSV). This virus has been designated a priority pathogen by the World Health Organization due to its capacity to induce a fatal cytokine storm. The study's findings attribute the pathogenesis to induction of the host inflammasome response by the SFTSV nonstructural protein.


Subject(s)
Bunyaviridae Infections , Encephalitis , Phlebovirus , Bunyaviridae Infections/immunology , Bunyaviridae Infections/virology , Encephalitis/immunology , Encephalitis/virology , Humans , Phlebovirus/metabolism , Signal Transduction/physiology , Viral Nonstructural Proteins/metabolism
5.
Sci Immunol ; 7(68): eabi6763, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148201

ABSTRACT

Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αß) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.


Subject(s)
Immunity, Innate/immunology , Interleukins/immunology , eIF-2 Kinase/immunology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , eIF-2 Kinase/deficiency
6.
Oncogene ; 37(14): 1939-1948, 2018 04.
Article in English | MEDLINE | ID: mdl-29367758

ABSTRACT

Hypermethylated-in-Cancer 1 (Hic1) is a tumor suppressor gene frequently inactivated by epigenetic silencing and loss-of-heterozygosity in a broad range of cancers. Loss of HIC1, a sequence-specific zinc finger transcriptional repressor, results in deregulation of genes that promote a malignant phenotype in a lineage-specific manner. In particular, upregulation of the HIC1 target gene SIRT1, a histone deacetylase, can promote tumor growth by inactivating TP53. An alternate line of evidence suggests that HIC1 can promote the repair of DNA double strand breaks through an interaction with MTA1, a component of the nucleosome remodeling and deacetylase (NuRD) complex. Using a conditional knockout mouse model of tumor initiation, we now show that inactivation of Hic1 results in cell cycle arrest, premature senescence, chromosomal instability and spontaneous transformation in vitro. This phenocopies the effects of deleting Brca1, a component of the homologous recombination DNA repair pathway, in mouse embryonic fibroblasts. These effects did not appear to be mediated by deregulation of Hic1 target gene expression or loss of Tp53 function, and rather support a role for Hic1 in maintaining genome integrity during sustained replicative stress. Loss of Hic1 function also cooperated with activation of oncogenic KRas in the adult airway epithelium of mice, resulting in the formation of highly pleomorphic adenocarcinomas with a micropapillary phenotype in vivo. These results suggest that loss of Hic1 expression in the early stages of tumor formation may contribute to malignant transformation through the acquisition of chromosomal instability.


Subject(s)
Chromosomal Instability/genetics , Kruppel-Like Transcription Factors/physiology , Neoplasms/genetics , Tumor Suppressor Protein p53/physiology , Animals , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence/genetics , Embryo, Mammalian , Female , Genes, Tumor Suppressor/physiology , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/pathology
7.
J Leukoc Biol ; 103(2): 185-192, 2018 02.
Article in English | MEDLINE | ID: mdl-28974542

ABSTRACT

IFNs protect us against infection from viral pathogens, but can also induce damaging inflammation and are associated with the development of autoimmune conditions. By dissecting the response that is mediated by different IFN-regulated genes, we hoped to identify targets that will enable us to preserve the defense against pathogens while minimizing immune disease. Toward this, several reports have identified that variability in the gene that encodes the melanoma differentiation-associated protein (MDA)-5 and other molecules in this pathway correlated with the risk of autoimmune diseases. The evidence for MDA5 activity as a cause of autoimmune disease is discussed.


Subject(s)
Autoimmune Diseases/metabolism , Interferon-Induced Helicase, IFIH1/physiology , Animals , Disease Models, Animal , Humans , Immunity, Innate/immunology , Interferons/metabolism , Viruses/immunology
8.
EMBO J ; 36(18): 2742-2757, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28851763

ABSTRACT

Melanoma differentiation-associated protein 5 (MDA5) mediates the innate immune response to viral infection. Polymorphisms in IFIH1, the gene coding for MDA5, correlate with the risk of developing type 1 diabetes (T1D). Here, we demonstrate that MDA5 is crucial for the immune response to enteric rotavirus infection, a proposed etiological agent for T1D. MDA5 variants encoded by minor IFIH1 alleles associated with lower T1D risk exhibit reduced activity against rotavirus infection. We find that MDA5 activity limits rotavirus infection not only through the induction of antiviral interferons and pro-inflammatory cytokines, but also by promoting cell death. Importantly, this MDA5-dependent antiviral response is specific to the pancreas of rotavirus-infected mice, similar to the autoimmunity associated with T1D. These findings imply that MDA5-induced cell death and inflammation in the pancreas facilitate progression to autoimmune destruction of pancreatic ß-cells.


Subject(s)
Cell Death , Host-Pathogen Interactions , Interferon-Induced Helicase, IFIH1/metabolism , Pancreas/pathology , Rotavirus Infections/immunology , Rotavirus Infections/pathology , Rotavirus/pathogenicity , Animals , Cells, Cultured , Inflammation/pathology , Mice
9.
Sci Rep ; 7: 44340, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281686

ABSTRACT

The central role of protein kinases in controlling disease processes has spurred efforts to develop pharmaceutical regulators of their activity. A rational strategy to achieve this end is to determine intrinsic auto-regulatory processes, then selectively target these different states of kinases to repress their activation. Here we investigate auto-regulation of the innate immune effector protein kinase R, which phosphorylates the eukaryotic initiation factor 2α to inhibit global protein translation. We demonstrate that protein kinase R activity is controlled by auto-inhibition via an intra-molecular interaction. Part of this mechanism of control had previously been reported, but was then controverted. We account for the discrepancy and extend our understanding of the auto-inhibitory mechanism by identifying that auto-inhibition is paradoxically instigated by incipient auto-phosphorylation. Phosphor-residues at the amino-terminus instigate an intra-molecular interaction that enlists both of the N-terminal RNA-binding motifs of the protein with separate surfaces of the C-terminal kinase domain, to co-operatively inhibit kinase activation. These findings identify an innovative mechanism to control kinase activity, providing insight for strategies to better regulate kinase activity.


Subject(s)
Eukaryotic Initiation Factor-2/chemistry , Recombinant Fusion Proteins/chemistry , eIF-2 Kinase/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/immunology , Eukaryotic Initiation Factor-2/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Immunity, Innate , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
10.
Cell Res ; 26(3): 367-79, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26794869

ABSTRACT

The protein kinase R (PKR) functions in the antiviral response by controlling protein translation and inflammatory cell signaling pathways. We generated a transgenic, knock-in mouse in which the endogenous PKR is expressed with a point mutation that ablates its kinase activity. This novel animal allows us to probe the kinase-dependent and -independent functions of PKR. We used this animal together with a previously generated transgenic mouse that is ablated for PKR expression to determine the role of PKR in regulating the activity of the cryopyrin inflammasome. Our data demonstrate that, in contradiction to earlier reports, PKR represses cryopyrin inflammasome activity. We demonstrate that this control is mediated through the established function of PKR to inhibit protein translation of constituents of the inflammasome to prevent initial priming during innate immune signaling. These findings identify an important role for PKR to dampen inflammation during the innate immune response and caution against the previously proposed therapeutic strategy to inhibit PKR to treat inflammation.


Subject(s)
Inflammasomes/antagonists & inhibitors , eIF-2 Kinase/metabolism , Animals , Inflammasomes/metabolism , Interleukin-18/biosynthesis , Interleukin-1beta/biosynthesis , Macrophages/enzymology , Macrophages/immunology , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics
11.
FEBS J ; 282(24): 4766-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26414443

ABSTRACT

The murine double-stranded RNA-binding protein termed protein kinase R (PKR)-associated protein X (RAX) and the human homolog, protein activator of PKR (PACT), were originally characterized as activators of PKR. Mice deficient in RAX show reproductive and developmental defects, including reduced body size, craniofacial defects and anterior pituitary hypoplasia. As these defects are not observed in PKR-deficient mice, the phenotype has been attributed to PKR-independent activities of RAX. Here we further investigated the involvement of PKR in the physiological function of RAX, by generating rax(-/-) mice deficient in PKR, or carrying a kinase-inactive mutant of PKR (K271R) or an unphosphorylatable mutant of the PKR substrate eukaryotic translation initiation factor 2 α subunit (eIF2α) (S51A). Ablating PKR expression rescued the developmental and reproductive deficiencies in rax(-/-) mice. Generating rax(-/-) mice with a kinase-inactive mutant of PKR resulted in similar rescue, confirming that the rax(-/-) defects are PKR dependent; specifically that the kinase activity of PKR was required for these defects. Moreover, generating rax(-/-) mice that were heterozygous for an unphosphorylatable mutant eIF2α provides partial rescue of the rax(-/-) defect, consistent with mutation of one copy of the Eif2s1 gene. These observations were further investigated in vitro by reducing RAX expression in anterior pituitary cells, resulting in increased PKR activity and induction of the PKR-regulated cyclin-dependent kinase inhibitor p21(WAF1/CIP1). These results demonstrate that PKR kinase activity is required for onset of the rax(-/-) phenotype, implying an unexpected function for RAX as a negative regulator of PKR in the context of postnatal anterior pituitary tissue, and identify a critical role for the regulation of PKR activity for normal development.


Subject(s)
Pituitary Gland, Anterior/metabolism , RNA-Binding Proteins/metabolism , eIF-2 Kinase/antagonists & inhibitors , Amino Acid Substitution , Animals , Cell Cycle , Cell Line , Cell Proliferation , Crosses, Genetic , Enzyme Activation , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Organ Size , Phosphorylation , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/enzymology , Pituitary Gland, Anterior/growth & development , Protein Processing, Post-Translational , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
12.
Nat Commun ; 6: 6795, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25865065

ABSTRACT

To date, the activities of protein kinases have formed the core of our understanding of cell signal transduction. Comprehension of the extent of protein acetylation has raised expectations that this alternate post-transcriptional modification will be shown to rival phosphorylation in its importance in mediating cellular responses. However, limited instances have been identified. Here we show that signalling from Toll-like or TNF-α receptors triggers the calcium/calmodulin-dependent protein kinase (CaMK2) to activate histone acetyltransferase-1 (HAT1), which then acetylates the transcriptional regulator PLZF. Acetylation of PLZF promotes the assembly of a repressor complex incorporating HDAC3 and the NF-κB p50 subunit that limits the NF-κB response. Accordingly, diminishing the activity of CaMK2, the expression levels of PLZF or HAT1, or mutating key residues that are covalently modified in PLZF and HAT1, curtails control of the production of inflammatory cytokines. These results identify a central role for acetylation in controlling the inflammatory NF-κB transcriptional programme.


Subject(s)
Histone Acetyltransferases/genetics , Kruppel-Like Transcription Factors/genetics , NF-kappa B/genetics , Protein Processing, Post-Translational , Transcription, Genetic , Acetylation , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/immunology , Histone Acetyltransferases/immunology , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Immunity, Innate , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/immunology , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NF-kappa B/immunology , Promyelocytic Leukemia Zinc Finger Protein , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction
13.
Proc Natl Acad Sci U S A ; 112(5): 1535-40, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605927

ABSTRACT

Inflammation is critical for host defense, but without appropriate control, it can cause chronic disease or even provoke fatal responses. Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like receptors, we identify that the Broad-complex, Tramtrack and Bric-a-brac/poxvirus and zinc finger (BTB/POZ), transcriptional regulator promyelocytic leukemia zinc finger (PLZF) limits the expression of inflammatory gene products. In accord with this finding, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated inflammatory responses to infectious stimuli. Temporal quantitation of inflammatory gene transcripts shows increased gene induction in the absence of PLZF. Genome-wide analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune homeostasis and limit damaging inflammation. We show that PLZF stabilizes a corepressor complex that encompasses histone deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while curbing damaging inflammation.


Subject(s)
Chromatin/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Signal Transduction , Animals , Bacterial Infections/metabolism , Chromatin Immunoprecipitation , Fluorescence Resonance Energy Transfer , Histone Deacetylases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Real-Time Polymerase Chain Reaction
14.
J Mol Cell Biol ; 6(6): 473-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25404612

ABSTRACT

A key control point in gene expression is the initiation of protein translation, with a universal stress response being constituted by inhibitory phosphorylation of the eukaryotic initiation factor 2α (eIF2α). In humans, four kinases sense diverse physiological stresses to regulate eIF2α to control cell differentiation, adaptation, and survival. Here we develop a computational molecular model of eIF2α and one of its kinases, the protein kinase R, to simulate the dynamics of their interaction. Predictions generated by coarse-grained dynamics simulations suggest a novel mode of action. Experimentation substantiates these predictions, identifying a previously unrecognized interface in the protein complex, which is constituted by dynamic residues in both eIF2α and its kinases that are crucial to regulate protein translation. These findings call for a reinterpretation of the current mechanism of action of the eIF2α kinases and demonstrate the value of conducting computational analysis to evaluate protein function.


Subject(s)
Eukaryotic Initiation Factor-2 , Molecular Dynamics Simulation , Protein Biosynthesis/physiology , eIF-2 Kinase , Eukaryotic Initiation Factor-2/chemistry , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , eIF-2 Kinase/chemistry , eIF-2 Kinase/metabolism
15.
EMBO Rep ; 14(9): 837-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877428

ABSTRACT

Here we report that ILK localizes in the mouse primary cilium, a sensory organelle required for signalling by the Hedgehog (Hh) pathway. Genetic or pharmacological inhibition of ILK blocks ciliary accumulation of the Hh pathway effector smoothened (Smo) and suppresses the induction of Gli transcription factor mRNAs by SHh. Conditional deletion of ILK or Smo also inhibits SHh-driven activation of Gli2 in the embryonic mouse cerebellum. ILK regulation of Hh signalling probably requires the physical interaction of ILK and Smo in the cilium, and we also show selective cilia-associated interaction of ILK with ß-arrestin, a known mediator of Smo-dependent signalling.


Subject(s)
Cerebellum/metabolism , Cilia/metabolism , Hedgehog Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Arrestins/metabolism , Cell Line , Cerebellum/embryology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Zinc Finger Protein Gli2
16.
Cancer Res ; 73(12): 3625-37, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23536558

ABSTRACT

Bladder cancer is associated with high recurrence and mortality rates due to metastasis. The elucidation of metastasis suppressors may offer therapeutic opportunities if their mechanisms of action can be elucidated and tractably exploited. In this study, we investigated the clinical and functional significance of the transcription factor activating transcription factor 3 (ATF3) in bladder cancer metastasis. Gene expression analysis revealed that decreased ATF3 was associated with bladder cancer progression and reduced survival of patients with bladder cancer. Correspondingly, ATF3 overexpression in highly metastatic bladder cancer cells decreased migration in vitro and experimental metastasis in vivo. Conversely, ATF3 silencing increased the migration of bladder cancer cells with limited metastatic capability in the absence of any effect on proliferation. In keeping with their increased motility, metastatic bladder cancer cells had increased numbers of actin filaments. Moreover, ATF3 expression correlated with expression of the actin filament severing protein gelsolin (GSN). Mechanistic studies revealed that ATF3 upregulated GSN, whereas ATF3 silencing reduced GSN levels, concomitant with alterations in the actin cytoskeleton. We identified six ATF3 regulatory elements in the first intron of the GSN gene confirmed by chromatin immunoprecipitation analysis. Critically, GSN expression reversed the metastatic capacity of bladder cancer cells with diminished levels of ATF3. Taken together, our results indicate that ATF3 suppresses metastasis of bladder cancer cells, at least in part through the upregulation of GSN-mediated actin remodeling. These findings suggest ATF3 coupled with GSN as prognostic markers for bladder cancer metastasis.


Subject(s)
Actin Cytoskeleton/metabolism , Activating Transcription Factor 3/metabolism , Gelsolin/metabolism , Urinary Bladder Neoplasms/metabolism , Activating Transcription Factor 3/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cells, Cultured , Female , Gelsolin/genetics , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Microscopy, Fluorescence , Middle Aged , RNA Interference , Response Elements/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
17.
Nucleic Acids Res ; 41(5): 3436-45, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23325848

ABSTRACT

The retinoic acid inducible gene-I (RIG-I)-like family of receptors is positioned at the front line of our innate cellular defence system. RIG-I detects and binds to foreign duplex RNA in the cytoplasm of both immune and non-immune cells, and initiates the induction of type I interferons and pro-inflammatory cytokines. The mechanism of RIG-I activation by double-stranded RNA (dsRNA) involves a molecular rearrangement proposed to expose the N-terminal pair of caspase activation recruitment domains, enabling an interaction with interferon-beta promoter stimulator 1 (IPS-1) and thereby initiating downstream signalling. dsRNA is particularly stimulatory when longer than 20 bp, potentially through allowing binding of more than one RIG-I molecule. Here, we characterize full-length RIG-I and RIG-I subdomains combined with a stimulatory 29mer dsRNA using multi-angle light scattering and size-exclusion chromatography-coupled small-angle X-ray scattering, to build up a molecular model of RIG-I before and after the formation of a 2:1 protein:dsRNA assembly. We report the small-angle X-ray scattering-derived solution structure of the human apo-RIG-I and observe that on binding of RIG-I to dsRNA in a 2:1 ratio, the complex becomes highly extended and flexible. Hence, here we present the first model of the fully activated oligomeric RIG-I.


Subject(s)
Apoproteins/chemistry , DEAD-box RNA Helicases/chemistry , RNA, Double-Stranded/chemistry , Chromatography, Gel , DEAD Box Protein 58 , Humans , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis , Receptors, Immunologic , Scattering, Small Angle , Trypsin/chemistry , X-Ray Diffraction
18.
Immunity ; 36(5): 795-806, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22633459

ABSTRACT

Primary resistance to pathogens is reliant on both basal and inducible immune defenses. To date, research has focused upon inducible innate immune responses. In contrast to resistance via cytokine induction, basal defense mechanisms are less evident. Here we showed that the antiviral protein kinase R (PKR) inhibited the key actin-modifying protein gelsolin to regulate actin dynamics and control cytoskeletal cellular functions under homeostatic conditions. Through this mechanism, PKR controlled fundamental innate immune, actin-dependent processes that included membrane ruffling and particle engulfment. Accordingly, PKR counteracted viral entry into the cell. These findings identify a layer of host resistance, showing that the regulation of actin-modifying proteins during the innate immune response bolsters first-line defense against intracellular pathogens and has a sustained effect on virus production. Moreover, these data provide proof of principle for a concept in which the cell cytoskeleton could be targeted to elicit broad antiviral protection.


Subject(s)
Actins/metabolism , Gelsolin/metabolism , Immunity, Innate/immunology , eIF-2 Kinase/metabolism , Actins/immunology , Cell Line, Transformed , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Cytoskeleton/immunology , Cytoskeleton/metabolism , Gelsolin/antagonists & inhibitors , Gelsolin/immunology , HEK293 Cells , HeLa Cells , Humans , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Protein Interaction Domains and Motifs/immunology , Viruses/immunology , Viruses/metabolism , eIF-2 Kinase/immunology
19.
Immunity ; 35(4): 491-3, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22035841

ABSTRACT

The crystal structure of the interferon-induced member of the dynamin family MxA presented by Gao et al. (2011) in this issue of Immunity reveals the molecule's higher-order structure, thereby providing insight into the protein's antiviral action as a molecular machine.

20.
J Immunol ; 185(10): 6013-22, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20956347

ABSTRACT

The initiation of antitumor immunity relies on dendritic cells (DCs) to cross-present cell-associated tumor Ag to CD8(+) T cells (T(CD8+)) due to a lack of costimulatory molecules on tumor cells. Innate danger signals have been demonstrated to enhance cross-priming of T(CD8+) to soluble as well as virally encoded Ags; however, their effect on enhancing T(CD8+) cross-priming to cell genome-encoded Ags remains unknown. Furthermore, influenza A virus (IAV) has not been shown to enhance antitumor immunity. Using influenza-infected allogeneic cell lines, we show in this study that T(CD8+) responses to cell-associated Ags can be dramatically enhanced due to enhanced T(CD8+) expansion. This enhanced cross-priming in part involves TLR7- but not TLR3-mediated sensing of IAV and is entirely dependent on MyD88 and IFN signaling pathways. We also showed that the inflammasome-induced IL-1 and IFN-γ did not play a role in enhancing cross-priming in our system. We further demonstrated in our ex vivo system that CD8(+) DCs are the only APCs able to prime TCR-transgenic T(CD8+). Importantly, plasmacytoid DCs and CD8(-) DCs were both able to enhance such priming when provided in coculture. These observations suggest that IAV infection of tumor cells may facilitate improved cross-presentation of tumor Ags and may be used to augment clinical vaccine efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cross-Priming/immunology , Interferon Type I/immunology , Membrane Glycoproteins/immunology , Orthomyxoviridae Infections/immunology , Toll-Like Receptor 7/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Influenza A virus/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Polymerase Chain Reaction , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...