Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Nat Commun ; 12(1): 4105, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215755

ABSTRACT

CCCH zinc finger proteins resolve immune responses by degrading the mRNAs of inflammatory cytokines such as tumor necrosis factor (TNF) and interleukin (IL)-6. Here we report that one such family member, monocyte chemotactic protein-induced protein 3 (MCPIP3, also named ZC3H12C or Regnase-3), promotes skin inflammation by simultaneously enhancing TNF in macrophages and repressing IL-6 in plasmacytoid dendritic cells (pDCs). MCPIP3 is positively associated with psoriasis pathogenesis, and highly expressed by macrophages and pDCs. MCPIP3-deficient macrophages produce less TNF and IL-12p40. However, MCPIP3-deficient pDCs secrete significantly more IL-6. This enhanced intradermal IL-6 may alleviate imiquimod-induced skin inflammation. As a result, MCPIP3-deficient mice are protected from imiquimod-induced psoriasiform lesions. Furthermore, early exposure to pDC-derived IL-6 suppresses macrophage-derived TNF and IL-12p40. Mechanistically, MCPIP3 could directly degrade mRNAs of IL-6, Regnase-1, and IκBζ. In turn, Regnase-1 could degrade MCPIP3 mRNAs. Our study identifies a critical post-transcriptional mechanism that synchronizes myeloid cytokine secretion to initiate autoimmune skin inflammation.


Subject(s)
Cell Cycle Proteins/metabolism , Cytokines/metabolism , Dermatitis/metabolism , Endoribonucleases/metabolism , Inflammation/metabolism , Myeloid Cells/metabolism , Ribonucleases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chemokine CCL2 , Dendritic Cells , Endoribonucleases/deficiency , Endoribonucleases/genetics , Epigenomics , Humans , Imiquimod , Inflammation/pathology , Interleukin-6/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Psoriasis , Ribonucleases/deficiency , Ribonucleases/genetics , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism
2.
Cancer Sci ; 112(7): 2739-2752, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33939241

ABSTRACT

DNA damage induces transcriptional repression of E2F1 target genes and a reduction in histone H3-Thr11 phosphorylation (H3-pThr11 ) at E2F1 target gene promoters. Dephosphorylation of H3-pThr11 is partly mediated by Chk1 kinase and protein phosphatase 1γ (PP1γ) phosphatase. Here, we isolated NIPP1 as a regulator of PP1γ-mediated H3-pThr11 by surveying nearly 200 PP1 interactor proteins. We found that NIPP1 inhibits PP1γ-mediated dephosphorylation of H3-pThr11 both in vivo and in vitro. By generating NIPP1-depleted cells, we showed that NIPP1 is required for cell proliferation and the expression of E2F1 target genes. Upon DNA damage, activated protein kinase A (PKA) phosphorylated the NIPP1-Ser199 residue, adjacent to the PP1 binding motif (RVxF), and triggered the dissociation of NIPP1 from PP1γ, leading to the activation of PP1γ. Furthermore, the inhibition of PKA activity led to the activation of E2F target genes. Statistical analysis confirmed that the expression of NIPP1 was positively correlated with E2F target genes. Taken together, these findings demonstrate that the PP1 regulatory subunit NIPP1 modulates E2F1 target genes by linking PKA and PP1γ during DNA damage.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Damage , E2F1 Transcription Factor/genetics , Endoribonucleases/metabolism , Histones/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 1/metabolism , RNA-Binding Proteins/metabolism , CRISPR-Cas Systems , Cell Proliferation , Cells, Cultured , Checkpoint Kinase 1/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endoribonucleases/deficiency , Endoribonucleases/isolation & purification , Epigenetic Repression , Gene Expression Regulation , Humans , Phosphoprotein Phosphatases/deficiency , Phosphoprotein Phosphatases/isolation & purification , Phosphorylation , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Messenger/metabolism , RNA-Binding Proteins/isolation & purification , Receptors, Neuropeptide Y/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Ultraviolet Rays
3.
PLoS One ; 16(2): e0247232, 2021.
Article in English | MEDLINE | ID: mdl-33600503

ABSTRACT

The appropriate regulation of T lymphocyte functions is key to achieve protective immune responses, while at the same time limiting the risks of tissue damage and chronic inflammation. Deciphering the mechanisms underpinning T cell responses in humans may therefore be beneficial for a range of infectious and chronic diseases. Recently, the development of methods based on CRISPR-Cas9 gene-editing has greatly expanded the available tool-box for the mechanistic studies of primary human T cell responses. While the deletion of a surface protein has become a relatively straightforward task, as long as an antibody for detection is available, the identification and selection of cells lacking an intracellular protein, a non-coding RNA or a protein for which no antibody is available, remain more problematic. Here, we discuss the options currently available to scientists interested in performing gene-editing in primary human T lymphocytes and we describe the optimization of a workflow for the screening and analysis of lymphocytes following gene-editing with CRISPR-Cas9 based on T cell cloning and T7 endonuclease I cleavage assay.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Membrane Proteins/genetics , Transcription Factors/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cells, Cultured , Endoribonucleases/deficiency , Endoribonucleases/genetics , Humans , Membrane Proteins/deficiency , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription Factors/deficiency
4.
Mol Cell Biochem ; 465(1-2): 53-64, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31834612

ABSTRACT

IRE1 is the most conserved endoplasmic reticulum (ER)-resident stress sensor. Its activation not only splices XBP1 but also participates in a variety of cell signaling. We elucidated the role of IRE1α in Neuro2a cells by establishing IRE1α-deficient cells and applying four IRE1 inhibitors. IRE1α deficiency prevented almost all spliced XBP1 (sXBP1) protein expression by treatment with thapsigargin (Tg) and tunicamycin (Tm); these phenomena paralleled the values measured by our two Nanoluciferase-based IRE1 assays. However, cell viability and protein expression of other ER stress-responsive factors in the IRE1α-deficient cells were comparable to those in the parental wild-type cells with or without Tm treatment. Next, we elucidated the IRE1 inhibitory actions and cytotoxicity of four compounds: STF083010, KIRA6, 4µ8C, and toyocamycin. KIRA6 attenuated IRE1 activity in a dose-dependent manner, but it showed severe cytotoxicity even in the IRE1α-deficient cells at a low concentration. The IRE1α-deficient cells were slightly resistant to KIRA6 at 0.1 µM in both the presence and absence of ER stress; however, resistance was not observed at 0.02 µM. Treatment with only KIRA6 at 0.1 µM for 12 h remarkably induced LC3 II, an autophagic marker, in both parental and IRE1α-deficient cells. Co-treatment with KIRA6 and Tm induced LC3 II, cleaved caspase-9, and cleaved caspase-3; however, IRE1α-deficiency did not abolish the expression of these two cleaved caspases. On the other hand, KIRA6 prohibited Tm-induced ATF4 induction in an IRE1-independent manner; however, co-treatment with KIRA6 and Tm also induced LC3 II and two cleaved caspases in the ATF4-deficient Neuro2a cells. Thus, we demonstrate that IRE1α deficiency has little impact on cell viability and expression of ER stress-responsive factors in Neuro2a cells, and the pharmacological actions of KIRA6 include IRE1-independent ways.


Subject(s)
CRISPR-Cas Systems , Cytotoxins/pharmacology , Endoribonucleases/deficiency , Gene Deletion , Gene Expression Regulation/drug effects , Microtubule-Associated Proteins , Protein Serine-Threonine Kinases/deficiency , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Mice
5.
Endocr Regul ; 53(4): 250-262, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31734650

ABSTRACT

OBJECTIVE: The aim of the present investigation was to study the effect of hypoxia on the expression of genes encoding endothelin-1 (EDN1) and its cognate receptors (EDNRA and EDNRB) as well as endothelin converting enzyme 1 (ECE1) in U87 glioma cells in response to inhibition of endoplasmic reticulum stress signaling mediated by ERN1/IRE1 (endoplasmic reticulum to nucleus signaling 1) for evaluation of their possible significance in the control of glioma growth through ERN1 and hypoxia. METHODS: The expression level of EDN1, EDNRA, EDNRB, and ECE1 genes as well as micro-RNA miR-19, miR-96, and miR-206 was studied in control and ERN1 knockdown U87 glioma cells under hypoxia by quantitative polymerase chain reaction. RESULTS: It was shown that the expression level of EDN1, EDNRA, EDNRB, and ECE1 genes was up-regulated in ERN1 knockdown glioma cells in comparison with the control glioma cells, being more significant for endothelin-1. We also observed down-regulation of microRNA miR-206, miR-96, and miR-19a, which have specific binding sites in mRNA EDN1, EDNRA, and EDNRB, correspondingly, and can participate in posttranscriptional regulation of these mRNA expressions. Furthermore, inhibition of ERN1 endoribonuclease lead to up-regulation of EDNRA and ECE1 gene expressions and down-regulation of the expression level of EDN1 and EDNRB genes in glioma cells. Thus, the expression of EDNRA and ECE1 genes is regulated by ERN1 endoribonuclease, but EDN1 and EDNRB genes preferentially by ERN1 protein kinase. We have also shown that hypoxia enhanced the expression of EDN1, EDNRA, and ECE1 genes and that knockdown of ERN1 signaling enzyme function significantly modified the response of all studied gene expressions to hypoxia. Thus, effect of hypoxia on the expression level of EDN1 and ECE1 genes was significantly or completely reduced in ERN1 knockdown glioma cells since the expression of EDNRA gene was down-regulated under hypoxia. Moreover, hypoxia is induced the expression of EDNRB gene in ERN1 knockdown glioma cells. CONCLUSIONS: Results of this investigation demonstrate that ERN1 knockdown significantly increased the expression of endothelin-1 and its receptors as well as ECE1 genes by different mechanisms and that all studied gene expressions were sensitive to hypoxia. It is possible that hypoxic regulation of the expression of these genes is a result of complex interaction of variable ERN1 related transcription and regulatory factors with HIF1A and possibly contributed to the control of glioma growth.


Subject(s)
Brain Neoplasms/genetics , Endoribonucleases/genetics , Glioma/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Hypoxia/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Endoribonucleases/deficiency , Endothelin-1/genetics , Endothelin-Converting Enzymes/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/metabolism , Glioma/pathology , Humans , Hypoxia/genetics , Hypoxia/pathology , Protein Serine-Threonine Kinases/deficiency , Receptor, Endothelin A/genetics , Receptor, Endothelin B/genetics
6.
Endocr Regul ; 53(4): 237-249, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31734653

ABSTRACT

OBJECTIVE: The aim of the present study was to examine the effect of glucose deprivation on the expression of genes encoded glucocorticoid receptor (NR3C1) and some related proteins (NR3C2, AHR, NRIP1, NNT, ARHGAP35, SGK1, and SGK3) in U87 glioma cells in response to inhibition of endoplasmic reticulum stress signaling mediated by ERN1/IRE1 (endoplasmic reticulum to nucleus signaling 1/inositol requiring enzyme 1) for evaluation of their possible significance in the control of glioma growth through endoplasmic reticulum stress signaling mediated by IRE1 and glucose deprivation. METHODS: The expression of NR3C1, NR3C2, AHR, NRIP1, NNT, ARHGAP35, SGK1, and SGK3 genes in U87 glioma cells transfected by empty vector pcDNA3.1 (control cells) and cells without ERN1 signaling enzyme function (transfected by dnERN1) under glucose deprivation was studied by real time quantitative polymerase chain reaction. RESULTS: It was shown that the expression level of NR3C2, AHR, SGK1, SGK3, and NNT genes was up-regulated in control U87 glioma cells under glucose deprivation condition in comparison with the control cells growing with glucose. At the same time, the expression of NRIP1 gene is down-regulated in these glioma cells under glucose deprivation, but NR3C1 and ARHGAP35 genes was resistant to this experimental condition. We also showed that inhibition of ERN1 signaling enzyme function significantly modified the response of most studied gene expressions to glucose deprivation condition. Thus, effect of glucose deprivation on the expression level of NR3C2, AHR, and SGK1 genes was significantly stronger in ERN1 knockdown U87 glioma cells since the expression of NNT gene was resistant to glucose deprivation condition. Moreover, the inhibition of ERN1 enzymatic activities in U87 glioma cells led to up-regulation of ARHGAP35 gene expression and significant down-regulation of the expression of SGK3 gene in response to glucose deprivation condition. CONCLUSIONS: Results of this study demonstrated that glucose deprivation did not change the expression level of NR3C1 gene but it significantly affected the expression of NR3C2, AHR, NRIP, SGK1, SGK3, and NNT genes in vector-transfected U87 glioma cells in gene specific manner and possibly contributed to the control of glioma growth since the expression of most studied genes in glucose deprivation condition was significantly dependent on the functional activity of IRE1 signaling enzyme.


Subject(s)
Brain Neoplasms/genetics , Endoribonucleases/genetics , Glioma/genetics , Glucose/deficiency , Protein Serine-Threonine Kinases/genetics , Receptors, Glucocorticoid/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endoribonucleases/deficiency , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glioma/pathology , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/genetics , Humans , Immediate-Early Proteins/genetics , Mitochondrial Proteins/genetics , NADP Transhydrogenase, AB-Specific/genetics , Nuclear Receptor Interacting Protein 1/genetics , Protein Serine-Threonine Kinases/deficiency , Repressor Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Cell ; 179(6): 1264-1275.e13, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31778653

ABSTRACT

TLR8 is among the highest-expressed pattern-recognition receptors in the human myeloid compartment, yet its mode of action is poorly understood. TLR8 engages two distinct ligand binding sites to sense RNA degradation products, although it remains unclear how these ligands are formed in cellulo in the context of complex RNA molecule sensing. Here, we identified the lysosomal endoribonuclease RNase T2 as a non-redundant upstream component of TLR8-dependent RNA recognition. RNase T2 activity is required for rendering complex single-stranded, exogenous RNA molecules detectable for TLR8. This is due to RNase T2's preferential cleavage of single-stranded RNA molecules between purine and uridine residues, which critically contributes to the supply of catabolic uridine and the generation of purine-2',3'-cyclophosphate-terminated oligoribonucleotides. Thus-generated molecules constitute agonistic ligands for the first and second binding pocket of TLR8. Together, these results establish the identity and origin of the RNA-derived molecular pattern sensed by TLR8.


Subject(s)
Endoribonucleases/metabolism , Proteolysis , Toll-Like Receptor 8/metabolism , Amino Acid Motifs , Base Sequence , Cell Line , Endoribonucleases/deficiency , Humans , Models, Molecular , Monocytes/metabolism , Myeloid Cells/metabolism , Nitrogen Isotopes , Oligonucleotides/metabolism , Purines/metabolism , RNA/metabolism , Staphylococcus aureus/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/chemistry , Uridine/metabolism
9.
J Bacteriol ; 201(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30885931

ABSTRACT

Riboregulation involving regulatory RNAs, RNA chaperones, and ribonucleases is fundamental for the rapid adaptation of gene expression to changing environmental conditions. The gene coding for the RNase YbeY belongs to the minimal prokaryotic genome set and has a profound impact on physiology in a wide range of bacteria. Here, we show that the Agrobacterium tumefaciensybeY gene is not essential. Deletion of the gene in the plant pathogen reduced growth, motility, and stress tolerance. Most interestingly, YbeY is crucial for A. tumefaciens-mediated T-DNA transfer and tumor formation. Comparative proteomics by using isobaric tags for relative and absolute quantitation (iTRAQ) revealed dysregulation of 59 proteins, many of which have previously been found to be dependent on the RNA chaperone Hfq. YbeY and Hfq have opposing effects on production of these proteins. Accumulation of a 16S rRNA precursor in the ybeY mutant suggests that A. tumefaciens YbeY is involved in rRNA processing. RNA coimmunoprecipitation-sequencing (RIP-Seq) showed binding of YbeY to the region immediately upstream of the 16S rRNA. Purified YbeY is an oligomer with RNase activity. It does not physically interact with Hfq and thus plays a partially overlapping but distinct role in the riboregulatory network of the plant pathogen.IMPORTANCE Although ybeY gene belongs to the universal bacterial core genome, its biological function is incompletely understood. Here, we show that YbeY is critical for fitness and host-microbe interaction in the plant pathogen Agrobacterium tumefaciens Consistent with the reported endoribonuclease activity of YbeY, A. tumefaciens YbeY acts as a RNase involved in maturation of 16S rRNA. This report adds a worldwide plant pathogen and natural genetic engineer of plants to the growing list of bacteria that require the conserved YbeY protein for host-microbe interaction.


Subject(s)
Agrobacterium tumefaciens/genetics , DNA, Bacterial/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/genetics , Ribosomes/genetics , Adaptation, Physiological , Agrobacterium tumefaciens/enzymology , Agrobacterium tumefaciens/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , DNA, Bacterial/metabolism , Endoribonucleases/deficiency , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Deletion , Gene Expression Profiling , Host Factor 1 Protein/metabolism , Metalloproteins/genetics , Metalloproteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Binding , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Ribosomes/metabolism , Sequence Homology, Nucleic Acid , Stress, Physiological , Virulence
10.
Nat Cell Biol ; 20(8): 942-953, 2018 08.
Article in English | MEDLINE | ID: mdl-30013108

ABSTRACT

Maintenance of endoplasmic reticulum (ER) proteostasis is controlled by a signalling network known as the unfolded protein response (UPR). Here, we identified filamin A as a major binding partner of the ER stress transducer IRE1α. Filamin A is an actin crosslinking factor involved in cytoskeleton remodelling. We show that IRE1α controls actin cytoskeleton dynamics and affects cell migration upstream of filamin A. The regulation of cytoskeleton dynamics by IRE1α is independent of its canonical role as a UPR mediator, serving instead as a scaffold that recruits and regulates filamin A. Targeting IRE1α expression in mice affected normal brain development, generating a phenotype resembling periventricular heterotopia, a disease linked to the loss of function of filamin A. IRE1α also modulated cell movement and cytoskeleton dynamics in fly and zebrafish models. This study unveils an unanticipated biological function of IRE1α in cell migration, whereby filamin A operates as an interphase between the UPR and the actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Movement , Endoribonucleases/metabolism , Fibroblasts/metabolism , Filamins/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Endoribonucleases/deficiency , Endoribonucleases/genetics , Evolution, Molecular , Female , Filamins/genetics , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Knockout , Neurons/pathology , Periventricular Nodular Heterotopia/genetics , Periventricular Nodular Heterotopia/metabolism , Periventricular Nodular Heterotopia/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Unfolded Protein Response , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
J Med Microbiol ; 67(5): 724-732, 2018 May.
Article in English | MEDLINE | ID: mdl-29620505

ABSTRACT

PURPOSE: In enterohaemorrhagic Escherichia coli (EHEC), stx1 or stx2 genes encode Shiga toxin (Stx1 or Stx2, respectively) and are carried by prophages. The production and release of both stx phages and toxin occur upon initiation of the phage lytic cycle. Phages can further disseminate stx genes by infecting naïve bacteria in the intestine. Here, the effect of RNase E deficiency on these two virulence traits was investigated. METHODOLOGY: Cultures of the EHEC strains TEA028-rne containing low versus normal RNase E levels or the parental strain (TEA028) were treated with mitomycin C (MMC) to induce the phage lytic cycle. Phages and Stx2 titres were quantified by the double-agar assay and the receptor ELISA technique, respectively. RESULTS: RNase E deficiency in MMC-treated cells significantly reduced the yield of infectious stx2 phages. Delayed cell lysis and the appearance of encapsidated phage DNA copies suggest a slow onset of the lytic cycle. However, these observations do not entirely explain the decrease of phage yields. stx1 phages were not detected under normal or deficient RNase E levels. After an initial delay, high levels of toxin were finally produced in MMC-treated cultures. CONCLUSION: RNase E scarcity reduces stx2 phage production but not toxin. Normal concentrations of RNase E are likely required for correct phage morphogenesis. Our future work will address the mechanism of RNase E action on phage morphogenesis.


Subject(s)
Coliphages/growth & development , Endoribonucleases/metabolism , Enterohemorrhagic Escherichia coli/enzymology , Enterohemorrhagic Escherichia coli/virology , Prophages/growth & development , Shiga Toxin 2/biosynthesis , Bacteriolysis , Coliphages/genetics , Endoribonucleases/deficiency , Enzyme-Linked Immunosorbent Assay , Humans , Prophages/genetics , Shiga Toxin 2/analysis , Viral Plaque Assay
12.
J Cell Biol ; 217(4): 1287-1301, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29507125

ABSTRACT

In mammalian pancreatic ß cells, the IRE1α-XBP1 pathway is constitutively and highly activated under physiological conditions. To elucidate the precise role of this pathway, we constructed ß cell-specific Ire1α conditional knockout (CKO) mice and established insulinoma cell lines in which Ire1α was deleted using the Cre-loxP system. Ire1α CKO mice showed the typical diabetic phenotype including impaired glycemic control and defects in insulin biosynthesis postnatally at 4-20 weeks. Ire1α deletion in pancreatic ß cells in mice and insulinoma cells resulted in decreased insulin secretion, decreased insulin and proinsulin contents in cells, and decreased oxidative folding of proinsulin along with decreased expression of five protein disulfide isomerases (PDIs): PDI, PDIR, P5, ERp44, and ERp46. Reconstitution of the IRE1α-XBP1 pathway restored the proinsulin and insulin contents, insulin secretion, and expression of the five PDIs, indicating that IRE1α functions as a key regulator of the induction of catalysts for the oxidative folding of proinsulin in pancreatic ß cells.


Subject(s)
Endoribonucleases/metabolism , Insulin-Secreting Cells/enzymology , Insulin/metabolism , Proinsulin/metabolism , Protein Folding , Protein Serine-Threonine Kinases/metabolism , X-Box Binding Protein 1/metabolism , Activating Transcription Factor 6/metabolism , Animals , Binding Sites , Blood Glucose/metabolism , Cell Line, Tumor , Diabetes Mellitus/blood , Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Endoribonucleases/deficiency , Endoribonucleases/genetics , Insulin/genetics , Insulinoma/enzymology , Insulinoma/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidation-Reduction , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Phosphorylation , Proinsulin/chemistry , Proinsulin/genetics , Promoter Regions, Genetic , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Thioredoxins/genetics , Thioredoxins/metabolism , X-Box Binding Protein 1/genetics , eIF-2 Kinase/metabolism
13.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29263275

ABSTRACT

Oncolytic virotherapy is an emerging treatment modality that uses replication-competent viruses to destroy cancer cells. M1 is a naturally occurring alphavirus (Togaviridae) which shows potent oncolytic activities against many cancers. Accumulation of unfolded proteins during virus replication leads to a transcriptional/translational response known as the unfolded protein response (UPR), which might counteract the antitumor effect of the oncolytic virus. In this report, we show that either pharmacological or biological inhibition of IRE1α or PERK, but not ATF6, substantially increases the oncolytic effects of the M1 virus. Moreover, inhibition of IRE1α blocks M1 virus-induced autophagy, which restricts the antitumor effects of the M1 virus through degradation of viral protein, in glioma cells. In addition, IRE1α suppression significantly increases the oncolytic effect of M1 virus in an orthotopic glioma model. From a molecular pathology study, we found that IRE1α is expressed at lower levels in higher-grade gliomas, suggesting greater antitumor efficacy of the oncolytic virus M1. Taken together, these findings illustrate a defensive mechanism of glioma cells against the oncolytic virus M1 and identify possible approaches to enhance the oncolytic viral protein accumulation and the subsequent lysis of tumor cells.IMPORTANCE Although oncolytic virotherapy is showing great promise in clinical applications, not all patients are benefiting. Identifying inhibitory signals in refractory cancer cells for each oncolytic virus would provide a good chance to increase the therapeutic effect. Here we describe that infection with the oncolytic virus M1 triggers the unfolded protein response (UPR) and subsequent autophagy, while blocking the UPR-autophagy axis significantly potentiates the antitumor efficacy of M1 in vitro and in vivo A survey of cancer tissue banks revealed that IRE1α, a key element in the UPR pathway, is commonly downregulated in higher-grade human gliomas, suggesting favorable prospects for the application of M1. Our work provides a potential predictor and target for enhancement of the therapeutic effectiveness of the M1 virus. We predict that the mechanism-based combination therapy will promote cancer virotherapy in the future.


Subject(s)
Autophagy/immunology , Endoribonucleases/deficiency , Glioma/therapy , Neoplasm Proteins/deficiency , Oncolytic Virotherapy , Oncolytic Viruses , Protein Serine-Threonine Kinases/deficiency , Togaviridae , Animals , Autophagy/genetics , Cell Line, Tumor , Chlorocebus aethiops , Cricetinae , Endoribonucleases/immunology , Female , Glioma/genetics , Glioma/immunology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Unfolded Protein Response/genetics , Unfolded Protein Response/immunology , Vero Cells , Xenograft Model Antitumor Assays
14.
Ukr Biochem J ; 89(1): 38-49, 2017.
Article in English | MEDLINE | ID: mdl-29236388

ABSTRACT

We have studied the effect of hypoxia on the expression level of mRNA of the basic enzymes of pentose-phosphate cycle (G6PD, TKT, TALDO1, PGLS and RPIA) and glucose-6-phosphate isomerase (GPI) in U87 glioma cells in relation to inhibition of IRE1 (inositol requiring enzyme 1). It was shown that hypoxia leads to up-regulation of the expression of GPI and PGLS genes and to down-regulation of TALDO1 and RPIA genes in control glioma cells. Changes for GPI gene were more significant than for other genes. At the same time, inhibition of IRE1 modified the effect of hypoxia on the expression of all studied genes. In particular, it increased sensitivity to hypoxia of G6PD and TKT genes expression and suppressed the effect of hypoxia on the expression of GPI and RPIA genes. Additionally, inhibition of IRE1 eliminated hypoxic regulation of PGLS gene and did not change significantly effect of hypoxia on the expression of TALDO1 gene in glioma cells. Present study demonstrated that hypoxia, which often contributes to tumor growth, affects the expression of most studied genes and inhibition of IRE1 modified the hypoxic regulation of pentose-phosphate cycle gene expressions in a gene specific manner and thus possibly contributes to slower glioma growth, but several aspects of this regulation warrant further investigation.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Neuroglia/metabolism , Pentose Phosphate Pathway/genetics , Protein Serine-Threonine Kinases/genetics , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endoribonucleases/deficiency , Gene Knockdown Techniques , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Humans , Neuroglia/pathology , Protein Serine-Threonine Kinases/deficiency , Signal Transduction , Transaldolase/genetics , Transaldolase/metabolism , Transketolase/genetics , Transketolase/metabolism
15.
FEMS Microbiol Lett ; 364(13)2017 07 06.
Article in English | MEDLINE | ID: mdl-28854682

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is a food-borne pathogen that assembles a type III secretion system (T3SS) on its surface. The last portion of the T3SS, called the 'translocon', is composed of a filament and a pore complex that is inserted into the membrane of intestinal epithelial cells. The genes encoding the translocon (espADB) are part of the LEE4 operon. Their expression is regulated by a complex post-transcriptional mechanism that involves the processing of LEE4 mRNA by the essential endoribonuclease RNase E. Here, we report the construction of an EHEC strain (TEA028-rne) in which RNase E can be induced by adding IPTG to the culture medium. EHEC cells deficient in RNase E displayed an abnormal morphology and slower growth, in agreement with published observations in E. coli K-12. Under those conditions, EspA and EspB were produced at higher concentrations, and protein secretion still occurred. These results indicate that RNase E negatively regulates translocon protein synthesis and demonstrate the utility of E. coli strain TEA028-rne as a tool for investigating the influence of this ribonuclease on EHEC gene expression in vitro.


Subject(s)
Bacterial Outer Membrane Proteins/biosynthesis , Endoribonucleases/deficiency , Escherichia coli O157/metabolism , Escherichia coli Proteins/biosynthesis , Type III Secretion Systems/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , DNA, Bacterial , Endoribonucleases/genetics , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Isopropyl Thiogalactoside/pharmacology , Operon
16.
J Immunol ; 199(7): 2215-2224, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28827284

ABSTRACT

Orosomucoid like 3 (ORMDL3), a gene localized to chromosome 17q21, has been linked in epidemiologic studies to childhood asthma and rhinovirus (RV) infections. As the single nucleotide polymorphisms linking ORMDL3 to asthma are associated with increased expression of ORMDL3, we have used hORMDL3zp3-Cre mice (which have universal increased expression of human ORMDL3) to determine whether infection of these transgenic mice with RV influences levels of airway inflammation or RV viral load. RV infection of hORMDL3zp3-Cre mice resulted in reduced RV viral load assessed by quantitative real-time PCR (lung and airway epithelium), as well as reduced airway inflammation (total bronchoalveolar lavage cells, neutrophils, macrophages, and lymphocytes) compared with RV-infected wild-type mice. Levels of the antiviral pathways including IFNs (IFN-α, IFN-ß, IFN-λ) and RNAse L were significantly increased in the lungs of RV-infected hORMDL3zp3-Cre mice. Levels of the antiviral mouse oligoadenylate synthetase (mOas)1g pathway and RNAse L were upregulated in the lungs of unchallenged hORMDL3zp3-Cre mice. In addition, levels of mOas2, but not mOas1 (mOas1a, mOas1b, mOas1g), or mOas3 pathways were significantly more upregulated by IFNs (IFN-α, IFN-ß, IFN-λ) in epithelial cells from hORMDL3zp3-Cre mice compared with RV-infected wild-type mouse epithelial cells. RNAse L-deficient mice infected with RV had increased RV viral load. Overall, these studies suggest that increased levels of ORMDL3 contribute to antiviral defense to RV infection in mice through pathways that may include IFNs (IFN-α, IFN-ß, IFN-λ), OAS, and RNAse L.


Subject(s)
Lung/virology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Rhinovirus/isolation & purification , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Animals , Asthma/immunology , Asthma/virology , Endoribonucleases/deficiency , Endoribonucleases/genetics , Endoribonucleases/metabolism , Epithelial Cells/virology , Inflammation/immunology , Inflammation/virology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/immunology , Interferons/biosynthesis , Interferons/genetics , Interferons/immunology , Lung/immunology , Mice , Mice, Transgenic , Picornaviridae Infections/metabolism , Real-Time Polymerase Chain Reaction , Viral Load
17.
PLoS One ; 12(5): e0177915, 2017.
Article in English | MEDLINE | ID: mdl-28542621

ABSTRACT

Escherichia coli RNase E (Eco-RNase E), encoded by rne (Eco-rne), is considered the global RNA decay initiator. Although Eco-RNase E is an essential gene product in E. coli, some bacterial species, such as Bacillus subtilis, do not possess Eco-RNase E sequence homologues. B. subtilis instead possesses RNase J1/J2 (Bsu-RNase J1/J2) and RNase Y (Bsu-RNase Y) to execute RNA decay. Here we found that E. coli lacking the Eco-rne gene (Δrne E. coli) was viable conditional on M9 minimal media by introducing Bsu-RNase J1/J2 or Bsu-RNase Y. We also cloned an extremely short Eco-RNase E homologue (Wpi-RNase E) and a canonical sized Bsu-RNase J1/J2 homologue (Wpi-RNase J) from Wolbachia pipientis, an α-proteobacterial endosymbiont of arthropods. We found that Wpi-RNase J restored the colony-forming ability (CFA) of Δrne E. coli, whereas Wpi-RNase E did not. Unexpectedly, Wpi-RNase E restored defective CFA due to lack of Eco-RNase G, a paralogue of Eco-RNase E. Our results indicate that bacterial species that lack Eco-RNase E homologues or bacterial species that possess Eco-RNase E homologues which lack Eco-RNase E-like activities have a modest Eco-RNase E-like function using RNase J and/or RNase Y. These results suggest that Eco-RNase E-like activities might distribute among a wide array of bacteria and that functions of RNases may have changed dynamically during evolutionary divergence of bacterial lineages.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli/genetics , Genetic Engineering , Sequence Homology, Amino Acid , Animals , Computer Simulation , Endoribonucleases/chemistry , Endoribonucleases/deficiency , Endoribonucleases/genetics , Escherichia coli/enzymology , Female , Mutation , Phenotype , Symbiosis , Wolbachia/enzymology
18.
Nature ; 539(7630): 588-592, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27851737

ABSTRACT

Small regulatory RNAs guide Argonaute (Ago) proteins in a sequence-specific manner to their targets and therefore have important roles in eukaryotic gene silencing. Of the three small RNA classes, microRNAs and short interfering RNAs are processed from double-stranded precursors into defined 21- to 23-mers by Dicer, an endoribonuclease with intrinsic ruler function. PIWI-interacting RNAs (piRNAs)-the 22-30-nt-long guides for PIWI-clade Ago proteins that silence transposons in animal gonads-are generated independently of Dicer from single-stranded precursors. piRNA 5' ends are defined either by Zucchini, the Drosophila homologue of mitoPLD-a mitochondria-anchored endonuclease, or by piRNA-guided target cleavage. Formation of piRNA 3' ends is poorly understood. Here we report that two genetically and mechanistically distinct pathways generate piRNA 3' ends in Drosophila. The initiating nucleases are either Zucchini or the PIWI-clade proteins Aubergine (Aub) or Ago3. While Zucchini-mediated cleavages directly define mature piRNA 3' ends, Aub/Ago3-mediated cleavages liberate pre-piRNAs that require extensive resection by the 3'-to-5' exoribonuclease Nibbler (Drosophila homologue of Mut-7). The relative activity of these two pathways dictates the extent to which piRNAs are directed to cytoplasmic or nuclear PIWI-clade proteins and thereby sets the balance between post-transcriptional and transcriptional silencing. Notably, loss of both Zucchini and Nibbler reveals a minimal, Argonaute-driven small RNA biogenesis pathway in which piRNA 5' and 3' ends are directly produced by closely spaced Aub/Ago3-mediated cleavage events. Our data reveal a coherent model for piRNA biogenesis, and should aid the mechanistic dissection of the processes that govern piRNA 3'-end formation.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Animals , Argonaute Proteins/metabolism , Cytoplasm/metabolism , Drosophila Proteins/deficiency , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Endoribonucleases/deficiency , Endoribonucleases/metabolism , Exoribonucleases/deficiency , Exoribonucleases/metabolism , Female , Nuclear Proteins/metabolism , Peptide Initiation Factors/metabolism , RNA Processing, Post-Transcriptional , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Transcription, Genetic
19.
Ukr Biochem J ; 88(2): 25-34, 2016.
Article in English | MEDLINE | ID: mdl-29227599

ABSTRACT

We have studied hypoxic regulation of the expression of genes encoded GADD (growth arrest and DNA damage) family proteins in U87 glioma cells in relation to inhibition of IRE1 (inositol requiring enzyme-1), which controls cell proliferation and tumor growth as a central mediator of endoplasmic reticulum stress. We have shown that hypoxia up-regulates the expression of GADD34, GADD45A, GADD45B, and GADD153 genes, which are related to cell proliferation and apoptosis, in control (transfected by empty vector) glioma cells in gene specific manner. At the same time, the expression level of EIF2AK 1 (eukaryotic translation initiation factor 2-alpha kinase 1) and AI FM1 (apoptosis inducing factor, mitochondria associated 1) genes in these cells is down-regulated upon hypoxic condition. It was also shown that inhibition of ІRE1 signaling enzyme function in U87 glioma cells enhances the effect of hypoxia on these genes expression, except EIF2AK 1 and AI FM1 genes. Furthermore, the expression of all studied genes in ІRE1 knockdown cells is significantly decreased upon normoxic condition, except GADD45B gene, which expression level is strongly up-regulated. Therefore, the expression level of genes encoding GADD34, GADD45A, GADD45B, GADD153, EIF2AK 1, and AI FM1 is affected by hypoxia and by inhibition of IRE1-mediated endoplasmic reticulum stress signaling in gene specific manner and correlates with suppression of glioma cell proliferation upon inhibition of the IRE1 enzyme function.


Subject(s)
Cell Cycle Proteins/genetics , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Neuroglia/metabolism , Nuclear Proteins/genetics , Protein Phosphatase 1/genetics , Protein Serine-Threonine Kinases/genetics , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Apoptosis/genetics , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Cell Cycle Proteins/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Endoribonucleases/deficiency , Gene Knockdown Techniques , Humans , Neuroglia/pathology , Nuclear Proteins/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Phosphatase 1/metabolism , Protein Serine-Threonine Kinases/deficiency , Signal Transduction , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transfection , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
20.
Ukr Biochem J ; 88(3): 54-65, 2016.
Article in English | MEDLINE | ID: mdl-29235326

ABSTRACT

We have studied the effect of hypoxia on the expression of nuclear genes encoding mitochondrial proteins in U87 glioma cells under the inhibition of IRE1 (inositol requiring enzyme-1), which controls cell proliferation and tumor growth as a central mediator of endoplasmic reticulum stress. It was shown that hypoxia down-regulated gene expression of malate dehydrogenase 2 (MDH2), malic enzyme 2 (ME2), mitochondrial aspartate aminotransferase (GOT2), and subunit B of succinate dehydrogenase (SDHB) in control (transfected by empty vector) glioma cells in a gene specific manner. At the same time, the expression level of mitochondrial NADP+-dependent isocitrate dehydrogenase 2 (IDH2) and subunit D of succinate dehydrogenase (SDHD) genes in these cells does not significantly change in hypoxic conditions. It was also shown that the inhibition of ІRE1 signaling enzyme function in U87 glioma cells decreases the effect of hypoxia on the expression of ME2, GOT2, and SDHB genes and introduces the sensitivity of IDH2 gene to hypoxia. Furthermore, the expression of all studied genes depends on IRE1-mediated endoplasmic reticulum stress signaling in gene specific manner, because ІRE1 knockdown significantly decreases their expression in normoxic conditions, except for IDH2 gene, which expression level is strongly up-regulated. Therefore, changes in the expression level of nuclear genes encoding ME2, MDH2, IDH2, SDHB, SDHD, and GOT2 proteins possibly reflect metabolic reprogramming of mitochondria by hypoxia and IRE1-mediated endoplasmic reticulum stress signaling and correlate with suppression of glioma cell proliferation under inhibition of the IRE1 enzyme function.


Subject(s)
Cell Nucleus/genetics , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Mitochondria/genetics , Neuroglia/enzymology , Protein Serine-Threonine Kinases/genetics , Aspartate Aminotransferases/genetics , Aspartate Aminotransferases/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Proliferation , Endoribonucleases/deficiency , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Malate Dehydrogenase/genetics , Malate Dehydrogenase/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neuroglia/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Transport , Signal Transduction , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...