Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(12): e23275, 2023 12.
Article in English | MEDLINE | ID: mdl-37902531

ABSTRACT

Ribosomal proteins play diverse roles in development and disease. Most ribosomal proteins have canonical roles in protein synthesis, while some exhibit extra-ribosomal functions. Previous studies in our laboratory revealed that ribosomal protein L13a (RPL13a) is involved in the translational silencing of a cohort of inflammatory proteins in myeloid cells. This prompted us to investigate the role of RPL13a in embryonic development. Here we report that RPL13a is required for early development in mice. Crosses between Rpl13a+/- mice resulted in no Rpl13a-/- offspring. Closer examination revealed that Rpl13a-/- embryos were arrested at the morula stage during preimplantation development. RNA sequencing analysis of Rpl13a-/- morulae revealed widespread alterations in gene expression, including but not limited to several genes encoding proteins involved in the inflammatory response, embryogenesis, oocyte maturation, stemness, and pluripotency. Ex vivo analysis revealed that RPL13a was localized to the cytoplasm and nucleus between the two-cell and morula stages. RNAi-mediated depletion of RPL13a phenocopied Rpl13a-/- embryos and knockdown embryos exhibited increased expression of IL-7 and IL-17 and decreased expression of the lineage specifier genes Sox2, Pou5f1, and Cdx2. Lastly, a protein-protein interaction assay revealed that RPL13a is associated with chromatin, suggesting an extra ribosomal function in transcription. In summary, our data demonstrate that RPL13a is essential for the completion of preimplantation embryo development. The mechanistic basis of the absence of RPL13a-mediated embryonic lethality will be addressed in the future through follow-up studies on ribosome biogenesis, global protein synthesis, and identification of RPL13a target genes using chromatin immunoprecipitation and RNA-immunoprecipitation-based sequencing.


Subject(s)
Embryonic Development , Ribosomal Proteins , Animals , Female , Humans , Mice , Pregnancy , Blastocyst , Chromatin Immunoprecipitation , Embryonic Development/genetics , Gene Expression , Ribosomal Proteins/genetics
2.
J Virol ; 96(2): e0167821, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34757848

ABSTRACT

The positive-sense, single-stranded RNA genome SARS-CoV-2 harbors functionally important cis-acting elements governing critical aspects of viral gene expression. However, insights on how these elements sense various signals from the host cell and regulate viral protein synthesis are lacking. Here, we identified two novel cis-regulatory elements in SARS-CoV-2 ORF1a and S RNAs and describe their role in translational control of SARS-CoV-2. These elements are sequence-unrelated but form conserved hairpin structures (validated by NMR) resembling gamma activated inhibitor of translation (GAIT) elements that are found in a cohort of human mRNAs directing translational suppression in myeloid cells in response to IFN-γ. Our studies show that treatment of human lung cells with receptor-binding S1 subunit, S protein pseudotyped lentivirus, and S protein-containing virus-like particles triggers a signaling pathway involving DAP-kinase1 that leads to phosphorylation and release of the ribosomal protein L13a from the large ribosomal subunit. Released L13a forms a virus activated inhibitor of translation (VAIT) complex that binds to ORF1a and S VAIT elements, causing translational silencing. Translational silencing requires extracellular S protein (and its interaction with host ACE2 receptor), but not its intracellular synthesis. RNA-protein interaction analyses and in vitro translation experiments showed that GAIT and VAIT elements do not compete with each other, highlighting differences between the two pathways. Sequence alignments of SARS-CoV-2 genomes showed a high level of conservation of VAIT elements, suggesting their functional importance. This VAIT-mediated translational control mechanism of SARS-CoV-2 may provide novel targets for small molecule intervention and/or facilitate development of more effective mRNA vaccines. IMPORTANCE Specific RNA elements in the genomes of RNA viruses play important roles in host-virus interaction. For SARS-CoV-2, the mechanistic insights on how these RNA elements could sense the signals from the host cell are lacking. Here we report a novel relationship between the GAIT-like SARS-CoV-2 RNA element (called VAITs) and the signal generated from the host cell. We show that for SARS-CoV-2, the interaction of spike protein with ACE2 not only serves the purpose for viral entry into the host cell, but also transduces signals that culminate into the phosphorylation and the release of L13a from the large ribosomal subunit. We also show that this event leads to the translational arrest of ORF1a and S mRNAs in a manner dependent on the structure of the RNA elements. Translational control of viral mRNA by a host-cell generated signal triggered by viral protein is a new paradigm in the host-virus relationship.


Subject(s)
COVID-19 , Host Microbial Interactions , RNA, Viral/immunology , SARS-CoV-2 , A549 Cells , COVID-19/immunology , COVID-19/virology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Protein Binding , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Virus Internalization
3.
FASEB J ; 35(11): e21990, 2021 11.
Article in English | MEDLINE | ID: mdl-34665898

ABSTRACT

Eukaryotic initiation factor 2A (eIF2A) is a 65 kDa protein that functions in minor initiation pathways, which affect the translation of only a subset of messenger ribonucleic acid (mRNAs), such as internal ribosome entry site (IRES)-containing mRNAs and/or mRNAs harboring upstream near cognate/non-AUG start codons. These non-canonical initiation events are important for regulation of protein synthesis during cellular development and/or the integrated stress response. Selective eIF2A knockdown in cellular systems significantly inhibits translation of such mRNAs, which rely on alternative initiation mechanisms for their translation. However, there exists a gap in our understanding of how eIF2A functions in mammalian systems in vivo (on the organismal level) and ex vivo (in cells). Here, using an eIF2A-knockout (KO) mouse model, we present evidence implicating eIF2A in the biology of aging, metabolic syndrome and central tolerance. We discovered that eIF2A-KO mice have reduced life span and that eIF2A plays an important role in maintenance of lipid homeostasis, the control of glucose tolerance, insulin resistance and also reduces the abundance of B lymphocytes and dendritic cells in the thymic medulla of mice. We also show the eIF2A KO affects male and female mice differently, suggesting that eIF2A may affect sex-specific pathways. Interestingly, our experiments involving pharmacological induction of endoplasmic reticulum (ER) stress with tunicamycin did not reveal any substantial difference between the response to ER stress in eIF2A-KO and wild-type mice. The identification of eIF2A function in the development of metabolic syndrome bears promise for the further identification of specific eIF2A targets responsible for these changes.


Subject(s)
Lipid Metabolism , Longevity , Metabolic Syndrome/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors
4.
FASEB J ; 34(5): 6888-6906, 2020 05.
Article in English | MEDLINE | ID: mdl-32232901

ABSTRACT

Previously, we identified a mechanism of inflammation control directed by ribosomal protein L13a and "GAIT" (Gamma Activated Inhibitor of Translation) elements in target mRNAs and showed that its elimination in myeloid cell-specific L13a knockout mice (L13a KO) increased atherosclerosis susceptibility and severity. Here, we investigated the mechanistic basis of this endogenous defense against atherosclerosis. We compared molecular and cellular aspects of atherosclerosis in high-fat diet (HFD)-fed L13a KO and intact (control) mice. HFD treatment of control mice induced release of L13a from 60S ribosome, formation of RNA-binding complex, and subsequent GAIT element-mediated translational silencing. Atherosclerotic plaques from HFD-treated KO mice showed increased infiltration of M1 type inflammatory macrophages. Macrophages from KO mice showed increased phagocytic activity and elevated expression of LDL receptor and pro-inflammatory mediators. NanoString analysis of the plaques from KO mice showed upregulation of a number of mRNAs encoding inflammatory proteins. Bioinformatics analysis suggests the presence of the potential GAIT elements in the 3'UTRs of several of these mRNAs. Macrophage induces L13a/GAIT-dependent translational silencing of inflammatory genes in response to HFD as an endogenous defense against atherosclerosis in ApoE-/- model.


Subject(s)
Atherosclerosis/prevention & control , Inflammation Mediators/metabolism , Macrophages/metabolism , Ribosomal Proteins/deficiency , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Differentiation , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Female , Macrophages/classification , Macrophages/pathology , Male , Mice , Mice, Knockout , Mice, Knockout, ApoE , Myeloid Cells/metabolism , Myeloid Cells/pathology , Phagocytosis , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
5.
RNA ; 25(10): 1377-1392, 2019 10.
Article in English | MEDLINE | ID: mdl-31308261

ABSTRACT

Eukaryotic ribosomal protein L13a is a member of the conserved universal ribosomal uL13 protein family. Structurally, L13a is distinguished from its prokaryotic counterparts by the presence of an ∼55 amino acid-long carboxy-terminal α-helical extension. The importance of these evolved residues in the carboxy-terminal extension for mammalian ribosome biogenesis as well as L13a's extraribosomal function in GAIT (γ interferon-activated inhibitor of translation) complex-mediated translation silencing during inflammation is not understood. Here, we present biochemical analyses of L13a mutant variants identifying several mutually exclusive amino acid residues in the eukaryote-specific carboxy-terminal extension of human L13a (Tyr149-Val203) important for ribosomal incorporation and translational silencing. Specifically, we show that mutation of Arg169, Lys170, and Lys171 to Ala abrogate GAIT-mediated translational silencing, but not L13a incorporation into ribosomes. Moreover, we show that the carboxy-terminal helix alone can silence translation of GAIT element-containing mRNAs in vitro. We also show through cellular immunofluorescence experiments that nuclear but not nucleolar localization of L13a is resistant to extensive amino acid alterations, suggesting that multiple complex nuclear import signals are present within this protein. These studies provide new insights into L13a structure and its ribosomal and extraribosomal functions in model human cells.


Subject(s)
Amino Acids/metabolism , Gene Silencing , Inflammation/prevention & control , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism , HEK293 Cells , Humans , Mutation , Nuclear Localization Signals , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics
7.
Nucleic Acids Res ; 45(22): 12987-13003, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29069516

ABSTRACT

RNA-protein interactions with physiological outcomes usually rely on conserved sequences within the RNA element. By contrast, activity of the diverse gamma-interferon-activated inhibitor of translation (GAIT)-elements relies on the conserved RNA folding motifs rather than the conserved sequence motifs. These elements drive the translational silencing of a group of chemokine (CC/CXC) and chemokine receptor (CCR) mRNAs, thereby helping to resolve physiological inflammation. Despite sequence dissimilarity, these RNA elements adopt common secondary structures (as revealed by 2D-1H NMR spectroscopy), providing a basis for their interaction with the RNA-binding GAIT complex. However, many of these elements (e.g. those derived from CCL22, CXCL13, CCR4 and ceruloplasmin (Cp) mRNAs) have substantially different affinities for GAIT complex binding. Toeprinting analysis shows that different positions within the overall conserved GAIT element structure contribute to differential affinities of the GAIT protein complex towards the elements. Thus, heterogeneity of GAIT elements may provide hierarchical fine-tuning of the resolution of inflammation.


Subject(s)
Chemokines/genetics , Gene Expression Regulation , RNA, Messenger/genetics , Silencer Elements, Transcriptional/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Chemokine CCL22/genetics , Chemokine CCL22/metabolism , Chemokines/metabolism , Conserved Sequence/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Mice, Knockout , Mice, Transgenic , Nucleic Acid Conformation , Operon , RNA, Messenger/chemistry , RNA, Messenger/metabolism , U937 Cells
8.
Cell Cycle ; 15(22): 3115-3120, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27686860

ABSTRACT

Eukaryotic initiation factor 2A (eIF2A) is a 65-kDa protein that was first identified in the early 1970s as a factor capable of stimulating initiator methionyl-tRNAi (Met-tRNAMeti) binding to 40S ribosomal subunits in vitro. However, in contrast to the eIF2, which stimulates Met-tRNAMeti binding to 40S ribosomal subunits in a GTP-dependent manner, eIF2A didn't reveal any GTP-dependence, but instead was found to direct binding of the Met-tRNAMeti to 40S ribosomal subunits in a codon-dependent manner. eIF2A appears to be highly conserved across eukaryotic species, suggesting conservation of function in evolution. The yeast Saccharomyces cerevisae eIF2A null mutant revealed no apparent phenotype, however, it was found that in yeast eIF2A functions as a suppressor of internal ribosome entry site (IRES)-mediated translation. It was thus suggested that eIF2A my act by impinging on the expression of specific mRNAs. Subsequent studies in mammalian cell systems implicated eIF2A in non-canonical (non-AUG-dependent) translation initiation events involving near cognate UUG and CUG codons. Yet, the role of eIF2A in cellular functions remains largely enigmatic. As a first step toward characterization of the eIF2A function in mammalian systems in vivo, we have obtained homozygous eIF2A-total knockout (KO) mice, in which a gene trap cassette was inserted between eIF2A exons 1 and 2 disrupting expression of all exons downstream of the insertion. The KO mice strain is viable and to date displays no apparent phenotype. We believe that the eIF2A KO mice strain will serve as a valuable tool for researchers studying non-canonical initiation of translation in vivo.


Subject(s)
Eukaryotic Initiation Factor-2/deficiency , Animals , Base Sequence , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Mice, Inbred C57BL , Mice, Knockout
9.
Cell Mol Immunol ; 13(6): 816-827, 2016 11.
Article in English | MEDLINE | ID: mdl-26166763

ABSTRACT

Sustained inflammation from infiltrated immune cells plays a pivotal role in the pathogenesis of ulcerative colitis (UC). Previously, we established the role of ribosomal protein L13a in the regulation of an inflammation-responsive post-transcriptional operon in myeloid cells. However, the role of this protein as a molecular cue to control the severity of colitis is not known. Here, we examined whether L13a-dependent translational control in macrophages could serve as an endogenous defense against colitis. The administration of dextran sodium sulfate induced experimental colitis in myeloid-specific L13a-knockout (KO) and control mice. Pathological scoring and injury to the colon mucosa evaluated the severity of colitis. The steady-state levels of several pro-inflammatory cytokines and chemokines were determined through ELISA and polyribosome profile analysis. Rapid weight loss, severe rectal bleeding, shortening of the colon, and significantly reduced survival rate were observed in the KO mice. Histopathological analysis of the colons of KO mice showed a severe disruption of epithelial crypts with immune cell infiltrates. Elevated levels of several inflammatory cytokines and chemokines and abrogation of their naturally imposed translational silencing were observed in the colons of the KO mice. Higher serum levels of several pro-inflammatory cytokines and the release of gut bacteria and endotoxins into the blood streams of KO mice were detected, suggesting the amplification of the inflammatory response to septicemia. Taken together, these results reveal an essential role for L13a in the endogenous protection against UC and demonstrate the potential for new therapeutic opportunities through the deliberate promotion of this mechanism.


Subject(s)
Colitis/metabolism , Colitis/pathology , Macrophages/metabolism , Protein Biosynthesis , Ribosomal Proteins/metabolism , Animals , Chemokines/metabolism , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Dextran Sulfate , Epithelial Cells/metabolism , Gene Deletion , Gene Silencing , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sepsis/immunology , Sepsis/pathology
10.
J Virol ; 88(16): 9100-10, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24899178

ABSTRACT

UNLABELLED: We report a novel extraribosomal innate immune function of mammalian ribosomal protein L13a, whereby it acts as an antiviral agent. We found that L13a is released from the 60S ribosomal subunit in response to infection by respiratory syncytial virus (RSV), an RNA virus of the Pneumovirus genus and a serious lung pathogen. Unexpectedly, the growth of RSV was highly enhanced in L13a-knocked-down cells of various lineages as well as in L13a knockout macrophages from mice. In all L13a-deficient cells tested, translation of RSV matrix (M) protein was specifically stimulated, as judged by a greater abundance of M protein and greater association of the M mRNA with polyribosomes, while general translation was unaffected. In silico RNA folding analysis and translational reporter assays revealed a putative hairpin in the 3'untranslated region (UTR) of M mRNA with significant structural similarity to the cellular GAIT (gamma-activated inhibitor of translation) RNA hairpin, previously shown to be responsible for assembling a large, L13a-containing ribonucleoprotein complex that promoted translational silencing in gamma interferon (IFN-γ)-activated myeloid cells. However, RNA-protein interaction studies revealed that this complex, which we named VAIT (respiratory syncytial virus-activated inhibitor of translation) is functionally different from the GAIT complex. VAIT is the first report of an extraribosomal L13a-mediated, IFN-γ-independent innate antiviral complex triggered in response to virus infection. We provide a model in which the VAIT complex strongly hinders RSV replication by inhibiting the translation of the rate-limiting viral M protein, which is a new paradigm in antiviral defense. IMPORTANCE: The innate immune mechanisms of host cells are diverse in nature and act as a broad-spectrum cellular defense against viruses. Here, we report a novel innate immune mechanism functioning against respiratory syncytial virus (RSV), in which the cellular ribosomal protein L13a is released from the large ribosomal subunit soon after infection and inhibits the translation of a specific viral mRNA, namely, that of the matrix protein M. Regarding its mechanism, we show that the recognition of a specific secondary structure in the 3' untranslated region of the M mRNA leads to translational arrest of the mRNA. We also show that the level of M protein in the infected cell is rate limiting for viral morphogenesis, providing a rationale for L13a to target the M mRNA for suppression of RSV growth. Translational silencing of a viral mRNA by a deployed ribosomal protein is a new paradigm in innate immunity.


Subject(s)
Antiviral Agents/immunology , Immunity, Innate/immunology , Immunologic Factors/immunology , Ribosomal Proteins/immunology , 3' Untranslated Regions/genetics , 3' Untranslated Regions/immunology , Animals , Base Sequence , Cell Line , Cell Line, Tumor , Humans , Immunity, Innate/genetics , Immunologic Factors/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polyribosomes/genetics , Polyribosomes/immunology , Protein Biosynthesis/genetics , Protein Biosynthesis/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Respiratory Syncytial Viruses/immunology , Ribosomal Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosome Subunits, Large, Eukaryotic/immunology , U937 Cells
11.
Arterioscler Thromb Vasc Biol ; 34(3): 533-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24436370

ABSTRACT

OBJECTIVE: Unresolved inflammatory response of macrophages plays a pivotal role in the pathogenesis of atherosclerosis. Previously we showed that ribosomal protein L13a-dependent translational silencing suppresses the synthesis of a cohort of inflammatory proteins in monocytes and macrophages. We also found that genetic abrogation of L13a expression in macrophages significantly compromised the resolution of inflammation in a mouse model of lipopolysaccharide-induced endotoxemia. However, its function in the pathogenesis of atherosclerosis is not known. Here, we examine whether L13a in macrophage has a protective role against high-fat diet-induced atherosclerosis. APPROACH AND RESULTS: We bred the macrophage-specific L13a knockout mice L13a Flox(+/+) Cre(+/+) onto apolipoprotein E-deficient background and generated the experimental double knockout mice L13a Flox(+/+) Cre(+/+) apolipoprotein E deficient (apoE(-/-)). L13a Flox(+/+) Cre(-/-) mice on apolipoprotein E-deficient background were used as controls. Control and knockout mice were subjected to high-fat diet for 10 weeks. Evaluation of aortic sinus sections and entire aorta by en face showed significantly higher atherosclerosis in the knockout mice. Severity of atherosclerosis in knockout mice was accompanied by thinning of the smooth muscle cell layer in the media, larger macrophage area in the intimal plaque region and higher plasma levels of inflammatory cytokines. In addition, macrophages isolated from knockout mice had higher polyribosomal abundance of several target mRNAs, thus showing defect in translation control. CONCLUSIONS: Our data demonstrate that loss of L13a in macrophages increases susceptibility to atherosclerosis in apolipoprotein E-deficient mice, revealing an important role of L13a-dependent translational control as an endogenous protection mechanism against atherosclerosis.


Subject(s)
Aortic Diseases/etiology , Atherosclerosis/etiology , Inflammation/genetics , Macrophages/metabolism , Protein Biosynthesis , Ribosomal Proteins/physiology , Animals , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Chemotaxis, Leukocyte , Cholesterol/blood , Cytokines/blood , Dietary Fats/toxicity , Disease Progression , Genetic Predisposition to Disease , Inflammation/complications , Mice , Mice, Knockout , Peritoneum/pathology , Polyribosomes/metabolism , RNA, Messenger/analysis , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Tunica Media/pathology
12.
Mol Cell Biol ; 33(15): 2829-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689135

ABSTRACT

In contrast to prokaryotes, the precise mechanism of incorporation of ribosomal proteins into ribosomes in eukaryotes is not well understood. For the majority of eukaryotic ribosomal proteins, residues critical for rRNA binding, a key step in the hierarchical assembly of ribosomes, have not been well defined. In this study, we used the mammalian ribosomal protein L13a as a model to investigate the mechanism(s) underlying eukaryotic ribosomal protein incorporation into ribosomes. This work identified the arginine residue at position 68 of L13a as being essential for L13a binding to rRNA and incorporation into ribosomes. We also demonstrated that incorporation of L13a takes place during maturation of the 90S preribosome in the nucleolus, but that translocation of L13a into the nucleolus is not sufficient for its incorporation into ribosomes. Incorporation of L13a into the 90S preribosome was required for rRNA methylation within the 90S complex. However, mutations abolishing ribosomal incorporation of L13a did not affect its ability to be phosphorylated or its extraribosomal function in GAIT element-mediated translational silencing. These results provide new insights into the mechanism of ribosomal incorporation of L13a and will be useful in guiding future studies aimed at fully deciphering mammalian ribosome biogenesis.


Subject(s)
Neoplasm Proteins/metabolism , RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Amino Acid Sequence , Animals , HEK293 Cells , Humans , Interferon-gamma/metabolism , Methylation , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Phosphorylation , Point Mutation , Protein Binding , Ribosomal Proteins/analysis , Ribosomal Proteins/genetics
13.
J Immunol ; 190(7): 3600-12, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460747

ABSTRACT

Inflammation is an obligatory attempt of the immune system to protect the host from infections. However, unregulated synthesis of proinflammatory products can have detrimental effects. Although mechanisms that lead to inflammation are well appreciated, those that restrain it are not adequately understood. Creating macrophage-specific L13a-knockout mice, we report that depletion of ribosomal protein L13a abrogates the endogenous translation control of several chemokines in macrophages. Upon LPS-induced endotoxemia, these animals displayed symptoms of severe inflammation caused by widespread infiltration of macrophages in major organs causing tissue injury and reduced survival rates. Macrophages from these knockout animals show unregulated expression of several chemokines (e.g., CXCL13, CCL22, CCL8, and CCR3). These macrophages failed to show L13a-dependent RNA binding complex formation on target mRNAs. In addition, increased polyribosomal abundance of these mRNAs shows a defect in translation control in the macrophages. Thus, to our knowledge, our studies provide the first evidence of an essential extraribosomal function of ribosomal protein L13a in resolving physiological inflammation in a mammalian host.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Ribosomal Proteins/metabolism , Animals , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/pathology , Endotoxins/metabolism , Female , Gene Deletion , Genetic Predisposition to Disease , Homozygote , Inflammation/genetics , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/immunology , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Organ Specificity/genetics , Peritoneum/immunology , Peritoneum/pathology , Protein Biosynthesis/genetics , Response Elements , Ribosomal Proteins/genetics , Spleen/immunology , Spleen/pathology
14.
Gene ; 502(2): 75-86, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22555019

ABSTRACT

Studies over the past 5 or so years have indicated that the traditional clustering of mechanisms for translation initiation in eukaryotes into cap-dependent and cap-independent (or IRES-mediated) is far too narrow. From individual studies of a number of mRNAs encoding proteins that are regulatory in nature (i.e. likely to be needed in small amounts such as transcription factors, protein kinases, etc.), it is now evident that mRNAs exist that blur these boundaries. This review seeks to set the basic ground rules for the analysis of different initiation pathways that are associated with these new mRNAs as well as related to the more traditional mechanisms, especially the cap-dependent translational process that is the major route of initiation of mRNAs for housekeeping proteins and thus, the bulk of protein synthesis in most cells. It will become apparent that a mixture of descriptions is likely to become the norm in the near future (i.e. m(7)G-assisted internal initiation).


Subject(s)
Protein Biosynthesis/genetics , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Humans
15.
Mol Cell Biol ; 31(22): 4482-99, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21930789

ABSTRACT

Protein syntheses mediated by cellular and viral internal ribosome entry sites (IRESs) are believed to have many features in common. Distinct mechanisms for ribosome recruitment and preinitiation complex assembly between the two processes have not been identified thus far. Here we show that the methylation status of rRNA differentially influenced the mechanism of 80S complex formation on IRES elements from the cellular sodium-coupled neutral amino acid transporter 2 (SNAT2) versus the hepatitis C virus mRNA. Translation initiation involves the assembly of the 48S preinitiation complex, followed by joining of the 60S ribosomal subunit and formation of the 80S complex. Abrogation of rRNA methylation did not affect the 48S complex but resulted in impairment of 80S complex assembly on the cellular, but not the viral, IRESs tested. Impairment of 80S complex assembly on the amino acid transporter SNAT2 IRES was rescued by purified 60S subunits containing fully methylated rRNA. We found that rRNA methylation did not affect the activity of any of the viral IRESs tested but affected the activity of numerous cellular IRESs. This work reveals a novel mechanism operating on a cohort of cellular IRESs that involves rRNA methylation for proper 80S complex assembly and efficient translation initiation.


Subject(s)
Amino Acid Transport System A/metabolism , Peptide Chain Initiation, Translational , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , RNA, Ribosomal/metabolism , Ribosome Subunits/metabolism , Amino Acid Transport System A/genetics , Cationic Amino Acid Transporter 1/biosynthesis , HEK293 Cells , HeLa Cells , Hepacivirus/genetics , Humans , Methylation , Protein Biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Messenger/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Ribosomal Proteins/biosynthesis , Ribosome Subunits/chemistry , Ribosome Subunits, Large/metabolism , Ribosome Subunits, Small/metabolism , Stress, Physiological
16.
J Immunol ; 184(7): 3311-9, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20304832

ABSTRACT

A robust innate immune response is essential to the protection of all vertebrates from infection, but it often comes with the price tag of acute inflammation. If unchecked, a runaway inflammatory response can cause significant tissue damage, resulting in myriad disorders, such as dermatitis, toxic shock, cardiovascular disease, acute pelvic and arthritic inflammatory diseases, and various infections. To prevent such pathologies, cells have evolved mechanisms to rapidly and specifically shut off these beneficial inflammatory activities before they become detrimental. Our review of recent literature, including our own work, reveals that the most dominant and common mechanism is translational silencing, in which specific regulatory proteins or complexes are recruited to cis-acting RNA structures in the untranslated regions of single or multiple mRNAs that code for the inflammatory protein(s). Enhancement of the silencing function may constitute a novel pharmacological approach to prevent immunity-related inflammation.


Subject(s)
Gene Expression Regulation/immunology , Inflammation/genetics , Inflammation/immunology , Protein Biosynthesis/immunology , Animals , Gene Expression , Humans
17.
J Biol Chem ; 284(16): 10367-75, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19224918

ABSTRACT

IRAK2, a member of the interleukin-1 receptor-associated kinase (IRAK) family, has been implicated in Toll-like receptor (TLR)-mediated signaling. We generated IRAK2-deficient mice to examine its function in detail. These mice are resistant to lipopolysaccharide-induced septic shock, because of impaired TLR4-mediated induction of pro-inflammatory cytokines and chemokines. Although IRAK2 deficiency did not affect TLR4-mediated NFkappaB activation, a reduction of lipopolysaccharide (LPS)-mediated mRNA stabilization contributed to the reduced cytokine and chemokine production observed in bone marrow-derived macrophages from IRAK2-deficient mice. Furthermore, the ratios of LPS-induced cytokine and chemokine mRNAs in translation-active (polysomal) versus translation-inactive (free ribosomes) pools were reduced in IRAK2-deficient macrophages compared with wild type macrophages. Importantly, LPS-induced phosphorylation of MKK3/6, MNK1, and eIF4E was significantly reduced in IRAK2-deficient macrophages compared with wild type macrophages. Moreover, LPS stimulation induced an interaction of IRAK2 with TRAF6, MKK3/6, and MK2, implicating a critical role for mitogen-activated protein kinase signaling in LPS-induced IRAK2-mediated post-transcriptional control. These results reveal that IRAK2 is required for LPS-mediated post-transcriptional control of cytokine and chemokine expression, which plays an essential role in TLR4-induced septic shock.


Subject(s)
Gene Expression Regulation/drug effects , Interleukin-1 Receptor-Associated Kinases/metabolism , Lipopolysaccharides/pharmacology , RNA Processing, Post-Transcriptional , Transcription, Genetic , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Chemokines/genetics , Chemokines/immunology , Cytokines/genetics , Cytokines/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Lipopolysaccharides/immunology , MAP Kinase Signaling System/physiology , Macrophages/cytology , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Biosynthesis , RNA Stability , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
18.
Mol Cell Biol ; 29(2): 458-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001086

ABSTRACT

We previously showed that ribosomal protein L13a is required for translational silencing of gamma interferon (IFN-gamma)-induced ceruloplasmin (Cp) synthesis in monocytes. This silencing also requires the presence of the GAIT (IFN-gamma activated inhibitor of translation) element in the 3' untranslated region (UTR) of Cp mRNA. Considering that Cp is an inflammatory protein, we hypothesized that this mechanism may have evolved to silence a family of proinflammatory proteins, of which Cp is just one member. To identify the other mRNAs that are targets for this silencing, we performed a genome-wide analysis of the polysome-profiled mRNAs by using an Affymetrix GeneChip and an inflammation-responsive gene array. A cluster of mRNAs encoding different chemokines and their receptors was identified as common hits in the two approaches and validated by real-time PCR. In silico predicted GAIT hairpins in the 3' UTRs of the target mRNAs were confirmed as functional cis-acting elements for translational silencing by luciferase reporter assays. Consistent with Cp, the newly identified target mRNAs also required L13a for silencing. Our studies have identified a new inflammation-responsive posttranscriptional operon that can be regulated directly at the level of translation in IFN-gamma-activated monocytes. This regulation of a cohort of mRNAs encoding inflammatory proteins may be important to resolve inflammation.


Subject(s)
Inflammation/genetics , Interferon-gamma/metabolism , Monocytes/immunology , Monocytes/metabolism , Operon , Polyribosomes/genetics , 3' Untranslated Regions , Ceruloplasmin/metabolism , Chemokines/genetics , Chemokines/metabolism , Computer Simulation , Electrophoretic Mobility Shift Assay , Gene Silencing , Humans , Inflammation/metabolism , Oligonucleotide Array Sequence Analysis , Polyribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Ribosomal Proteins/metabolism , U937 Cells
19.
RNA ; 13(12): 2224-37, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17921318

ABSTRACT

Previously, we demonstrated that treatment of monocytic cells with IFN-gamma causes release of ribosomal protein L13a from the 60S ribosome and subsequent translational silencing of Ceruloplasmin (Cp) mRNA. Here, evidence using cultured cells demonstrates that Cp mRNA silencing is dependent on L13a and that L13a-deficient ribosomes are competent for global translational activity. Human monocytic U937 cells were stably transfected with two different shRNA sequences for L13a and clonally selected for more than 98% abrogation of total L13a expression. Metabolic labeling of these cells showed rescue of Cp translation from the IFN-gamma mediated translational silencing activity. Depletion of L13a caused significant reduction of methylation of ribosomal RNA and of cap-independent translation mediated by Internal Ribosome Entry Site (IRES) elements derived from p27, p53, and SNAT2 mRNAs. However, no significant differences in the ribosomal RNA processing, polysome formation, global translational activity, translational fidelity, and cell proliferation were observed between L13a-deficient and wild-type control cells. These results support the notion that ribosome can serve as a depot for releasable translation-regulatory factors unrelated to its basal polypeptide synthetic function. Unlike mammalian cells, the L13a homolog in yeast is indispensable for growth. Thus, L13a may have evolved from an essential ribosomal protein in lower eukaryotes to having a role as a dispensable extra-ribosomal function in higher eukaryotes.


Subject(s)
RNA, Ribosomal/metabolism , Ribosomal Proteins/metabolism , Ribosomes/physiology , tRNA Methyltransferases/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Lentivirus/genetics , RNA Interference , RNA, Neoplasm/genetics , Recombinant Proteins/metabolism , Ribosomal Proteins/genetics , Transfection , U937 Cells
20.
RNA ; 13(12): 2116-28, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17901157

ABSTRACT

Ribosomal protein (rp) S5 belongs to a family of ribosomal proteins that includes bacterial rpS7. rpS5 forms part of the exit (E) site on the 40S ribosomal subunit and is essential for yeast viability. Human rpS5 is 67% identical and 79% similar to Saccharomyces cerevisiae rpS5 but lacks a negatively charged (pI approximately 3.27) 21 amino acid long N-terminal extension that is present in fungi. Here we report that replacement of yeast rpS5 with its human homolog yielded a viable yeast strain with a 20%-25% decrease in growth rate. This replacement also resulted in a moderate increase in the heavy polyribosomal components in the mutant strain, suggesting either translation elongation or termination defects, and in a reduction in the polyribosomal association of the elongation factors eEF3 and eEF1A. In addition, the mutant strain was characterized by moderate increases in +1 and -1 programmed frameshifting and hyperaccurate recognition of the UAA stop codon. The activities of the cricket paralysis virus (CrPV) IRES and two mammalian cellular IRESs (CAT-1 and SNAT-2) were also increased in the mutant strain. Consistently, the rpS5 replacement led to enhanced direct interaction between the CrPV IRES and the mutant yeast ribosomes. Taken together, these data indicate that rpS5 plays an important role in maintaining the accuracy of translation in eukaryotes and suggest that the negatively charged N-terminal extension of yeast rpS5 might affect the ribosomal recruitment of specific mRNAs.


Subject(s)
Ribosomal Proteins/metabolism , Saccharomyces cerevisiae/genetics , Base Sequence , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nucleic Acid Conformation , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...