Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
BMC Cancer ; 24(1): 432, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589831

ABSTRACT

BACKGROUND: One-third of diffuse large B-cell lymphoma (DLBCL) patients suffer relapse after standard treatment. Eukaryotic initiation factor 3a (eIF3a) is a key player in the initial stage of translation, which has been widely reported to be correlated with tumorigenesis and therapeutic response. This study aimed to explore the biological role of eIF3a, evaluate its prognostic and therapeutic potential in DLBCL. METHODS: RNA-seq datasets from GEO database were utilized to detect the expression and prognostic role of eIF3a in DLBCL patients. Protein level of eIF3a was estimated by western blot and immunohistochemical. Next, DLBCL cells were transfected with lentiviral vector either eIF3a-knockdown or empty to assess the biological role of eIF3a. Then, samples were divided into 2 clusters based on eIF3a expression and differentially expressed genes (DEGs) were identified. Function enrichment and mutation analysis of DEGs were employed to detect potential biological roles. Moreover, we also applied pan-cancer and chemosensitivity analysis for deep exploration. RESULTS: eIF3a expression was found to be higher in DLBCL than healthy controls, which was associated with worse prognosis. The expression of eIF3a protein was significantly increased in DLBCL cell lines compared with peripheral blood mononuclear cells (PBMCs) from healthy donors. eIF3a knockdown inhibited the proliferation of DLBCL cells and the expression of proliferation-related proteins and increase cell apoptosis rate. Besides, 114 DEGs were identified which had a close linkage to cell cycle and tumor immune. eIF3a and DEGs mutations were found to be correlated to chemosensitivity and vital signal pathways. Pan-cancer analysis demonstrated that high eIF3a expression was associated with worse prognosis in several tumors. Moreover, eIF3a expression was found to be related to chemosensitivity of several anti-tumor drugs in DLBCL, including Vincristine and Wee1 inhibitor. CONCLUSIONS: We firstly revealed the high expression and prognostic role of eIF3a in DLBCL, and eIF3a might promote the development of DLBCL through regulating cell proliferation and apoptosis. eIF3a expression was related to immune profile and chemosensitivity in DLBCL. These results suggest that eIF3a could serve as a potential prognostic biomarker and therapeutic target in DLBCL.


Subject(s)
Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Humans , Leukocytes, Mononuclear , Cell Proliferation/genetics , Antineoplastic Agents/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/diagnosis , Peptide Initiation Factors/pharmacology , Peptide Initiation Factors/therapeutic use , Cell Line, Tumor
2.
J Ethnopharmacol ; 325: 117859, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38316218

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Endometriosis (EMs) is characterized by inflammatory lesions, dysmenorrhea, infertility, and chronic pelvic pain. Single-target medications often fail to provide systemic therapeutic results owing to the complex mechanism underlying endometriosis. Although traditional Chinese medicines-such as Juan-Tong-Yin (JTY)-have shown promising results, their mechanisms of action remain largely unknown. AIM OF THE STUDY: To elucidate the therapeutic mechanism of JTY in EMs, focusing on endoplasmic reticulum (ER) stress-induced autophagy. MATERIALS AND METHODS: The major components of JTY were detected using high-performance liquid chromatography-mass spectrometry (HPLC-MS). The potential mechanism of JTY in EMs treatment was predicted using network pharmacological analysis. Finally, the pathogenesis of EMs was validated in a clinical case-control study and the molecular mechanism of JTY was validated in vitro using endometrial stromal cells (ESCs). RESULTS: In total, 241 compounds were analyzed and identified from JTY using UPLC-MS. Network pharmacology revealed 288 targets between the JTY components and EMs. Results of the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses indicated that regulating autophagy, migration, apoptosis, and inflammation were the key mechanisms of JTY in treating EMs. Meanwhile, we found that protein kinase R-like endoplasmic reticulum kinase (PERK), Beclin-1, and microtubule-associated protein light chain 3 B (LC3B) expressions were lower in endometria of patients with EMs than in those with normal eutopic endometria (p < 0.05). Additionally, during in vitro experiments, treatment with 20% JTY-containing serum significantly suppressed ESC proliferation, achieving optimal effects after 48 h. Electron microscopy revealed significantly increased autophagy flux in the JTY group compared with the control group. Moreover, JTY treatment significantly reduced the migratory and invasive abilities of ESCs and upregulated protein expression of PERK, eukaryotic initiation factor 2α (eIF2α)/phospho-eukaryotic initiation factor 2α (p-eIF2α), activating Transcription Factor-4 (ATF4), Beclin-1, and LC3BII/I, while subsequently downregulating NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and interleukin 18 (IL-18) expression. However, administration of GSK2656157-a highly selective PERK inhibitor-reversed these changes. CONCLUSION: JTY ameliorates EMs by activating PERK associated with unfolded protein reaction, enhancing cell ER stress and autophagy, improving the inflammatory microenvironment, and decreasing the migration and invasion of ESCs.


Subject(s)
Endometriosis , Signal Transduction , Female , Humans , Beclin-1/metabolism , Endometriosis/pathology , Case-Control Studies , Chromatography, Liquid , Tandem Mass Spectrometry , Endoplasmic Reticulum Stress , Autophagy , Apoptosis , Stromal Cells/metabolism , Stromal Cells/pathology , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/pharmacology
3.
Gene ; 884: 147702, 2023 Oct 30.
Article in English | MEDLINE | ID: mdl-37567453

ABSTRACT

Acute lung injury (ALI) is a common inflammatory respiratory disorder characterized by a high incidence and mortality rate. This study aimed to investigate the potential therapeutic effects of the neutrophil elastase inhibitor Sivelestat sodium (SIV) in improving endoplasmic reticulum stress (ERS) while treating lipopolysaccharide (LPS)-induced ALI. An ALI model was established using LPS induction. The effects of SIV on ALI were observed both in vivo and in vitro, along with its impact on ERS. Lung tissue damage was assessed using Hematoxylin-eosin (H&E) staining. Lung edema was measured by the lung wet/dry weight ratio. The expression levels of protein kinase R-like ER kinase (PERK), Phospho-protein kinase R-like ER kinase (p-PERK), activating transcription factor 4 (ATF4), eukaryotic translation initiation factor 2α (EIF2a), phosphorylated α subunit of eukaryotic initiation factor 2α (P-EIF2a), and C/EBP homologous protein (CHOP) were analyzed by Western blotting in vivo and in vitro. The levels of tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) in Lung tissue samples supernatants were measured by ELISA. Oxidative stress markers were measured by ELISA. Apoptosis was measured using the TUNEL assay. Apoptosis-associated proteins B-cell lymphoma-2 (Bcl-2)、Bcl2-associated × (Bax)、caspase-3 were evaluated through Western blotting in vivo and in vitro. The expression levels of ERS-related proteins, including p-PERK, ATF4, P-EIF2a, and CHOP, were significantly increased in the LPS-induced ALI model. However, SIV markedly reduced the expression levels of these proteins, suppressing the LPS-induced ERS response. Further investigations revealed that SIV exerted a protective effect on ALI by alleviating lung tissue damage and apoptosis, improving lung function, and reducing inflammation and oxidative stress levels. However, when SIV was co-administered with Tunicamycin (TUN), TUN blocked the beneficial effects of SIV on ERS and reversed the protective effects of SIV on ALI. In conclusion, SIV alleviated lung tissue damage and apoptosis, improving lung function, and reducing inflammation and oxidative stress in LPS-induced ALI by improving ERS.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Humans , Lipopolysaccharides/pharmacology , Endoplasmic Reticulum Stress , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Apoptosis , Inflammation , Protein Kinases , Peptide Initiation Factors/pharmacology , Sodium
4.
Acta Cir Bras ; 38: e380323, 2023.
Article in English | MEDLINE | ID: mdl-36888755

ABSTRACT

PURPOSE: Sepsis is characterized by an acute inflammatory response to infection, often with multiple organ failures, especially severe lung injury. This study was implemented to probe circular RNA (circRNA) protein tyrosine kinase 2 (circPTK2)-associated regulatory mechanisms in septic acute lung injury (ALI). METHODS: A cecal ligation and puncture-based mouse model and an lipopolysaccharides (LPS)-based alveolar type II cell (RLE-6TN) model were generated to mimic sepsis. In the two models, inflammation- and pyroptosis-related genes were measured. RESULTS: The degree of lung injury in mice was analyzed by hematoxylin and eosin (H&E) staining and the apoptosis was by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining. In addition, pyroptosis and toxicity were detected in cells. Finally, the binding relationship between circPTK2, miR-766, and eukaryotic initiation factor 5A (eIF5A) was detected. Data indicated that circPTK2 and eIF5A were up-regulated and miR-766 was down-regulated in LPS-treated RLE-6TN cells and lung tissue of septic mice. Lung injury in septic mice was ameliorated after inhibition of circPTK2. CONCLUSIONS: It was confirmed in the cell model that knockdown of circPTK2 effectively ameliorated LPS-induced ATP efflux, pyroptosis, and inflammation. Mechanistically, circPTK2 mediated eIF5A expression by competitively adsorbing miR-766. Taken together, circPTK2/miR-766/eIF5A axis ameliorates septic ALI, developing a novel therapeutic target for the disease.


Subject(s)
Acute Lung Injury , MicroRNAs , Sepsis , Animals , Mice , Pyroptosis , RNA, Circular/genetics , RNA, Circular/pharmacology , Focal Adhesion Kinase 1/pharmacology , Lipopolysaccharides/adverse effects , Lung/metabolism , Apoptosis , Acute Lung Injury/metabolism , Sepsis/genetics , Peptide Initiation Factors/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Adenosine Triphosphate/pharmacology
5.
Fundam Clin Pharmacol ; 37(2): 245-252, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36355605

ABSTRACT

Anlotinib is approved for refractory cases in advanced non-small-cell lung cancer (NSCLC). This is a novel oral multitarget tyrosine kinase inhibitor, but patients inevitably face prospects of drug resistance during the treatment process. Using anlotinib-resistant NSCLC models, this work investigated the underlying molecular mechanism and systematically addressed the issue of anlotinib resistance. We demonstrated that expression and activity of eukaryotic translation initiation factor 4E (eIF4E) were upregulated in NSCLC cells due to prolonged exposure to anlotinib. eIF4E depletion resulted in significant effects to anlotinib-resistant cells, showing proliferation inhibition and apoptosis inducement. We further showed that MAP kinase interacting serine/threonine kinase (MNK)-dependent eIF4E inhibition by cercosporamide was active against anlotinib-resistant cells and significantly augmented anlotinib's efficacy in parental NSCLC cells. Importantly, observations from in-vitro experiments are consistent in in vivo anlotinib-resistant and anlotinib-sensitive NSCLC cancer xenograft mouse models. Our work is the first to reveal that eIF4E is involved intimately in anlotinib resistance development in NSCLC, and this eIF4E activation can be reversed by cercosporamide or other MNK inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Intracellular Signaling Peptides and Proteins , Phosphorylation , Cell Line, Tumor , Lung Neoplasms/drug therapy , Peptide Initiation Factors/pharmacology , Peptide Initiation Factors/therapeutic use , Cell Proliferation
6.
Mol Med ; 28(1): 103, 2022 09 04.
Article in English | MEDLINE | ID: mdl-36058905

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is still a critical problem in clinical practice, with a heavy burden for national health system around the world. It is notable that sepsis is the predominant cause of AKI for patients in the intensive care unit and the mortality remains considerably high. The treatment for AKI relies on supportive therapies and almost no specific treatment is currently available. Spermidine is a naturally occurring polyamine with pleiotropic effects. However, the renoprotective effect of spermidine and the underlying mechanism remain elusive. METHODS: We employed mice sepsis-induced AKI model and explored the potential renoprotective effect of spermidine in vivo with different administration time and routes. Macrophage depleting was utilized to probe the role of macrophage. In vitro experiments were conducted to examine the effect of spermidine on macrophage cytokine secretion, NLRP3 inflammasome activation and mitochondrial respiration. RESULTS: We confirmed that spermidine improves AKI with different administration time and routes and that macrophages serves as an essential mediator in this protective effect. Meanwhile, spermidine downregulates NOD-like receptor protein 3 (NLRP3) inflammasome activation and IL-1 beta production in macrophages directly. Mechanically, spermidine enhances mitochondrial respiration capacity and maintains mitochondria function which contribute to the NLRP3 inhibition. Importantly, we showed that eukaryotic initiation factor 5A (eIF5A) hypusination plays an important role in regulating macrophage bioactivity. CONCLUSIONS: Spermidine administration practically protects against sepsis-induced AKI in mice and macrophages serve as an essential mediator in this protective effect. Our study identifies spermidine as a promising pharmacologic approach to prevent AKI.


Subject(s)
Acute Kidney Injury , Sepsis , Acute Kidney Injury/metabolism , Animals , Disease Models, Animal , Inflammasomes/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/pharmacology , Peptide Initiation Factors/therapeutic use , Respiration , Sepsis/metabolism , Spermidine/metabolism , Spermidine/pharmacology , Spermidine/therapeutic use
7.
Biochimie ; 84(8): 815-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12457568

ABSTRACT

Polyamines are involved in various cellular processes including embryonic development, cell growth and cell cycle regulation, apoptosis and carcinogenesis. Growing evidence suggests the importance of RNA processing and stability mediated by oncogenes and tumor suppressor genes in the development of cancer. Polyamines, which are found to be increased in neoplastic cells and tissues compared to normal, bind to RNA to influence its structure and function. Polyamines mediate RNA processing through the spermidine-regulated protein, eIF-5A. To further investigate how polyamines influence RNA expression, we characterized the polyamine-dependent RNA expression of a cancer-related gene, cyclooxygenase-2 (COX-2), which contains eIF-5A consensus binding elements. Depletion of polyamines by DL-alpha-difluoromethylornithine (DFMO) treatment caused an induction of COX-2 mRNA steady-state levels. Polyamines appear to regulate expression of COX-2 by a post-transcriptional mechanism.


Subject(s)
Biogenic Polyamines/pharmacology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Processing, Post-Translational , RNA-Binding Proteins , 3' Untranslated Regions/genetics , Base Sequence , Biogenic Polyamines/biosynthesis , Blotting, Northern , Caco-2 Cells , Cyclooxygenase 2 , Eflornithine/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Membrane Proteins , Models, Biological , Peptide Initiation Factors/pharmacology , Promoter Regions, Genetic/physiology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/biosynthesis , Eukaryotic Translation Initiation Factor 5A
8.
Br J Cancer ; 86(7): 1023-7, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953842

ABSTRACT

Regulation of protein synthesis at the level of translation initiation is fundamentally important for the control of cell proliferation under normal physiological conditions. Conversely, misregulation of protein synthesis is emerging as a major contributory factor in cancer development. Most bulk protein synthesis is initiated via recognition of the mRNA 5' cap and subsequent recognition of the initiator AUG codon by a directional scanning mechanism. However, several key regulators of tumour development are translated by a cap-independent pathway. Here we review eukaryotic translation initiation, its regulation and the ways in which this regulation can break down during tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Peptide Initiation Factors/pharmacology , Protein Biosynthesis , Animals , Eukaryotic Cells/physiology , Humans , Neoplasms/physiopathology , RNA, Messenger
9.
Wei Sheng Yan Jiu ; 31(4): 255-7, 2002 Aug.
Article in Chinese | MEDLINE | ID: mdl-12600033

ABSTRACT

TIF3 (GenBank Accession Number AF 271072) is identified as a novel cadmium- responsive proto-oncogene. In order to determine whether the antisense TIF3 reverses the oncogenic potential of Cd-transformed BALB/c-3T3 cells or not, a stable expression system of CdCl2-transformed BALB/c-3T3 cells with the expression vector containing TIF3 cDNA in the antisense orientation using calcium phosphate and G418 selection protocols is firstly established. Then, the reversal of the oncogenic potential of these cells is tested by soft agar and nude mouse tumorigenicity assay. The results demonstrated that expression of the antisense TIF3 in the CdCl2-transformed BALB/c-3T3 cells results in reversal of the transformed phenotype of the cells. This is evidenced by a 25%-70% decrease in the number of anchorage-independent colonies growing on soft agar and the significant reduced tumorigenic potential of cells in nude mice compared with the corresponding controls. In addition to a significant delay in the onset of appearance of tumors, a significant reduction in size and a 50.8%-55.1% decrease in weight of the tumors are also observed in the mice injected with the TIF3 antisense expressing cells compared with the corresponding controls. The results indicate that antisense TIF3 mRNA expression reverses its oncogenic potential of Cd-transformed BALB/c-3T3 cells and may have therapeutic potential to cancer induced by cadmium.


Subject(s)
3T3 Cells/cytology , Cadmium Chloride/toxicity , Cell Transformation, Neoplastic/drug effects , Peptide Initiation Factors/pharmacology , Animals , Antisense Elements (Genetics) , Mice , Mice, Inbred BALB C , Mice, Nude , Peptide Initiation Factors/genetics , Proto-Oncogenes/genetics , Transfection
10.
FEBS Lett ; 495(3): 167-71, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11334885

ABSTRACT

Sequence determinants and structural features of the RNA govern mRNA-ribosome interaction in bacteria. However, ribosomal recruitment to leaderless mRNAs, which start directly with the AUG start codon and do not bear a Shine-Dalgarno sequence like canonical mRNAs, does not appear to rely on 16S rRNA-mRNA interactions. Here, we have studied the effects of translation initiation factors IF2 and IF3 on 30S initiation at a 5'-terminal AUG and at a competing downstream canonical ribosome binding site. We show that IF2 affects the forward kinetics of 30S initiation complex formation at the 5'-terminal AUG as well as the stability of these complexes. Moreover, the IF2:IF3 molar ratio was found to play a decisive role in translation initiation of a leaderless mRNA both in vitro and in vivo indicating that the translational efficiency of an mRNA is not only intrinsically determined but can be altered depending on the availability of components of the translational machinery.


Subject(s)
Codon, Initiator , Peptide Initiation Factors/pharmacology , Protein Biosynthesis , Ribosomes/metabolism , Escherichia coli/genetics , Eukaryotic Initiation Factor-3 , Genes, Reporter , Macromolecular Substances , Peptide Initiation Factors/metabolism , Prokaryotic Initiation Factor-2 , RNA, Transfer, Met/metabolism , Transformation, Genetic
11.
Curr Biol ; 10(13): 793-6, 2000 Jun 29.
Article in English | MEDLINE | ID: mdl-10898981

ABSTRACT

Overexpression of the translation initiation factor eIF4E leads to cell transformation and occurs in a number of human cancers [1]. mRNA translation and cell growth can be regulated through the availability of eIF4E to form initiation complexes by binding to eIF4G. The availability of eIF4E is blocked through the binding of members of a family of eIF4E-binding proteins (4E-BPs) [2] [3]. Indeed, cell transformation caused by the overexpression of eIF4E can be reversed by the overexpression of 4E-BPs [4] [5] [6] [7] [8]. To study the role of eIF4E in cell transformation, we developed a series of peptides based on the conserved eIF4E-binding motifs in 4E-BPs and eIF4G [9] linked to the penetratin peptide-carrier sequence, which mediates the rapid transport of peptides across cell membranes. Surprisingly, introduction of these eIF4E-binding peptides into MRC5 cells led to rapid, dose-dependent cell death, with characteristics of apoptosis. Single alanine substitutions at key positions in the peptides impair their binding to eIF4E and markedly reduce their ability to induce apoptosis. A triple alanine substitution, which abolishes binding to eIF4E, renders the peptide unable to induce apoptosis. Our data provide strong evidence that the peptides induce apoptosis through binding to eIF4E. They do not induce apoptosis through inhibition of protein synthesis, as chemical inhibitors of translation did not induce apoptosis or affect peptide-induced cell death. Thus these new data indicate that eIF4E has a direct role in controlling cell survival that is not linked to its known role in mRNA translation.


Subject(s)
Apoptosis/drug effects , Eukaryotic Initiation Factors , Oligopeptides/pharmacology , Peptide Initiation Factors/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/pharmacology , Cell Cycle Proteins , Cell Line , Cell Survival/drug effects , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Humans , In Situ Nick-End Labeling , Oligopeptides/metabolism , Pactamycin/pharmacology , Peptide Initiation Factors/chemistry , Peptide Initiation Factors/pharmacology , Phosphoproteins/chemistry , Phosphoproteins/pharmacology , Protein Binding , Protein Synthesis Inhibitors/pharmacology
12.
J Immunol ; 161(11): 6171-9, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834103

ABSTRACT

We have evaluated the ability of the Leishmania protein LeIF to influence the Th1/Th2 cytokine responses and the generation of LeIF-specific T cell clones in the absence of adjuvant. We characterized LeIF-specific T cell responses in Leishmania major-infected and uninfected BALB/c mice. These mice develop a strong Th2 response during infection with L. major. When lymph node cells from infected BALB/c mice were stimulated in vitro with LeIF, only IFN-gamma (and no detectable IL-4) was found in the culture supernatant. In addition, LeIF down-regulated Leishmania Ag-specific IL-4 production by lymph node cells from infected BALB/c mice. Subsequently, Th responses were evaluated in naive BALB/c mice following immunization with LeIF. T cell clones derived from mice immunized with LeIF preferentially secreted IFN-gamma. Finally, to understand the basis for the preferential Th1 cytokine bias observed with LeIF, the ability of LeIF to influence the early cytokine profile was evaluated in splenocytes of SCID mice. We found that LeIF stimulated fresh spleen cells from naive SCID mice to secrete IFN-gamma by IL-12/IL-18-dependent mechanisms. The N-terminal half of the molecule (amino acid residues 1-226) maintained the ability to stimulate IFN-gamma from splenocytes of SCID mice. Finally, we also demonstrated that LeIF was able to provide partial protection of BALB/c mice against L. major. Thus, our results suggest the potential of LeIF as a Th1-type adjuvant and as a therapeutic and prophylactic vaccine Ag for leishmaniasis when used with other leishmanial Ags.


Subject(s)
Interleukin-12/physiology , Leishmania major/immunology , Peptide Initiation Factors/pharmacology , Protozoan Proteins/pharmacology , Recombinant Proteins/pharmacology , Th1 Cells/metabolism , Adjuvants, Immunologic/pharmacology , Amino Acid Sequence , Animals , Antigens, Protozoan/pharmacology , Clone Cells , Cloning, Molecular , Down-Regulation/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-18/physiology , Interleukin-4/biosynthesis , Leishmania major/genetics , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/prevention & control , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, SCID , Molecular Sequence Data , Peptide Initiation Factors/genetics , Peptide Initiation Factors/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Spleen/cytology , Spleen/immunology
13.
J Mol Biol ; 281(2): 241-52, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9698545

ABSTRACT

A novel function of initiation factors IF1 and IF2 in Escherichia coli translation has been identified. It is shown that these factors efficiently catalyse dissociation of peptidyl-tRNAs with polypeptides of different length from the P-site of E. coli ribosomes, and that the simultaneous presence of both factors is required for induction of drop-off. The factor-induced drop-off occurs with both sense and stop codons in the A-site and competes with peptide elongation or termination. The efficiency with which IF1 and IF2 catalyse drop-off decreases with increasing length of the nascent polypeptide, but is quite significant for hepta-peptidyl-tRNAs, the longest polypeptide chains studied. In the absence of IF1 and IF2 the rate of drop-off varies considerably for different peptidyl-tRNAs, and depends both on the length and sequence of the nascent peptide. Efficient factor-catalysed drop-off requires GTP but not GTP hydrolysis, as shown in experiments without guanine nucleotides, with GDP or with the non-cleavable analogue GMP-PNP.Simultaneous overexpression of IF1 and IF2 in vivo inhibits cell growth specifically in some peptidyl-tRNA hydrolase deficient mutants, suggesting that initiation factor-catalysed drop-off of peptidyl-tRNA can occur on a significant scale in the bacterial cell. Consequences for the bacterial physiology of this previously unknown function of IF1 and IF2 are discussed.


Subject(s)
Escherichia coli/genetics , Oligopeptides/metabolism , Peptide Initiation Factors/metabolism , Protein Biosynthesis/physiology , Ribosomes/metabolism , Carboxylic Ester Hydrolases/metabolism , Escherichia coli/growth & development , Guanosine Triphosphate/physiology , Peptide Initiation Factors/pharmacology , Prokaryotic Initiation Factor-2 , RNA, Transfer, Amino Acyl/metabolism , RNA, Transfer, Met/metabolism
14.
Genes Dev ; 12(1): 67-83, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9420332

ABSTRACT

Initiation of translation of hepatitis C virus and classical swine fever virus mRNAs results from internal ribosomal entry. We reconstituted internal ribosomal entry in vitro from purified translation components and monitored assembly of 48S ribosomal preinitiation complexes by toe-printing. Ribosomal subunits (40S) formed stable binary complexes on both mRNAs. The complex structure of these RNAs determined the correct positioning of the initiation codon in the ribosomal "P" site in binary complexes. Ribosomal binding and positioning on these mRNAs did not require the initiation factors eIF3, eIF4A, eIF4B, and eIF4F and translation of these mRNAs was not inhibited by a trans-dominant eIF4A mutant. Addition of Met-tRNAiMet, eIF2, and GTP to these binary ribosomal complexes resulted in formation of 48S preinitiation complexes. The striking similarities between this eukaryotic initiation mechanism and the mechanism of translation initiation in prokaryotes are discussed.


Subject(s)
Classical Swine Fever Virus/genetics , Codon, Initiator , Eukaryotic Initiation Factors , Hepacivirus/genetics , Protein Biosynthesis , RNA, Viral/metabolism , Ribosomes/metabolism , Animals , Base Sequence , Cytoplasm/metabolism , DNA Primers , Eukaryotic Cells , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/pharmacology , Eukaryotic Initiation Factor-3 , Eukaryotic Initiation Factor-4A , Eukaryotic Initiation Factor-4F , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Peptide Chain Initiation, Translational , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/pharmacology , Prokaryotic Cells , RNA, Transfer, Met/metabolism , Rabbits , Ribosomal Protein S9 , Ribosomal Proteins/metabolism , Structure-Activity Relationship
15.
Eur J Immunol ; 27(10): 2634-42, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9368620

ABSTRACT

LeIF, a gene homologue of the eukaryotic initiation factor 4A was first described as a leishmanial antigen that induced a Th1-type T cell response in peripheral blood mononuclear cells (PBMC) from leishmaniasis patients. Moreover, the interferon (IFN)-gamma production by PBMC was found to be interleukin (IL)-12 dependent. Herein, we characterize the effects of LeIF on cytokine production and expression of surface molecules by normal human monocytes as well as by monocyte-derived macrophages and dendritic cells (MoDC). LeIF was a strong inducer of IL-12 and, to a lesser extent, of IL-10 and tumor necrosis factor (TNF)-alpha in macrophages and MoDC. IL-12 production did not require CD40 triggering, confirming that the ability of LeIF to induce IL-12 was not mediated through an effect on T cells. However, addition of soluble CD40 ligand (L) synergistically augmented IL-12 production in macrophages and MoDC. The cytokine-inducing activity of LeIF is located in the N-terminal portion of the molecule and was both proteinase K sensitive and polymyxin B resistant. LeIF, lipopolysaccharide and fixed Staphylococcus aureus all induced comparable amounts of IL-12, validating the potent cytokine-inducing effects of LeIF. Moreover, of these stimuli, LeIF had the highest IL-12/IL-10 and IL-12/TNF-alpha ratio demonstrating the preference of LeIF for IL-12 induction. Studies investigating the expression of surface molecules showed that LeIF up-regulated B7-1 and CD54 (ICAM-1) on macrophages and MoDC. To our knowledge this is the first report describing IL-12 production, up-regulation of co-stimulatory and intercellular adhesion molecules by monocytic antigen-presenting cells in response to a protein from a pathogenic microorganism. These immunomodulatory characteristics of LeIF might be excellent properties for a Th1-type adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigen-Presenting Cells/drug effects , B7-1 Antigen/biosynthesis , Gene Expression Regulation/drug effects , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Leishmania major/immunology , Macrophages/drug effects , Peptide Initiation Factors/pharmacology , Protozoan Proteins/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Antigen-Presenting Cells/metabolism , B7-1 Antigen/genetics , CD40 Ligand , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-12/genetics , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Membrane Glycoproteins/pharmacology , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
EMBO J ; 16(13): 4107-16, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9233819

ABSTRACT

For recognition by the translational machinery, most eukaryotic cellular mRNAs have a 5' cap structure [e.g. m7G(5')ppp(5')N]. We describe a translation enhancer sequence (3'TE) located in the 3'-untranslated region (UTR) of the genome of the PAV barley yellow dwarf virus (BYDV-PAV) which stimulates translation from uncapped mRNA by 30- to 100-fold in vitro and in vivo to a level equal to that of efficient capped mRNAs. A four base duplication within the 3'TE destroyed the stimulatory activity. Efficient translation was recovered by addition of a 5' cap to this mRNA. Translation of both uncapped mRNA containing the 3'TE in cis and capped mRNA lacking any BYDV-PAV sequence was inhibited specifically by added 3'TE RNA in trans. This inhibition was reversed by adding initiation factor 4F (eIF4F), suggesting that the 3'TE, like the 5' cap, mediates eIF4F-dependent translation initiation. The BYDV-PAV 5'UTR was necessary for the 3'TE to function, except when the 3'TE itself was moved to the 5'UTR. Thus, the 3'TE is sufficient for recruiting the translation factors and ribosomes, while the viral 5'UTR may serve only for the long distance 3'-5' communication. Models are proposed to explain this novel mechanism of cap-independent translation initiation facilitated by the 3'UTR.


Subject(s)
Luteovirus/genetics , Protein Biosynthesis , RNA Caps , RNA, Messenger/metabolism , RNA, Viral/metabolism , Conserved Sequence , Eukaryotic Initiation Factor-4F , Genome, Viral , Introns , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/pharmacology
17.
EMBO J ; 16(4): 844-55, 1997 Feb 17.
Article in English | MEDLINE | ID: mdl-9049313

ABSTRACT

A common feature of viral infection is the subversion of the host cell machinery towards the preferential translation of viral products. In some instances, this is partly mediated by the expression of virally encoded proteases which lead to the cleavage of initiation factor eIF4G. The foot-and-mouth disease virus encodes two forms of a cysteine proteinase (L protease) which bisects the eIF4G polypeptide into an N-terminal fragment containing the eIF4E binding site, and a C-terminal fragment which contains binding sites for eIF4A and eIF3 and which associates with the 40S ribosomal subunit. Previously, we have demonstrated that the cleavage of eIF4G by L protease stimulates the translation of uncapped transcripts encoding cellular proteins and supports internal initiation driven by picornavirus internal ribosome entry segment (IRES) elements. Use of reticulocyte lysates manipulated to deplete them of eIF4E and the N-terminal fragment suggests that the C-terminal fragment of eIF4G is responsible for these effects, and we have now confirmed this by purifying the C-terminal fragment and analysing its effects directly in the absence of L protease. Interestingly, we find that pre-incubation of reticulocyte lysates or ribosomal salt wash fractions with the specific eIF4E binding protein, PHAS-I (eIF4E-BP1), blocks the proteolytic cleavage of eIF4G by L protease. This effect can be reversed by addition of recombinant eIF4E. These data are consistent with a model whereby the L protease cleavage site in eIF4G is inaccessible until a change in conformation is induced by the binding of eIF4E. This may have implications for a role for eIF4E binding in triggering changes that expose other domains in the eIF4G molecule during initiation of translation.


Subject(s)
Carrier Proteins , Peptide Initiation Factors/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis/physiology , Animals , Cell-Free System , Cyclins/genetics , Endopeptidases/metabolism , Endopeptidases/pharmacology , Eukaryotic Initiation Factor-4E , Eukaryotic Initiation Factor-4G , Peptide Fragments , Peptide Initiation Factors/genetics , Peptide Initiation Factors/pharmacology , Phosphoproteins/pharmacology , RNA Caps , RNA, Messenger/metabolism , Rabbits , Recombinant Fusion Proteins , Reticulocytes , Ribosomes/metabolism
18.
Mol Cell Biol ; 15(8): 4525-35, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7623843

ABSTRACT

The Saccharomyces cerevisiae PRT1 gene product Prt1p is a component of translation initiation factor eIF-3, and mutations in PRT1 inhibit translation initiation. We have investigated structural and functional aspects of Prt1p and its gene. Transcript analysis and deletion of the PRT1 5' end revealed that translation of PRT1 mRNA is probably initiated at the second in-frame ATG in the open reading frame. The amino acid changes encoded by six independent temperature-sensitive prt1 mutant alleles were found to be distributed throughout the central and C-terminal regions of Prt1p. The temperature sensitivity of each mutant allele was due to a single missense mutation, except for the prt1-2 allele, in which two missense mutations were required. In-frame deletion of an N-terminal region of Prt1p generated a novel, dominant-negative form of Prt1p that inhibits translation initiation even in the presence of wild-type Prt1p. Subcellular fractionation suggested that the dominant-negative Prt1p competes with wild-type Prt1p for association with a component of large Prt1p complexes and as a result inhibits the binding of wild-type Prt1p to the 40S ribosome.


Subject(s)
Fungal Proteins/genetics , Peptide Chain Initiation, Translational , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Alleles , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Mutational Analysis , Eukaryotic Initiation Factor-3 , Fungal Proteins/pharmacology , Macromolecular Substances , Molecular Sequence Data , Open Reading Frames/genetics , Peptide Chain Initiation, Translational/drug effects , Peptide Fragments/pharmacology , Peptide Initiation Factors/pharmacology , Polymorphism, Genetic , Prokaryotic Initiation Factor-3 , Protein Synthesis Inhibitors/pharmacology , RNA Precursors/genetics , RNA-Binding Proteins/metabolism , Sequence Analysis, DNA , Sequence Deletion , Transcription, Genetic
19.
Nucleic Acids Res ; 23(3): 334-40, 1995 Feb 11.
Article in English | MEDLINE | ID: mdl-7885827

ABSTRACT

Infection of cells with the foot-and-mouth-disease virus, a member of the picornavirus family, results in the shut-off of host protein synthesis. A major contributory mechanism is the proteolytic destruction of the gamma subunit of the complex eIF-4, which functions in translation to promote the binding of the 43S ribosomal preinitiation complex to the 5' end of the cellular mRNA molecules bearing a 5' terminal cap structure. Picornavirus RNA molecules, which are uncapped, use a distinct mechanism for translational initiation, which can operate in the absence, or at low levels, of eIF-4. The proteolysis of eIF-4 gamma in cells infected by foot-and-mouth-disease virus results from expression of a virus-encoded cysteine proteinase known as Leader (or L) protease. We have used a transcription plasmid encoding this protease as a tool to deplete in vitro translation systems of eIF-4 gamma in order to elucidate in more detail the role of this polypeptide in the control of translation. Using in vitro transcribed mRNAs we have observed a marked contrast between capped and uncapped transcripts in the response of their translation to the proteolysis of eIF-4 gamma. Translation of capped mRNAs is, as expected, severely impaired, and is restored by addition of eIF-4 complex containing the intact gamma-subunit. On the other hand, translation of uncapped transcripts, normally inefficient, is substantially enhanced. The data suggest that the translation of uncapped mRNAs may be stimulated in this system by one or more of the proteolytic degradation products of eIF-4 gamma.


Subject(s)
CDC2-CDC28 Kinases , Peptide Initiation Factors/metabolism , Protein Biosynthesis/physiology , RNA Caps/genetics , RNA, Messenger/genetics , Viral Proteins , 3C Viral Proteases , Animals , Cell-Free System , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/pharmacology , Peptide Initiation Factors/pharmacology , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/genetics , Rabbits , Reticulocytes
20.
J Virol ; 68(3): 1544-50, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8107217

ABSTRACT

Initiation of poliovirus RNA translation by internal entry of ribosomes is believed to require the participation of trans-acting factors. The mechanism of action of these factors is poorly defined. The limiting amount of one of these factors, La protein, in rabbit reticulocyte lysates (RRL) has been postulated to partially explain the inefficient translation of poliovirus RNA in this system. To further characterize La activity in translation and to identify other potential limiting factors, we assayed the ability of La protein as well as purified initiation factors, eIF-2, guanine nucleotide exchange factor (GEF), eIF-4A, eIF-4B, eIF-4F, and eIF-3, to stimulate the synthesis of P1, the capsid precursor protein, in poliovirus type 1 (Mahoney) RNA-programmed RRL. Of the proteins tested, only La, GEF, and to some extent eIF-2 stimulated the synthesis of P1. The enhanced translation of P1 in response to La occurred concomitantly with the inhibition of synthesis of most aberrant polypeptides, resulting from initiation in the middle of the genome. Deletion of the carboxy-terminal half (214 amino acids) of La did not decrease its binding to the poliovirus 5' untranslated region but abrogated the stimulatory and correcting activity in translation. In contrast to La, GEF and eIF-2 stimulated the overall translation and increased the synthesis of aberrant products as well as P1. Neither La, GEF, nor any other factor stimulated translation of encephalomyocarditis virus RNA in RRL. The implications of these findings for the mechanism of internal translation initiation on picornavirus RNAs are discussed.


Subject(s)
Autoantigens/pharmacology , Peptide Initiation Factors/pharmacology , Poliovirus/metabolism , Protein Biosynthesis/drug effects , Ribonucleoproteins/pharmacology , Animals , Autoantigens/genetics , Cell-Free System , Eukaryotic Initiation Factor-2/pharmacology , Guanine Nucleotide Exchange Factors , Mutation , Peptide Chain Initiation, Translational/drug effects , Proteins/pharmacology , Rabbits , Reticulocytes , Ribonucleoproteins/genetics , Sequence Deletion , SS-B Antigen
SELECTION OF CITATIONS
SEARCH DETAIL
...