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










Publication year range
1.
Trends Biochem Sci ; 44(12): 1009-1021, 2019 12.
Article in English | MEDLINE | ID: mdl-31353284

ABSTRACT

Selection of the translation initiation site (TIS) is a crucial step during translation. In the 1980s Marylin Kozak performed key studies on vertebrate mRNAs to characterize the optimal TIS consensus sequence, the Kozak motif. Within this motif, conservation of nucleotides in crucial positions, namely a purine at -3 and a G at +4 (where the A of the AUG is numbered +1), is essential for TIS recognition. Ever since its characterization the Kozak motif has been regarded as the optimal sequence to initiate translation in all eukaryotes. We revisit here published in silico data on TIS consensus sequences, as well as experimental studies from diverse eukaryotic lineages, and propose that, while the -3A/G position is universally conserved, the remaining variability of the consensus sequences enables their classification as optimal, strong, and moderate TIS sequences.


Subject(s)
Codon, Initiator/physiology , Eukaryota/physiology , Nucleotide Motifs , Peptide Chain Initiation, Translational/physiology , RNA, Messenger/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L58-75, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25326576

ABSTRACT

Surfactant protein A (SP-A), a molecule with roles in lung innate immunity and surfactant-related functions, is encoded by two genes in humans: SFTPA1 (SP-A1) and SFTPA2 (SP-A2). The mRNAs from these genes differ in their 5'-untranslated regions (5'-UTR) due to differential splicing. The 5'-UTR variant ACD' is exclusively found in transcripts of SP-A1, but not in those of SP-A2. Its unique exon C contains two upstream AUG codons (uAUGs) that may affect SP-A1 translation efficiency. The first uAUG (u1) is in frame with the primary start codon (p), but the second one (u2) is not. The purpose of this study was to assess the impact of uAUGs on SP-A1 expression. We employed RT-qPCR to determine the presence of exon C-containing SP-A1 transcripts in human RNA samples. We also used in vitro techniques including mutagenesis, reporter assays, and toeprinting analysis, as well as in silico analyses to determine the role of uAUGs. Exon C-containing mRNA is present in most human lung tissue samples and its expression can, under certain conditions, be regulated by factors such as dexamethasone or endotoxin. Mutating uAUGs resulted in increased luciferase activity. The mature protein size was not affected by the uAUGs, as shown by a combination of toeprint and in silico analysis for Kozak sequence, secondary structure, and signal peptide and in vitro translation in the presence of microsomes. In conclusion, alternative splicing may introduce uAUGs in SP-A1 transcripts, which in turn negatively affect SP-A1 translation, possibly affecting SP-A1/SP-A2 ratio, with potential for clinical implication.


Subject(s)
Alternative Splicing/physiology , Codon, Initiator/physiology , Lung/metabolism , Pulmonary Surfactant-Associated Protein A/biosynthesis , RNA, Messenger/biosynthesis , Alternative Splicing/drug effects , Anti-Inflammatory Agents/pharmacology , Cell Line, Tumor , Dexamethasone/pharmacology , Endotoxins/pharmacology , Exons/physiology , Female , Humans , Male , Microsomes/metabolism , Mutation , Protein Sorting Signals/physiology , Pulmonary Surfactant-Associated Protein A/genetics , RNA, Messenger/genetics
3.
RNA ; 17(3): 439-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21220547

ABSTRACT

Translation initiation in eukaryotes involves more than a dozen protein factors. Alterations in six factors have been found to reduce the fidelity of start codon recognition by the ribosomal preinitiation complex in yeast, a phenotype referred to as Sui(-). No small molecules are known that affect the fidelity of start codon recognition. Such compounds would be useful tools for probing the molecular mechanics of translation initiation and its regulation. To find compounds with this effect, we set up a high-throughput screen using a dual luciferase assay in S. cerevisiae. Screening of over 55,000 compounds revealed two structurally related molecules that decrease the fidelity of start codon selection by approximately twofold in the dual luciferase assay. This effect was confirmed using additional in vivo assays that monitor translation from non-AUG start codons. Both compounds increase translation of a natural upstream open reading frame previously shown to initiate translation at a UUG. The compounds were also found to exacerbate increased use of UUG as a start codon (Sui(-) phenotype) conferred by haploinsufficiency of wild-type eukaryotic initiation factor (eIF) 1, or by mutation in eIF1. Furthermore, the effects of the compounds are suppressed by overexpressing eIF1, which is known to restore the fidelity of start codon selection in strains harboring Sui(-) mutations in various other initiation factors. Together, these data strongly suggest that the compounds affect the translational machinery itself to reduce the accuracy of selecting AUG as the start codon.


Subject(s)
Codon, Initiator/physiology , Eukaryotic Initiation Factor-1/metabolism , Heterocyclic Compounds/pharmacology , Peptide Chain Initiation, Translational/drug effects , Peptide Chain Initiation, Translational/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Eukaryotic Initiation Factor-1/genetics , Haploinsufficiency , Luciferases/metabolism , Mutation/genetics , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Small Molecule Libraries
4.
J Biol Chem ; 285(7): 4595-602, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20018845

ABSTRACT

Mammalian cytosolic and mitochondrial thioredoxin reductases are essential selenocysteine-containing enzymes that control thioredoxin functions. Thioredoxin/glutathione reductase (TGR) is a third member of this enzyme family. It has an additional glutaredoxin domain and shows highest expression in testes. Herein, we found that human and several other mammalian TGR genes lack any AUG codons that could function in translation initiation. Although mouse and rat TGRs have such codons, we detected protein sequences upstream of them by immunoblot assays and direct proteomic analyses. Further gene engineering and expression analyses demonstrated that a CUG codon, located upstream of the sequences previously thought to initiate translation, is the actual start codon in mouse TGR. The use of this codon relies on the Kozak consensus sequence and ribosome-scanning mechanism. However, CUG serves as an inefficient start codon that allows downstream initiation, thus generating two isoforms of the enzyme in vivo and in vitro. The use of CUG evolved in mammalian TGRs, and in some of these organisms, GUG is used instead. The newly discovered longer TGR form shows cytosolic localization in cultured cells and is expressed in spermatids in mouse testes. This study shows that CUG codon is used as an inefficient start codon to generate protein isoforms in mouse.


Subject(s)
Isoenzymes/genetics , Multienzyme Complexes/genetics , NADH, NADPH Oxidoreductases/genetics , Testis/enzymology , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Codon, Initiator/genetics , Codon, Initiator/physiology , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/metabolism , NIH 3T3 Cells , Rats , Seminiferous Epithelium/metabolism , Sequence Homology, Amino Acid
5.
J Neurochem ; 104(6): 1663-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18205747

ABSTRACT

Peripherin is a type III neuronal intermediate filament protein detected within the intraneuronal inclusions characteristic of amyotrophic lateral sclerosis. The constitutively expressed peripherin isoform is encoded by all nine exons of the human and mouse peripherin genes to generate a protein species of approximately 58 kDa on sodium dodecyl sulfate-polyacrylamide gels. Expression of this isoform, termed Per-58, generates a filament network in transfected SW13 vim cells. On immunoblots of cell lysates derived from these transfected cells, we have consistently observed a second peripherin species of approximately 45 kDa. In this study, we show that this species is a novel peripherin isoform generated through the use of an in-frame downstream initiation codon. This isoform, that we have designated Per-45, is co-expressed together with Per-58 and, thus, constitutive in both human and mouse. Using mutational analysis, we show that Per-45 is required for normal network formation, with the absence of Per-45 leading to irregular filamentous structures. We further show that peripherin expression in the normal nervous system is characterized by tissue-specific Per-58 : Per-45 isoform ratios. Taken together, these results identify novel processing requirements for peripherin expression and indicate a hitherto unrecognized role for neuronal intermediate filament network formation through intra-isoform associations.


Subject(s)
Intermediate Filament Proteins/genetics , Intermediate Filaments/physiology , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Neurons/physiology , Protein Biosynthesis/physiology , Adrenal Gland Neoplasms , Animals , Cell Line, Tumor , Codon, Initiator/physiology , DNA, Complementary , Female , Gene Expression/physiology , Humans , Intermediate Filament Proteins/chemistry , Isomerism , Male , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Neurons/cytology , Peripherins , Plasmids
6.
BMC Mol Biol ; 8: 90, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17937806

ABSTRACT

BACKGROUND: ADAM15 is a metalloprotease-disintegrin implicated in ectodomain shedding and cell adhesion. Aberrant ADAM15 expression has been associated with human cancer and other disorders. We have previously shown that the alternative splicing of ADAM15 transcripts is mis-regulated in cancer cells. To gain a better understanding of ADAM15 regulation, its genomic organization and regulatory elements as well as the alternative exon use in human tissues were characterized. RESULTS: Human ADAM15, flanked by the FLJ32785/DCST1 and ephrin-A4 genes, spans 11.4 kb from the translation initiation codon to the polyadenylation signal, being the shortest multiple-exon ADAM gene. The gene contains 23 exons varying from 63 to 316 bp and 22 introns from 79 to 1283 bp. The gene appeared to have several transcription start sites and their location suggested the promoter location within a CpG island proximal to the translation start. Reporter expression experiments confirmed the location of functional GC-rich, TATAless and CAATless promoter, with the most critical transcription-supporting elements located -266 to -23 bp relative to the translation start. Normal human tissues showed different complex patterns of at least 13 different ADAM15 splice variants arising from the alternative use of the cytosolic-encoding exons 19, 20a/b, and 21a/b. The deduced ADAM15 protein isoforms have different combinations of cytosolic regulatory protein interaction motifs. CONCLUSION: Characterization of human ADAM15 gene and identification of elements involved in the regulation of transcription and alternative splicing provide important clues for elucidation of physiological and pathological roles of ADAM15. The present results also show that the alternative exon use is a physiological post-transcriptional mechanism regulating ADAM15 expression in human tissues.


Subject(s)
ADAM Proteins/genetics , Alternative Splicing/physiology , Exons/physiology , Gene Expression Regulation, Enzymologic/physiology , Membrane Proteins/genetics , ADAM Proteins/biosynthesis , Amino Acid Motifs/genetics , Cell Adhesion/physiology , Cell Line , Codon, Initiator/physiology , CpG Islands/physiology , Ephrin-A4/genetics , Ephrin-A4/metabolism , Humans , Membrane Proteins/biosynthesis , Neoplasms/enzymology , Neoplasms/genetics , Organ Specificity/physiology , Promoter Regions, Genetic/physiology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
7.
Genes Dev ; 21(10): 1217-30, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17504939

ABSTRACT

Selection of the AUG start codon is a key step in translation initiation requiring hydrolysis of GTP in the eIF2*GTP*Met-tRNA(i)(Met) ternary complex (TC) and subsequent P(i) release from eIF2*GDP*P(i). It is thought that eIF1 prevents recognition of non-AUGs by promoting scanning and blocking P(i) release at non-AUG codons. We show that Sui(-) mutations in Saccharomyces cerevisiae eIF1, which increase initiation at UUG codons, reduce interaction of eIF1 with 40S subunits in vitro and in vivo, and both defects are diminished in cells by overexpressing the mutant proteins. Remarkably, Sui(-) mutation ISQLG(93-97)ASQAA (abbreviated 93-97) accelerates eIF1 dissociation and P(i) release from reconstituted preinitiation complexes (PICs), whereas a hyperaccuracy mutation in eIF1A (that suppresses Sui(-) mutations) decreases the eIF1 off-rate. These findings demonstrate that eIF1 dissociation is a critical step in start codon selection, which is modulated by eIF1A. We also describe Gcd(-) mutations in eIF1 that impair TC loading on 40S subunits or destabilize the multifactor complex containing eIF1, eIF3, eIF5, and TC, showing that eIF1 promotes PIC assembly in vivo beyond its important functions in AUG selection.


Subject(s)
Codon, Initiator/physiology , Eukaryotic Initiation Factor-1/metabolism , Protein Biosynthesis/physiology , Ribosome Subunits, Small, Eukaryotic/metabolism , Saccharomyces cerevisiae/genetics , Codon, Initiator/genetics , Electrophoresis, Polyacrylamide Gel , Eukaryotic Initiation Factor-1/genetics , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics
8.
Int J Immunogenet ; 34(2): 107-13, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17373935

ABSTRACT

Interleukin 7 (IL-7) is a key factor in the survival, development and proliferation of B and T lymphocytes. Elevation of plasma IL-7 has been reported in several lymphopenia cases such as HIV-1 patients. After patients started to receive antiretroviral drugs and their CD4(+) cell counts had recovered, IL-7 in plasma decreased to normal levels. There are considerable variations in the levels of plasma IL-7 as well as the rate of CD4(+) T-cell restoration. Although pre-treatment plasma IL-7 levels have been shown to be prognostic for the rate of post-treatment CD4(+) T-cell restoration, the mechanisms responsible for the variations in plasma IL-7 and rate of CD4(+) T-cell restoration are still completely unknown. In the study here, we searched for genetic polymorphisms that might affect levels of IL-7 gene expression. For this purpose, we used 1658-bp PCR-amplified fragments of the IL-7 gene containing 1470 bp of the upstream non-coding region obtained from 151 Japanese and 234 Thai subjects. We found two novel human genetic polymorphisms in the upstream non-coding region of the IL-7 gene. The luciferase reporter assay demonstrated that one of those polymorphisms could increase the gene expression of IL-7. We speculate that this polymorphism, a three base ATC deletion just upstream of an out-of-frame ATG codon in the upstream non-coding region of the IL-7 gene, reduces the efficiency of translation from the upstream, out-of-frame ATG, resulting in increased translation efficiency from the authentic ATG of IL-7. Although the frequency of this allele is very low, it would be interesting to analyse this polymorphism in HIV-1-infected individuals with different rates of immune reconstitution after treatment with a highly active antiretroviral therapy.


Subject(s)
Codon, Initiator/genetics , Interleukin-7/genetics , Polymorphism, Genetic , Protein Biosynthesis/genetics , Untranslated Regions/genetics , Base Sequence , Codon, Initiator/physiology , Gene Expression Regulation , Gene Frequency , Genes, Reporter , Genotype , Humans , Luciferases/genetics , Molecular Sequence Data , Sequence Deletion
9.
Sci STKE ; 2007(367): re1, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-17200520

ABSTRACT

Several thousand human genes, amounting to about one-third of the whole genome, are potential targets for regulation by the several hundred microRNAs (miRNAs) encoded in the genome. The regulation occurs posttranscriptionally and involves the approximately 21-nucleotide miRNA interacting with a target site in the mRNA that generally has imperfect complementarity to the miRNA. The target sites are almost invariably in the 3'-untranslated region of the messenger RNA (mRNA), often in multiple copies. Metazoan miRNAs were previously thought to down-regulate protein expression by inhibiting target mRNA translation at some stage after the translation initiation step, without much effect on mRNA abundance. However, recent studies have questioned these suppositions. With some targets, an increase in the rate of mRNA degradation by the normal decay pathway contributes to the decrease in protein expression. miRNAs can also inhibit translation initiation, specifically the function of the cap-binding initiation factor, eIF4E. Repressed target mRNAs as well as miRNAs themselves accumulate in cytoplasmic foci known as P-bodies, where many enzymes involved in mRNA degradation are concentrated. However, P-bodies may also serve as repositories for the temporary and reversible storage of untranslated mRNA, and reducing the expression (knockdown) of several distinct P-body protein components can alleviate miRNA-mediated repression of gene expression.


Subject(s)
Gene Expression Regulation/physiology , MicroRNAs/physiology , Animals , Base Sequence , Codon, Initiator/physiology , Down-Regulation , Eukaryotic Initiation Factor-4E/physiology , Humans , Models, Biological , Protein Biosynthesis/physiology , RNA Stability/physiology , RNA, Small Interfering/physiology , Sequence Alignment
10.
Mol Cell Biol ; 26(9): 3353-64, 2006 May.
Article in English | MEDLINE | ID: mdl-16611980

ABSTRACT

beta-Site beta-amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) is the beta-secretase in vivo for processing APP to generate amyloid beta protein (Abeta). Abeta deposition in the brain is the hallmark of Alzheimer's disease (AD) neuropathology. Inhibition of BACE1 activity has major pharmaceutical potential for AD treatment. The expression of the BACE1 gene is relatively low in vivo. The control of BACE1 expression has not been well defined. There are six upstream AUGs (uAUGs) in the 5' leader sequence of the human BACE1 mRNA. We investigated the role of the promoter and the uATGs in the 5' untranslated region (UTR) of the human BACE1 gene in BACE1 gene transcription and translation initiation. Our results show that the first and second uATGs are the integral part of the core minimal promoter of the human BACE1 gene, while the third uAUG is skipped over by ribosomal scanning. The fourth uAUG can function as a translation initiation codon, and deletion or mutation of this uAUG increases downstream gene expression. The fourth uAUG of the BACE1 5'UTR is responsible for inhibiting the expression of BACE1. Translation initiation by the BACE1 uAUGs and physiological AUG requires intact eIF4G. Our results demonstrate that during human BACE1 gene expression, ribosomes skipped some uAUGs by leaky scanning and translated an upstream open reading frame, initiated efficiently at the fourth uAUG, and subsequently reinitiated BACE1 translation at the physiological AUG site. Such leaky scanning and reinitiation resulted in weak expression of BACE1 under normal conditions. Alterations of the leaky scanning and reinitiation in BACE1 gene expression could play an important role in AD pathogenesis.


Subject(s)
Codon, Initiator/physiology , Endopeptidases/genetics , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Protein Biosynthesis/genetics , 5' Untranslated Regions/genetics , Amyloid Precursor Protein Secretases , Apoptosis Regulatory Proteins/pharmacology , Aspartic Acid Endopeptidases , Base Sequence , Cysteine Endopeptidases/pharmacology , Humans , Membrane Glycoproteins/pharmacology , Molecular Sequence Data , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/pharmacology , Viral Proteins/pharmacology
11.
Mol Cell Endocrinol ; 249(1-2): 21-31, 2006 04 25.
Article in English | MEDLINE | ID: mdl-16504375

ABSTRACT

At least four alternatively spliced mRNAs can be synthesized from the human AT(1)R (hAT(1)R) gene that differ only in the inclusion or exclusion of exon 2 and/or 3. RT-PCR experiments demonstrate that splice variants harboring exon 2 accounts for at least 30% of all the hAT(1)R mRNA transcripts expressed in the human tissues investigated. Since exon 2 contains two upstream AUGs or open reading frames (uORFs), we hypothesized that these AUGs would inhibit the translation of the downstream hAT(1)R protein ORF harbored in exon 4. This study demonstrates that the inclusion of exon 2 in hAT(1)R mRNA transcripts dramatically reduces hAT(1)R protein levels (nine-fold) and significantly attenuates Ang II responsiveness ( approximately four-fold). Interestingly, only when both AUGs were mutated in combination were the hAT(1)R density and Ang II signaling levels comparable with those values obtained using mRNA splice variants that did not include exon 2. This observation is consistent with a model where the majority of the ribosomes likely translate uORF#1 and are then unable to reinitiate at the downstream hAT(1)R ORF, in part due to the presence of AUG#2 and to the short intercistronic spacing. Importantly, TGF-beta(1) treatment (4ng/ml for 4h) of fibroblasts up-regulated hAT(1)R mRNA splice variants, which harbored exon 2, six-fold. Since AT(1)R activation is closely associated with cardiovascular disease, the inclusion of exon 2 by alternative splicing represents a novel mechanism to reduce the overall production of the hAT(1)R protein and possibly limit the potential pathological effects of AT(1)R activation.


Subject(s)
Alternative Splicing , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1/genetics , Transforming Growth Factor beta/pharmacology , Up-Regulation , Base Sequence , Codon, Initiator/physiology , Exons , Humans , Molecular Sequence Data , Receptor, Angiotensin, Type 1/biosynthesis , Sequence Alignment , Transforming Growth Factor beta/physiology
12.
Biochim Biophys Acta ; 1575(1-3): 54-62, 2002 May 03.
Article in English | MEDLINE | ID: mdl-12020819

ABSTRACT

Apobec-1 catalyzes C to U editing of apolipoprotein B (apoB) mRNA in the mammalian intestine. Rat apobec-1 is transcribed from three distinct promoters, which contain distinct 5' untranslated regions (5'UTRs) accompanied by variable numbers of in-frame upstream AUGs (uAUGs). We have observed a shift in apobec-1 promoter usage in an experimental model of colon carcinogenesis, resulting in transcripts loaded with 5'AUGs. In colon cancer, apobec-1 protein levels decreased by 90% in the cancer tissue as compared to normal tissue, suggesting an inhibitory effect of the 5'UTR on apobec-1 translation. We investigated the effects of these different 5'UTRs by site-directed mutagenesis coupled with in vitro translation studies. These studies established that the uAUGs within the 5'UTR of the alternative transcripts inhibit apobec-1 translation. This effect was independent of the length of the 5'UTR. Further analysis demonstrated that these uAUGs altered the polysome distribution, shifting the mRNA towards a denser, post-polyribosomal fraction. These findings were confirmed in transient transfection studies in vivo using HepG2 cells, where functional expression of apobec-1 was restored by mutagenesis of the uAUGs. Taken together, these data imply that rat apobec-1 gene expression is downregulated through alternative promoter usage. This dominant translational control of apobec-1 gene expression is most plausibly exerted through uAUGs.


Subject(s)
Alternative Splicing , Codon, Initiator/genetics , Colonic Neoplasms/genetics , Cytidine Deaminase/genetics , Gene Expression Regulation, Neoplastic/genetics , Transcription, Genetic , 5' Untranslated Regions/genetics , APOBEC-1 Deaminase , Alternative Splicing/genetics , Animals , Codon, Initiator/physiology , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/physiology , Male , Polyribosomes/genetics , Polyribosomes/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , Rats , Rats, Inbred F344
13.
Nat Cell Biol ; 4(6): 462-7, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032546

ABSTRACT

Activation of the p53 tumour suppressor protein can lead to cell-cycle arrest or apoptosis. p53 function is controlled by the mdm2 oncogene product, which targets p53 for proteasomal degradation. In this report we demonstrate that Mdm2 induces translation of the p53 mRNA from two alternative initiation sites, giving full-length p53 and another protein with a relative molecular mass (M(r)) of approximately 47K; we designate this protein as p53/47. This translation induction requires Mdm2 to interact directly with the nascent p53 polypeptide. The alternatively translated p53/47 does not contain the Mdm2-binding site and it lacks the most amino-terminal transcriptional-activation domain of p53. Increased expression of p53/47 stabilizes p53 in the presence of Mdm2, and alters the expression levels of p53-induced gene products. These results show how the interaction of Mdm2 with p53 leads to a change in the ratio of full-length p53 to p53/47 by inducing translation of both p53 proteins and the subsequent selective degradation of full-length p53. Thus, Mdm2 controls the expression levels of p53 through a dual mechanism that involves induction of synthesis and targeting for degradation.


Subject(s)
Nuclear Proteins , Protein Biosynthesis/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Apoptosis/physiology , Breast Neoplasms , Codon, Initiator/physiology , Female , Fibroblasts/cytology , Gene Expression Regulation/physiology , Humans , Lung Neoplasms , Molecular Sequence Data , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2 , RNA, Ribosomal, 5S/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/chemistry
14.
Exp Eye Res ; 73(6): 827-36, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11846513

ABSTRACT

Lens development and response to peroxide stress are associated with dramatic changes in protein ubiquitination, reflecting dynamic changes in activity of the ubiquitin-activating enzyme (E1). Two isoforms of E1 (E1A and E1B) have been identified in lens cells although only one E1 mRNA, containing three potential translational start sites, has been detected. Novel, site-specific antibodies to E1 were generated and the hypothesis that the two isoforms of E1 are translated from alternative initiation codons of a single mRNA was tested. Antibodies raised against E1A-N peptide (Met(1)to Cys(23)of E1A) reacted only with E1A by immunoblot and immunoprecipitation. Antibodies raised against E1B-N peptide (Met(1)to Glu(25)of E1B or Met(41)to Glu(65)of E1A) and E1AB-C peptide (His(1030)to Arg(1058)of E1A or His(990)to Arg(1018)of E1B) reacted with both E1A and E1B. These results indicate that (1) E1A and E1B contain the same C-terminal residues; (2) E1A contains the N terminal sequence of E1B; and (3) E1B does not contain the N terminal sequence of E1A. The two isoforms of lens E1 are therefore translated from a single mRNA. Specifically, E1A is translated from the first initiation codon, and E1B translated from the second initiation codon. E1A and E1B were affinity-purified, and their ability to 'charge' ubiquitin carrier proteins (E2s) with activated ubiquitin was compared in a cell-free system. E1A and E1B were indistinguishable with respect to charging different E2s. However, E1 immunolocalization studies with human lens epithelial cells indicate that E1A and E1B are preferentially localized to the nucleus and cytosol, respectively. This observation suggests that E1A and E1B ubiquitinate different proteins and serve different functions in intact cells.


Subject(s)
Epithelial Cells/enzymology , Lens, Crystalline/cytology , Ligases/physiology , Animals , Cattle , Cell-Free System , Codon, Initiator/physiology , Humans , Immunoblotting , Isoenzymes , Lens, Crystalline/enzymology , Mice , Precipitin Tests , Protein Biosynthesis , RNA, Messenger , Rabbits , Ubiquitin-Activating Enzymes , Ubiquitin-Protein Ligases
15.
J Invest Dermatol ; 115(4): 668-73, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998141

ABSTRACT

The melanoma cell adhesion molecule is a membrane glycoprotein whose expression is associated with tumor progression and the development of metastatic potential. The mechanisms for upregulation of the melanoma cell adhesion molecule during melanoma progression are still poorly understood. In this study, we show further evidence that melanoma cell adhesion molecule expression is tightly regulated at the transcriptional level. Using a combination of chloramphenicol acetyl transferase reporter assays and DNA mobility shift experiments, we investigated the role played by three putative melanoma cell adhesion molecule regulatory elements, namely the initiator sequence, the SCA element, and the ASp element. The SCA and the ASp boxes can potentially interact with the transcription factors Sp1 and AP-2. Sp1 binding to both sites was confirmed, but only the SCA sequence could form a complex with AP-2. AP-2-driven downregulation of the melanoma cell adhesion molecule promoter, however, did not depend only on a functional SCA element. The pyrimidine-rich CTCACTTG initiator, which overlaps the RNA start site, was essential for promoter function and was shown to interact with proteins related to basic helix-loop-helix transcription factors. Binding in nonmetastatic melanoma cells was induced by cAMP. In metastatic cells, however, binding was constitutive, but could be markedly decreased upon treatment with phorbol esters. As melanoma cell adhesion molecule expression is modulated by cAMP and phorbol ester signaling, these results suggest that the initiator is the central element that mediates cAMP and phorbol ester sensitivity and initiates melanoma cell adhesion molecule overexpression in melanomas.


Subject(s)
Antigens, CD , Antigens, Surface/genetics , Codon, Initiator/physiology , Membrane Glycoproteins , Neural Cell Adhesion Molecules , Animals , CD146 Antigen , DNA-Binding Proteins/physiology , Down-Regulation , Gene Expression Regulation , Mice , Promoter Regions, Genetic/physiology , Skin Neoplasms/chemistry , Skin Neoplasms/genetics , Transcription Factors/physiology , Transcription, Genetic/drug effects
16.
Mol Gen Genet ; 261(1): 11-20, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10071205

ABSTRACT

A scanning ribosome will usually initiate translation as soon as it encounters the first favourable AUG codon and only a few eukaryotic transcripts have more complex arrangements. These relatively few complex transcripts are normally characterized by structural features such as multiple AUGs and significant secondary structure. However, the functional relevance of these features has rarely been established. We present here a study of the functional significance of the multiple AUGs in the leader of STA2 transcripts of the budding yeast Saccharomyces cerevisiae, and extrapolate, where applicable, these results to a co-regulated gene, MUC1. The STA2 gene (a representative member of the polymorphic STA1-3 gene family), encodes an extracellular glucoamylase, and is evolutionarily linked to, and transcriptionally co-regulated with, the MUC1 gene, which encodes a mucin-like protein essential for pseudohyphal/invasive growth and cell-adhesion in S. cerevisiae. Each of these genes contains a putative upstream ORF, while STA2 has two additional in-frame AUG codons 5' to the major cistron. We show that utilization of the alternative translational start-sites of STA2 results in glucoamylases that differ at their N-termini, which are associated with differences in their localization patterns. Analysis of mutants revealed the presence of a putative secretion-enhancing signal that might prove to be relevant to the alternative targeting mechanism recently uncovered in S. cerevisiae. We show that a short up-stream ORF present in the leaders of STA1-3 and MUC1 is probably bypassed by a process of leaky scanning.


Subject(s)
Codon, Initiator/genetics , Genes, Fungal/genetics , Glucan 1,4-alpha-Glucosidase/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Codon, Initiator/physiology , Fungal Proteins/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Molecular Sequence Data , Mucins/genetics , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/enzymology , Transcription, Genetic , beta-Galactosidase/genetics
18.
Nature ; 394(6696): 854-9, 1998 Aug 27.
Article in English | MEDLINE | ID: mdl-9732867

ABSTRACT

The scanning model of translation initiation is a coherent description of how eukaryotic ribosomes reach the initiation codon after being recruited to the capped 5' end of messenger RNA. Five eukaryotic initiation factors (eIF 2, 3, 4A, 4B and 4F) with established functions have been assumed to be sufficient to mediate this process. Here we report that eIF1 and eIF1A are also both essential for translation initiation. In their absence, 43S ribosomal preinitiation complexes incubated with ATP, eIF4A, eIF4B and eIF4F bind exclusively to the cap-proximal region but are unable to reach the initiation codon. Individually, eIF1A enhances formation of this cap-proximal complex, and eIF1 weakly promotes formation of a 48S ribosomal complex at the initiation codon. These proteins act synergistically to mediate assembly of ribosomal initiation complexes at the initiation codon and dissociate aberrant complexes from the mRNA.


Subject(s)
Codon, Initiator/physiology , Eukaryotic Initiation Factor-1/physiology , Peptide Initiation Factors/physiology , Protein Biosynthesis/physiology , Ribosomes/physiology , Animals , Macromolecular Substances , RNA Caps , RNA, Messenger/metabolism , Rabbits
19.
Dev Genes Evol ; 207(7): 482-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9510543

ABSTRACT

Promoters active in the germline produce OVO-A and OVO-B mRNAs encoding isoforms of a putative transcription factor. The isoforms have a common C2H2 zinc-finger domain but different N-termini that include potential effector domains. Single point mutations in three dominant-negative ovoD mutations result in new in-frame initiation codons in OVO-B mRNAs and amino acid substitutions within charged regions of OVO-A proteins. Three lines of evidence suggest that the dominant activity is due to the new initiation codons in OVO-B mRNAs and not the amino acid substitutions in OVO-A. First, we made a fourth ovoD allele by inserting a new in-frame AUG. This ovoD4 allele encodes a nearly full-length OVO-A isoform from OVO-B mRNAs. Second, engineered stop codons in ovoD1 downstream of the new AUG abolished dominant negative activity. Third, a substantial deletion of an OVO-A region encoding a highly charged amino acid domain fully rescued loss-of-function ovo alleles. These data suggest that ovoD mutations result in inappropriate expression of OVO-A in the female germline.


Subject(s)
Alleles , Codon, Initiator/physiology , DNA-Binding Proteins/genetics , Drosophila Proteins , Drosophila/genetics , Genes, Dominant/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Female , Fertility/genetics , Genes, Insect/genetics , Molecular Sequence Data , Ovary/growth & development , Phenotype , Point Mutation/genetics , RNA, Messenger/genetics , Regulatory Sequences, Nucleic Acid/genetics , Sex Determination Processes , Zinc Fingers
20.
Int J Biochem Cell Biol ; 27(5): 469-73, 1995 May.
Article in English | MEDLINE | ID: mdl-7641076

ABSTRACT

The AGG and AGA are the least used arginine codons in E. coli but they are the most preferable ones in eukaryotes. The low expression of some eucaryotic genes (such as human alpha-1 interferon gene) which contain clusters of AGG codons is explained either by the limited pool of the tRNA(AGG) (Varenne and Lazdunski, 1986) or by the competition of these clusters with the Shine-Dalgarno (SD) sequence (Ivanov et al., 1992). The aim of the present study is to demonstrate the in vivo capacity of AGG tandems to bind to bacterial ribosomes. The two tandems of AGG codons (Arg12 Arg13 and Arg163 Arg164) of hIF alpha 1 with their surrounding nucleotides were cloned in a bacterial expression plasmid containing a strong promoter and a reporter gene (chloramphenicol acetyltransferase, CAT) devoid of a ribosome binding site. The results obtained showed that both AGG tandems initiated translation of the CAT mRNA with an efficiency equal to that of the consensus SD sequence and several fold higher than the native SD sequence of the CAT gene.


Subject(s)
Arginine/physiology , Codon, Initiator/physiology , Interferon Type I/genetics , Peptide Chain Initiation, Translational/physiology , Repetitive Sequences, Nucleic Acid , Amino Acid Sequence , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Escherichia coli/genetics , Genes, Reporter , Humans , Molecular Sequence Data , Recombinant Proteins , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...