Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lev L. Kisselev is active.

Publication


Featured researches published by Lev L. Kisselev.


RNA | 1999

Mutations in the highly conserved GGQ motif of class 1 polypeptide release factors abolish ability of human eRF1 to trigger peptidyl-tRNA hydrolysis

Ludmila Frolova; Ruslan Yu. Tsivkovskii; Galina F. Sivolobova; Nina Yu. Oparina; Oleg Igorevich Serpinsky; Vladimir M. Blinov; Sergey I. Tatkov; Lev L. Kisselev

Although the primary structures of class 1 polypeptide release factors (RF1 and RF2 in prokaryotes, eRF1 in eukaryotes) are known, the molecular basis by which they function in translational termination remains obscure. Because all class 1 RFs promote a stop-codon-dependent and ribosome-dependent hydrolysis of peptidyl-tRNAs, one may anticipate that this common function relies on a common structural motif(s). We have compared amino acid sequences of the available class 1 RFs and found a novel, common, unique, and strictly conserved GGQ motif that should be in a loop (coil) conformation as deduced by programs predicting protein secondary structure. Site-directed mutagenesis of the human eRF1 as a representative of class 1 RFs shows that substitution of both glycyl residues in this motif, G183 and G184, causes complete inactivation of the protein as a release factor toward all three stop codons, whereas two adjacent amino acid residues, G181 and R182, are functionally nonessential. Inactive human eRF1 mutants compete in release assays with wild-type eRF1 and strongly inhibit their release activity. Mutations of the glycyl residues in this motif do not affect another function, the ability of eRF1 together with the ribosome to induce GTPase activity of human eRF3, a class 2 RF. We assume that the novel highly conserved GGQ motif is implicated directly or indirectly in the activity of class 1 RFs in translation termination.


Structure | 2002

Polypeptide Release Factors in Prokaryotes and Eukaryotes: Same Function, Different Structure

Lev L. Kisselev

Although the eukaryotic (eRF1) and prokaryotic (RF2) polypeptide release (translation termination) factors are functionally similar, they turn out to be very different in overall shape and architecture and in the location of key functional elements.


The EMBO Journal | 2003

Termination of translation: interplay of mRNA, rRNAs and release factors?

Lev L. Kisselev; Måns Ehrenberg; Ludmila Frolova

Termination of translation in eukaryotes has focused recently on functional anatomy of polypeptide chain release factor, eRF1, by using a variety of different approaches. The tight correlation between the domain structure and different functions of eRF1 has been revealed. Independently, the role of prokaryotic RF1/2 in GTPase activity of RF3 has been deciphered, as well as RF3 function itself.


Cell | 2006

In Vitro Reconstitution of Eukaryotic Translation Reveals Cooperativity between Release Factors eRF1 and eRF3

Elena Z. Alkalaeva; Andrey V. Pisarev; Lyudmila Frolova; Lev L. Kisselev; Tatyana V. Pestova

Eukaryotic translation termination is triggered by peptide release factors eRF1 and eRF3. Whereas eRF1 recognizes all three termination codons and induces hydrolysis of peptidyl tRNA, eRF3s function remains obscure. Here, we reconstituted all steps of eukaryotic translation in vitro using purified ribosomal subunits; initiation, elongation, and termination factors; and aminoacyl tRNAs. This allowed us to investigate termination using pretermination complexes assembled on mRNA encoding a tetrapeptide and to propose a model for translation termination that accounts for the cooperative action of eRF1 and eRF3 in ensuring fast release of nascent polypeptide. In this model, binding of eRF1, eRF3, and GTP to pretermination complexes first induces a structural rearrangement that is manifested as a 2 nucleotide forward shift of the toeprint attributed to pretermination complexes that leads to GTP hydrolysis followed by rapid hydrolysis of peptidyl tRNA. Cooperativity between eRF1 and eRF3 required the eRF3 binding C-terminal domain of eRF1.


Trends in Biochemical Sciences | 2000

Translational termination comes of age

Lev L. Kisselev; Richard H. Buckingham

Translational termination has been a largely ignored aspect of protein synthesis for many years. However, the recent identification of new release-factor genes, the mapping of release-factor functional sites and in vitro reconstitution experiments have provided a deeper understanding of the termination mechanism. In addition, protein-protein interactions among release factors and with other proteins have been revealed. The three-dimensional structures of a prokaryotic ribosome recycling factor and eukaryotic release factor 1 (eRF1) mimic the shape of transfer RNA, indicating that they bind to the same ribosomal site. Post-termination events in bacteria have been clarified, linking termination, ribosomal recycling and translation initiation.


RNA | 2002

Highly conserved NIKS tetrapeptide is functionally essential in eukaryotic translation termination factor eRF1.

Ludmila Frolova; Alim Seit-Nebi; Lev L. Kisselev

Class-1 polypeptide chain release factors (RFs) play a key role in translation termination. Eukaryotic (eRF1) and archaeal class-1 RFs possess a highly conserved Asn-Ile-Lys-Ser (NIKS) tetrapeptide located at the N-terminal domain of human eRF1. In the three-dimensional structure, NIKS forms a loop between helices. The universal occurrence and exposed nature of this motif provoke the appearance of hypotheses postulating an essential role of this tetrapeptide in stop codon recognition and ribosome binding. To approach this problem experimentally, site-directed mutagenesis of the NIKS (positions 61-64) in human eRF1 and adjacent amino acids has been applied followed by determination of release activity and ribosome-binding capacity of mutants. Substitutions of Asn61 and Ile62 residues of the NIKS cause a decrease in the ability of eRF1 mutants to promote termination reaction in vitro, but to a different extent depending on the stop codon specificity, position, and nature of the substituting residues. This observation points to a possibility that Asn-Ile dipeptide modulates the specific recognition of the stop codons by eRF1. Some replacements at positions 60, 63, and 64 cause a negligible (if any) effect in contrast to what has been deduced from some current hypotheses predicting the structure of the termination codon recognition site in eRF1. Reduction in ribosome binding revealed for Ile62, Ser64, Arg65, and Arg68 mutants argues in favor of the essential role played by the right part of the NIKS loop in interaction with the ribosome, most probably with ribosomal RNA.


Molecular Microbiology | 2002

The essential role of the invariant GGQ motif in the function and stability in vivo of bacterial release factors RF1 and RF2.

Liliana Mora; Valérie Heurgué-Hamard; Stéphanie Champ; Måns Ehrenberg; Lev L. Kisselev; Richard H. Buckingham

Release factors RF1 and RF2 are required in bacteria for the cleavage of peptidyl‐tRNA. A single sequence motif, GGQ, is conserved in all eubacterial, archaebacterial and eukaryotic release factors and may mimic the CCA end of tRNA, although the position of the motif in the crystal structures of human eRF1 and Escherichia coli RF2 is strikingly different. Mutations have been introduced at each of the three conserved positions. Changing the Gln residue to Ala or Glu allowed the factors to retain about 22% of tetrapeptide release activity in vitro, but these mutants could not complement thermosensitive RF mutants in vivo. None of several mutants with altered Gly residues retained activity in vivo or in vitro. Many GGQ mutants were poorly expressed and are presumably unstable; many were also toxic to the cell. The toxic mutant factors or their degradation products may bind to ribosomes inhibiting the action of the normal factor. These data are consistent with a common role for the GGQ motif in bacterial and eukaryotic release factors, despite strong divergence in primary, secondary and tertiary structure, but are difficult to reconcile with the hypothesis that the amide nitrogen of the Gln plays a vital role in peptidyl‐tRNA hydrolysis.


Oncogene | 2004

Discovery of frequent homozygous deletions in chromosome 3p21.3 LUCA and AP20 regions in renal, lung and breast carcinomas.

Vera N. Senchenko; Jian Liu; Witaly Loginov; Igor Bazov; Debora Angeloni; Yury Seryogin; V. D. Ermilova; T. P. Kazubskaya; R. F. Garkavtseva; Veronika Zabarovska; Lev L. Kisselev; John D. Minna; Michael I. Lerman; George Klein; E. A. Braga; Eugene R. Zabarovsky

We searched for chromosome 3p homo- and hemizygous losses in 23 lung cancer cell lines, 53 renal cell and 22 breast carcinoma biopsies using 31 microsatellite markers located in frequently deleted 3p regions. In addition, two sequence-tagged site markers (NLJ-003 and NL3-001) located in the Alu-PCR clone 20 region (AP20) and lung cancer (LUCA) regions, respectively, were used for quantitative real-time PCR (QPCR). We found frequent (10–18%) homozygous deletions (HDs) in both 3p21.3 regions in the biopsies and lung cancer cell lines. In addition, we discovered that amplification of 3p is a very common (15–42.5%) event in these cancers and probably in other epithelial malignancies. QPCR showed that aberrations of either NLJ-003 or NL3-001 were detected in more than 90% of all studied cases. HDs were frequently detected simultaneously both in NLJ-003 or NL3-001 loci in the same tumour (P<3–10−7). This observation suggests that tumour suppressor genes (TSG) in these regions could have a synergistic effect. The exceptionally high frequency of chromosome aberrations in NLJ-003 and NL3-001 loci suggests that multiple TSG(s) involved in different malignancies are located very near to these markers. Precise mapping of 15 independent HDs in the LUCA region allowed us to establish the smallest HD region in 3p21.3C located between D3S1568 (CACNA2D2 gene) and D3S4604 (SEMA3F gene). This region contains 17 genes. Mapping of 19 HDs in the AP20 region resulted in the localization of the minimal region to the interval flanked by D3S1298 and D3S3623 markers. Only four genes were discovered in this interval, namely, APRG1, ITGA9, HYA22 and VILL.


FEBS Letters | 1998

Eukaryotic mRNAs encoding abundant and scarce proteins are statistically dissimilar in many structural features.

Alex V. Kochetov; Igor V Ischenko; Denis G. Vorobiev; Alexander E. Kel; V. N. Babenko; Lev L. Kisselev; N. A. Kolchanov

It is well known that non‐coding mRNA sequences are dissimilar in many structural features. For individual mRNAs correlations were found for some of these features and their translational efficiency. However, no systematic statistical analysis was undertaken to relate protein abundance and structural characteristics of mRNA encoding the given protein. We have demonstrated that structural and contextual features of eukaryotic mRNAs encoding high‐ and low‐abundant proteins differ in the 5′ untranslated regions (UTR). Statistically, 5′ UTRs of low‐expression mRNAs are longer, their guanine plus cytosine content is higher, they have a less optimal context of the translation initiation codons of the main open reading frames and contain more frequently upstream AUG than 5′ UTRs of high‐expression mRNAs. Apart from the differences in 5′ UTRs, high‐expression mRNAs contain stronger termination signals. Structural features of low‐ and high‐expression mRNAs are likely to contribute to the yield of their protein products.


EMBO Reports | 2002

Conversion of omnipotent translation termination factor eRF1 into ciliate-like UGA-only unipotent eRF1

Alim Seit-Nebi; Ludmila Frolova; Lev L. Kisselev

In eukaryotic ribosomes, termination of translation is triggered by class 1 polypeptide release factor, eRF1. In organisms with a universal code, eRF1 responds to three stop codons, whereas, in ciliates with variant codes, only one or two codon(s) remain(s) as stop signals. By mutagenesis of the Y–C–F minidomain of the N domain, we converted an omnipotent human eRF1 recognizing all three stop codons into a unipotent ‘ciliate‐like’ UGA‐only eRF1. The conserved Cys127 located in the Y–C–F minidomain plays a critical role in stop codon recognition. The UGA‐only response has also been achieved by concomitant substitutions of four other amino acids located at the Y–C–F and NIKS minidomains of eRF1. We suggest that for eRF1 the stop codon decoding is of a non‐linear (non‐protein‐anticodon) type and explores a combination of positive and negative determinants. We assume that stop codon recognition is profoundly different by eukaryotic and prokaryotic class 1 RFs.

Collaboration


Dive into the Lev L. Kisselev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ludmila Frolova

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Tamara D. Mashkova

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ol'ga O. Favorova

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Sergey F. Beresten

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Galina K. Kovaleva

Engelhardt Institute of Molecular Biology

View shared research outputs
Researchain Logo
Decentralizing Knowledge