Network


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

Hotspot


Dive into the research topics where Jan Paleček is active.

Publication


Featured researches published by Jan Paleček.


Molecular and Cellular Biology | 2005

Nse2, a component of the Smc5-6 complex, is a SUMO ligase required for the response to DNA damage.

Emily A. Andrews; Jan Paleček; John Sergeant; Elaine M. Taylor; Alan R. Lehmann; Felicity Z. Watts

ABSTRACT The Schizosaccharomyces pombe SMC proteins Rad18 (Smc6) and Spr18 (Smc5) exist in a high-M r complex which also contains the non-SMC proteins Nse1, Nse2, Nse3, and Rad62. The Smc5-6 complex, which is essential for viability, is required for several aspects of DNA metabolism, including recombinational repair and maintenance of the DNA damage checkpoint. We have characterized Nse2 and show here that it is a SUMO ligase. Smc6 (Rad18) and Nse3, but not Smc5 (Spr18) or Nse1, are sumoylated in vitro in an Nse2-dependent manner, and Nse2 is itself autosumoylated, predominantly on the C-terminal part of the protein. Mutations of C195 and H197 in the Nse2 RING-finger-like motif abolish Nse2-dependent sumoylation. nse2.SA mutant cells, in which nse2.C195S-H197A is integrated as the sole copy of nse2, are viable, whereas the deletion of nse2 is lethal. Smc6 (Rad18) is sumoylated in vivo: the sumoylation level is increased upon exposure to DNA damage and is drastically reduced in the nse2.SA strain. Since nse2.SA cells are sensitive to DNA-damaging agents and to exposure to hydroxyurea, this implicates the Nse2-dependent sumoylation activity in DNA damage responses but not in the essential function of the Smc5-6 complex.


Chemical Reviews | 2015

Electrochemistry of Nonconjugated Proteins and Glycoproteins. Toward Sensors for Biomedicine and Glycomics

Emil Paleček; Jan Tkac; Martin Bartošík; Tomas Bertok; Veronika Ostatná; Jan Paleček

In this review, we wish to show that in the recent years a significant progress was done in the EC analysis of practically all proteins, based on electroactivity of amino acid (aa) residues in proteins. Also electrochemistry of polysaccharides, oligosaccharides and glycoproteins greatly advanced in creating important steps for its larger application in the glycoprotein research. In recent decades, a great effort was devoted to the discovery and application of biomarkers for analysis of different diseases, including cancer. In the following paragraphs, special attention will be paid (i) to intrinsic electroactivity of peptides and proteins, including the sensitivity to changes in protein 3D structures, as well as to recent advances in EC investigations of DNA-protein interactions, (ii) to intrinsic electroactivity of glycans and polysaccharides, advances in EC detection of lectin-glycoprotein interactions and to introduction of electroactive labels to polysaccharides and glycans and finally (iii) to EC detection of protein biomarkers, based predominantly on application of antibodies in immunoassays, nucleic acid and peptide aptamers for construction of aptasensors, and lectin biosensors for detection of glycoprotein biomarkers.


Molecular and Cellular Biology | 2005

Composition and Architecture of the Schizosaccharomyces pombe Rad18 (Smc5-6) Complex

John Sergeant; Elaine M. Taylor; Jan Paleček; Maria Fousteri; Emily A. Andrews; Sara Sweeney; Hideo Shinagawa; Felicity Z. Watts; Alan R. Lehmann

ABSTRACT The rad18 gene of Schizosaccharomyces pombe is an essential gene that is involved in several different DNA repair processes. Rad18 (Smc6) is a member of the structural maintenance of chromosomes (SMC) family and, together with its SMC partner Spr18 (Smc5), forms the core of a high-molecular-weight complex. We show here that both S. pombe and human Smc5 and -6 interact through their hinge domains and that four independent temperature-sensitive mutants of Rad18 (Smc6) are all mutated at the same glycine residue in the hinge region. This mutation abolishes the interactions between the hinge regions of Rad18 (Smc6) and Spr18 (Smc5), as does mutation of a conserved glycine in the hinge region of Spr18 (Smc5). We purified the Smc5-6 complex from S. pombe and identified four non-SMC components, Nse1, Nse2, Nse3, and Rad62. Nse3 is a novel protein which is related to the mammalian MAGE protein family, many members of which are specifically expressed in cancer tissue. In initial steps to understand the architecture of the complex, we identified two subcomplexes containing Rad18-Spr18-Nse2 and Nse1-Nse3-Rad62. The subcomplexes are probably bridged by a weaker interaction between Nse2 and Nse3.


Journal of Biological Chemistry | 2006

The Smc5-Smc6 DNA Repair Complex BRIDGING OF THE Smc5-Smc6 HEADS BY THE KLEISIN, Nse4, AND NON-KLEISIN SUBUNITS

Jan Paleček; Susanne Vidot; Min Feng; Aidan J. Doherty; Alan R. Lehmann

Structural maintenance of chromosomes (SMC) proteins play fundamental roles in many aspects of chromosome organization and dynamics. The SMC complexes form unique structures with long coiled-coil arms folded at a hinge domain, so that the globular N- and C-terminal domains are brought together to form a “head.” Within the Smc5-Smc6 complex, we previously identified two subcomplexes containing Smc6-Smc5-Nse2 and Nse1-Nse3-Nse4. A third subcomplex containing Nse5 and -6 has also been identified recently. We present evidence that Nse4 is the kleisin component of the complex, which bridges the heads of Smc5 and -6. The C-terminal part of Nse4 interacts with the head domain of Smc5, and structural predictions for Nse4 proteins suggest similar motifs that are shared within the kleisin family. Specific mutations within a predicted winged helix motif of Nse4 destroy the interaction with Smc5. We propose that Nse4 and its orthologs form the δ-kleisin subfamily. We further show that Nse3, as well as Nse5 and Nse6, also bridge the heads of Smc5 and -6. The Nse1-Nse3-Nse4 and Nse5-Nse6 subcomplexes bind to the Smc5-Smc6 heads domain at different sites.


FEBS Letters | 2008

Mapping of interaction domains of putative telomere-binding proteins AtTRB1 and AtPOT1b from Arabidopsis thaliana

Petra Procházková Schrumpfová; Milan Kuchař; Jan Paleček; Jiří Fajkus

MINT‐6440051: AtTRB1 (uniprotkb:Q8VWK4) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by two‐hybrid (MI:0018) MINT‐6440068: AtTRB2 (uniprotkb:Q8VX38) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by two‐hybrid (MI:0018) MINT‐6440083: AtTRB3 (uniprotkb:Q9M2X3) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by two‐hybrid (MI:0018) MINT‐6440099: AtPOT1b (uniprotkb:Q6Q835) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by two‐hybrid (MI:0018) MINT‐6440119: AtPOT1b (uniprotkb:Q6Q835) physically interacts (MI:0218) with AtTRB2 (uniprotkb:Q8VX38) by two‐hybrid (MI:0018) MINT‐6440138: AtPOT1b (uniprotkb:Q6Q835) physically interacts (MI:0218) with AtTRB3 (uniprotkb:Q9M2X3) by two‐hybrid (MI:0018) MINT‐6440216: AtPOT1b (uniprotkb:Q6Q835) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by coimmunoprecipitation (MI:0019) MINT‐6440157: AtTRB2 (uniprotkb:Q8VX38) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by coimmunoprecipitation (MI:0019) MINT‐6440177: AtTRB3 (uniprotkb:Q9M2X3) physically interacts (MI:0218) with AtTRB1 (uniprotkb:Q8VWK4) by coimmunoprecipitation (MI:0019)


PLOS ONE | 2011

Interactions between the Nse3 and Nse4 Components of the SMC5-6 Complex Identify Evolutionarily Conserved Interactions between MAGE and EID Families

Jessica Hudson; Katerina Bednarova; Lucie Kozáková; Chunyan Liao; Marc Guérineau; Rita Colnaghi; Susanne Vidot; Jaromír Marek; Sreenivas Reddy Bathula; Alan R. Lehmann; Jan Paleček

Background The SMC5-6 protein complex is involved in the cellular response to DNA damage. It is composed of 6–8 polypeptides, of which Nse1, Nse3 and Nse4 form a tight sub-complex. MAGEG1, the mammalian ortholog of Nse3, is the founding member of the MAGE (melanoma-associated antigen) protein family and Nse4 is related to the EID (E1A-like inhibitor of differentiation) family of transcriptional repressors. Methodology/Principal Findings Using site-directed mutagenesis, protein-protein interaction analyses and molecular modelling, we have identified a conserved hydrophobic surface on the C-terminal domain of Nse3 that interacts with Nse4 and identified residues in its N-terminal domain that are essential for interaction with Nse1. We show that these interactions are conserved in the human orthologs. Furthermore, interaction of MAGEG1, the mammalian ortholog of Nse3, with NSE4b, one of the mammalian orthologs of Nse4, results in transcriptional co-activation of the nuclear receptor, steroidogenic factor 1 (SF1). In an examination of the evolutionary conservation of the Nse3-Nse4 interactions, we find that several MAGE proteins can interact with at least one of the NSE4/EID proteins. Conclusions/Significance We have found that, despite the evolutionary diversification of the MAGE family, the characteristic hydrophobic surface shared by all MAGE proteins from yeast to humans mediates its binding to NSE4/EID proteins. Our work provides new insights into the interactions, evolution and functions of the enigmatic MAGE proteins.


Cell Cycle | 2015

The melanoma-associated antigen 1 (MAGEA1) protein stimulates the E3 ubiquitin-ligase activity of TRIM31 within a TRIM31-MAGEA1-NSE4 complex

Lucie Kozáková; Lucie Vondrová; Karel Stejskal; Panagoula Charalabous; Peter Kolesár; Alan R. Lehmann; Stjepan Uldrijan; Christopher M. Sanderson; Zbynek Zdrahal; Jan Paleček

The MAGE (Melanoma-associated antigen) protein family members are structurally related to each other by a MAGE-homology domain comprised of 2 winged helix motifs WH/A and WH/B. This family specifically evolved in placental mammals although single homologs designated NSE3 (non-SMC element) exist in most eukaryotes. NSE3, together with its partner proteins NSE1 and NSE4 form a tight subcomplex of the structural maintenance of chromosomes SMC5–6 complex. Previously, we showed that interactions of the WH/B motif of the MAGE proteins with their NSE4/EID partners are evolutionarily conserved (including the MAGEA1-NSE4 interaction). In contrast, the interaction of the WH/A motif of NSE3 with NSE1 diverged in the MAGE paralogs. We hypothesized that the MAGE paralogs acquired new RING-finger-containing partners through their evolution and form MAGE complexes reminiscent of NSE1-NSE3-NSE4 trimers. In this work, we employed the yeast 2-hybrid system to screen a human RING-finger protein library against several MAGE baits. We identified a number of potential MAGE-RING interactions and confirmed several of them (MDM4, PCGF6, RNF166, TRAF6, TRIM8, TRIM31, TRIM41) in co-immunoprecipitation experiments. Among these MAGE-RING pairs, we chose to examine MAGEA1-TRIM31 in detail and showed that both WH/A and WH/B motifs of MAGEA1 bind to the coiled-coil domain of TRIM31 and that MAGEA1 interaction stimulates TRIM31 ubiquitin-ligase activity. In addition, TRIM31 directly binds to NSE4, suggesting the existence of a TRIM31-MAGEA1-NSE4 complex reminiscent of the NSE1-NSE3-NSE4 trimer. These results suggest that MAGEA1 functions as a co-factor of TRIM31 ubiquitin-ligase and that the TRIM31-MAGEA1-NSE4 complex may have evolved from an ancestral NSE1-NSE3-NSE4 complex.


PLOS ONE | 2012

Analysis of the Nse3/MAGE-Binding Domain of the Nse4/EID Family Proteins

Marc Guérineau; Kríz Z; Lucie Kozáková; Katerina Bednarova; Pavel Janoš; Jan Paleček

Background The Nse1, Nse3 and Nse4 proteins form a tight sub-complex of the large SMC5-6 protein complex. hNSE3/MAGEG1, the mammalian ortholog of Nse3, is the founding member of the MAGE (melanoma-associated antigen) protein family and the Nse4 kleisin subunit is related to the EID (E1A-like inhibitor of differentiation) family of proteins. We have recently shown that human MAGE proteins can interact with NSE4/EID proteins through their characteristic conserved hydrophobic pocket. Methodology/Principal Findings Using mutagenesis and protein-protein interaction analyses, we have identified a new Nse3/MAGE-binding domain (NMBD) of the Nse4/EID proteins. This short domain is located next to the Nse4 N-terminal kleisin motif and is conserved in all NSE4/EID proteins. The central amino acid residues of the human NSE4b/EID3 domain were essential for its binding to hNSE3/MAGEG1 in yeast two-hybrid assays suggesting they form the core of the binding domain. PEPSCAN ELISA measurements of the MAGEC2 binding affinity to EID2 mutant peptides showed that similar core residues contribute to the EID2-MAGEC2 interaction. In addition, the N-terminal extension of the EID2 binding domain took part in the EID2-MAGEC2 interaction. Finally, docking and molecular dynamic simulations enabled us to generate a structure model for EID2-MAGEC2. Combination of our experimental data and the structure modeling showed how the core helical region of the NSE4/EID domain binds into the conserved pocket characteristic of the MAGE protein family. Conclusions/Significance We have identified a new Nse4/EID conserved domain and characterized its binding to Nse3/MAGE proteins. The conservation and binding of the interacting surfaces suggest tight co-evolution of both Nse4/EID and Nse3/MAGE protein families.


Nucleic Acids Research | 2016

Chromatin association of the SMC5/6 complex is dependent on binding of its NSE3 subunit to DNA

Katerina Zabrady; Marek Adamus; Lucie Vondrová; Chunyan Liao; Hana Skoupilová; Markéta Nováková; Lenka Jurčišinová; Aaron Alt; Antony W. Oliver; Alan R. Lehmann; Jan Paleček

SMC5/6 is a highly conserved protein complex related to cohesin and condensin, which are the key components of higher-order chromatin structures. The SMC5/6 complex is essential for proliferation in yeast and is involved in replication fork stability and processing. However, the precise mechanism of action of SMC5/6 is not known. Here we present evidence that the NSE1/NSE3/NSE4 sub-complex of SMC5/6 binds to double-stranded DNA without any preference for DNA-replication/recombination intermediates. Mutations of key basic residues within the NSE1/NSE3/NSE4 DNA-binding surface reduce binding to DNA in vitro. Their introduction into the Schizosaccharomyces pombe genome results in cell death or hypersensitivity to DNA damaging agents. Chromatin immunoprecipitation analysis of the hypomorphic nse3 DNA-binding mutant shows a reduced association of fission yeast SMC5/6 with chromatin. Based on our results, we propose a model for loading of the SMC5/6 complex onto the chromatin.


Journal of Clinical Investigation | 2016

Destabilized SMC5/6 complex leads to chromosome breakage syndrome with severe lung disease

Saskia N. van der Crabben; Marije P Hennus; Grant A. McGregor; Deborah I. Ritter; Sandesh C.S. Nagamani; Owen S. Wells; Magdalena Harakalova; Ivan K. Chinn; Aaron Alt; Lucie Vondrová; Ron Hochstenbach; Joris M. van Montfrans; Suzanne W. J. Terheggen-Lagro; Stef van Lieshout; Markus J. van Roosmalen; Ivo Renkens; Karen Duran; Isaac J. Nijman; Wigard P. Kloosterman; Eric A.M. Hennekam; Jordan S. Orange; Peter M. van Hasselt; David A. Wheeler; Jan Paleček; Alan R. Lehmann; Antony W. Oliver; Laurence H. Pearl; Sharon E. Plon; Johanne M. Murray; Gijs van Haaften

The structural maintenance of chromosomes (SMC) family of proteins supports mitotic proliferation, meiosis, and DNA repair to control genomic stability. Impairments in chromosome maintenance are linked to rare chromosome breakage disorders. Here, we have identified a chromosome breakage syndrome associated with severe lung disease in early childhood. Four children from two unrelated kindreds died of severe pulmonary disease during infancy following viral pneumonia with evidence of combined T and B cell immunodeficiency. Whole exome sequencing revealed biallelic missense mutations in the NSMCE3 (also known as NDNL2) gene, which encodes a subunit of the SMC5/6 complex that is essential for DNA damage response and chromosome segregation. The NSMCE3 mutations disrupted interactions within the SMC5/6 complex, leading to destabilization of the complex. Patient cells showed chromosome rearrangements, micronuclei, sensitivity to replication stress and DNA damage, and defective homologous recombination. This work associates missense mutations in NSMCE3 with an autosomal recessive chromosome breakage syndrome that leads to defective T and B cell function and acute respiratory distress syndrome in early childhood.

Collaboration


Dive into the Jan Paleček's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucie Vondrová

Central European Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lucie Kozáková

Central European Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emil Paleček

Academy of Sciences of the Czech Republic

View shared research outputs
Researchain Logo
Decentralizing Knowledge