TY - JOUR AU - Cinege, Gyöngyi Ilona AU - Fodor, K. AU - Magyar, Lilla Brigitta AU - Lipinszki, Zoltán AU - Hultmark, D. AU - Andó, István TI - Cellular Immunity of Drosophila willistoni Reveals Novel Complexity in Insect Anti-Parasitoid Defense JF - CELLS J2 - CELLS-BASEL VL - 13 PY - 2024 IS - 7 SN - 2073-4409 DO - 10.3390/cells13070593 UR - https://m2.mtmt.hu/api/publication/34804520 ID - 34804520 N1 - Innate Immunity Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, 6726, Hungary Doctoral School of Biology, University of Szeged, Szeged, 6720, Hungary MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, Szeged, 6726, Hungary National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, 6726, Hungary Department of Molecular Biology, Umea University, Umea, 901 87, Sweden Export Date: 22 April 2024 Correspondence Address: Cinege, G.; Innate Immunity Group, Hungary; email: cinege.gyongyi@brc.hu LA - English DB - MTMT ER - TY - JOUR AU - Kovács, Zoltán AU - Bajusz, Csaba AU - Szabó, Anikó AU - Borkúti, Péter AU - Vedelek, Balázs AU - Benke, Reka AU - Lipinszki, Zoltán AU - Kristó, Ildikó AU - Vilmos, Péter TI - A bipartite NLS motif mediates the nuclear import of Drosophila moesin JF - FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY J2 - FRONT CELL DEV BIOL VL - 12 PY - 2024 PG - 14 SN - 2296-634X DO - 10.3389/fcell.2024.1206067 UR - https://m2.mtmt.hu/api/publication/34743202 ID - 34743202 N1 - Funding Agency and Grant Number: NKFIH (Hungarian National Research, Development and Innovation Office) through the National Laboratory for Biotechnology program [PD127968, LP2017-7/2017]; Hungarian Academy of Sciences Lendulet Grant; [2022-2.1.1-NL-2022-00008] Funding text: This work was supported by NKFIH (Hungarian National Research, Development and Innovation Office) through the National Laboratory for Biotechnology program, grant 2022-2.1.1-NL-2022-00008 (PV), and PD127968 (IK), and the Hungarian Academy of Sciences Lendulet Grant LP2017-7/2017 (ZL). AB - The ERM protein family, which consists of three closely related proteins in vertebrates, ezrin, radixin, and moesin (ERM), is an ancient and important group of cytoplasmic actin-binding and organizing proteins. With their FERM domain, ERMs bind various transmembrane proteins and anchor them to the actin cortex through their C-terminal F-actin binding domain, thus they are major regulators of actin dynamics in the cell. ERMs participate in many fundamental cellular processes, such as phagocytosis, microvilli formation, T-cell activation and tumor metastasis. We have previously shown that, besides its cytoplasmic activities, the single ERM protein of Drosophila melanogaster, moesin, is also present in the cell nucleus, where it participates in gene expression and mRNA export. Here we study the mechanism by which moesin enters the nucleus. We show that the nuclear import of moesin is an NLS-mediated, active process. The nuclear localization sequence of the moesin protein is an evolutionarily highly conserved, conventional bipartite motif located on the surface of the FERM domain. Our experiments also reveal that the nuclear import of moesin does not require PIP2 binding or protein activation, and occurs in monomeric form. We propose, that the balance between the phosphorylated and non-phosphorylated protein pools determines the degree of nuclear import of moesin. LA - English DB - MTMT ER - TY - CHAP AU - Sávai, Gergő AU - Kartali, Tünde AU - Benci, Dániel Attila AU - Patai, Roland AU - Lipinszki, Zoltán AU - Vágvölgyi, Csaba AU - Papp, Tamás TI - Mikovírusok azonosítása Rhizopus fajokban T2 - Biotechnológiai Szakmai Nap Absztraktfüzet PB - Doktoranduszok Országos Szövetsége (DOSZ) CY - Budapest SN - 9786156457448 PY - 2024 UR - https://m2.mtmt.hu/api/publication/34723493 ID - 34723493 LA - Hungarian DB - MTMT ER - TY - JOUR AU - Ábrahám, Edit AU - Bajusz, Csaba AU - Marton, Annamária AU - Borics, Attila AU - Mdluli, Thandiswa AU - Pardi, Norbert AU - Lipinszki, Zoltán TI - Expression and purification of the receptor-binding domain of SARS-CoV-2 spike protein in mammalian cells for immunological assays JF - FEBS OPEN BIO J2 - FEBS OPEN BIO VL - 14 PY - 2024 IS - 3 SP - 380 EP - 389 PG - 10 SN - 2211-5463 DO - 10.1002/2211-5463.13754 UR - https://m2.mtmt.hu/api/publication/34575640 ID - 34575640 N1 - Funding Agency and Grant Number: National Laboratory for Biotechnology [2022-2.1.1-NL-2022-00008]; Hungarian Academy of Sciences (Lenduelet Program Grant) [LP2017-7/2017]; National Research, Development and Innovation Office [K143124]; National Institute of Allergy and Infectious Diseases [R01AI146101, R01AI153064] Funding text: This work was supported by the National Laboratory for Biotechnology (2022-2.1.1-NL-2022-00008) to CB and ZL, the Hungarian Academy of Sciences (Lenduelet Program Grant (LP2017-7/2017)) to ZL, and the National Research, Development and Innovation Office (K143124) to AB. NP was supported by the National Institute of Allergy and Infectious Diseases (R01AI146101 and R01AI153064). The authors are grateful for Adam Pap and Zsuzsanna Darula (BRC) for the PNGase F enzyme, and Petar Lambrev (BRC) for the Jasco J-815 CD spectropolarimeter. AB - The receptor-binding domain (RBD) of the spike glycoprotein of SARS-CoV-2 virus mediates the interaction with the host cell and is required for virus internalization. It is, therefore, the primary target of neutralizing antibodies. The receptor-binding domain soon became the major target for COVID-19 research and the development of diagnostic tools and new-generation vaccines. Here, we provide a detailed protocol for high-yield expression and one-step affinity purification of recombinant RBD from transiently transfected Expi293F cells. Expi293F mammalian cells can be grown to extremely high densities in a specially formulated serum-free medium in suspension cultures, which makes them an excellent tool for secreted protein production. The highly purified RBD is glycosylated, structurally intact, and forms homomeric complexes. With this quick and easy method, we are able to produce large quantities of RBD (80 mg center dot L-1 culture) that we have successfully used in immunological assays to examine antibody titers and seroconversion after mRNA-based vaccination of mice. LA - English DB - MTMT ER - TY - JOUR AU - Réthi-Nagy, Zsuzsánna AU - Ábrahám, Edit AU - Sinka, Rita AU - Juhász, Szilvia AU - Lipinszki, Zoltán TI - Protein Phosphatase 4 Is Required for Centrobin Function in DNA Damage Repair JF - CELLS J2 - CELLS-BASEL VL - 12 PY - 2023 IS - 18 PG - 17 SN - 2073-4409 DO - 10.3390/cells12182219 UR - https://m2.mtmt.hu/api/publication/34129677 ID - 34129677 N1 - Export Date: 26 October 2023 Correspondence Address: Lipinszki, Z.; MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Hungary; email: lipinszki.zoltan@brc.hu Correspondence Address: Juhász, S.; Institute of Biochemistry, Hungary; email: juhasz.szilvia@brc.hu AB - Genome stability in human cells relies on the efficient repair of double-stranded DNA breaks, which is mainly achieved by homologous recombination (HR). Among the regulators of various cellular functions, Protein phosphatase 4 (PP4) plays a pivotal role in coordinating cellular response to DNA damage. Meanwhile, Centrobin (CNTRB), initially recognized for its association with centrosomal function and microtubule dynamics, has sparked interest due to its potential contribution to DNA repair processes. In this study, we investigate the involvement of PP4 and its interaction with CNTRB in HR-mediated DNA repair in human cells. Employing a range of experimental strategies, we investigate the physical interaction between PP4 and CNTRB and shed light on the importance of two specific motifs in CNTRB, the PP4-binding FRVP and the ATR kinase recognition SQ sequences, in the DNA repair process. Moreover, we examine cells depleted of PP4 or CNTRB and cells harboring FRVP and SQ mutations in CNTRB, which result in similar abnormal chromosome morphologies. This phenomenon likely results from the impaired resolution of Holliday junctions, which serve as crucial intermediates in HR. Taken together, our results provide new insights into the intricate mechanisms of PP4 and CNTRB-regulated HR repair and their interrelation. LA - English DB - MTMT ER - TY - JOUR AU - Verster, Kirsten I. AU - Cinege, Gyöngyi Ilona AU - Lipinszki, Zoltán AU - Magyar, Lilla Brigitta AU - Kurucz, Judit Éva AU - Tarnopol, Rebecca L. AU - Ábrahám, Edit AU - Darula, Zsuzsanna AU - Karageorgi, Marianthi AU - Tamsil, Josephine A. AU - Akalu, Saron M. AU - Andó, István AU - Whiteman, Noah K. TI - Evolution of insect innate immunity through domestication of bacterial toxins JF - PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA J2 - P NATL ACAD SCI USA VL - 120 PY - 2023 IS - 16 PG - 9 SN - 0027-8424 DO - 10.1073/pnas.2218334120 UR - https://m2.mtmt.hu/api/publication/33744872 ID - 33744872 N1 - Department of Integrative Biology, University of California, Berkeley, CA 94720, United States Innate Immunity Group, Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, 6726, Hungary MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, 6726, Hungary Doctoral School of Biology, University of Szeged, Szeged, 6720, Hungary Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720, United States Single Cell Omics Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Szeged, 6728, Hungary Laboratory of Proteomics Research, Biological Research Centre, Eötvös Loránd Research Network, Szeged, 6726, Hungary Department of Biology, Stanford University, Palo Alto, CA 94305, United States Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, United States Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, United States Cited By :2 Export Date: 10 July 2023 CODEN: PNASA Correspondence Address: Andó, I.; Innate Immunity Group, Hungary; email: ando@brc.hu Correspondence Address: Whiteman, N.K.; Department of Integrative Biology, United States; email: whiteman@berkeley.edu AB - Toxin cargo genes are often horizontally transferred by phages between bacterial species and are known to play an important role in the evolution of bacterial pathogenesis. Here, we show how these same genes have been horizontally transferred from phage or bacteria to animals and have resulted in novel adaptations. We discovered that two widespread bacterial genes encoding toxins of animal cells, cytolethal distending toxin subunit B ( cdtB ) and apoptosis-inducing protein of 56 kDa ( aip56) , were captured by insect genomes through horizontal gene transfer from bacteria or phages. To study the function of these genes in insects, we focused on Drosophila ananassae as a model. In the D. ananassae subgroup species, cdtB and aip56 are present as singular ( cdtB ) or fused copies ( cdtB::aip56 ) on the second chromosome. We found that cdtB and aip56 genes and encoded proteins were expressed by immune cells, some proteins were localized to the wasp embryo’s serosa, and their expression increased following parasitoid wasp infection. Species of the ananassae subgroup are highly resistant to parasitoid wasps, and we observed that D. ananassae lines carrying null mutations in cdtB and aip56 toxin genes were more susceptible to parasitoids than the wild type. We conclude that toxin cargo genes were captured by these insects millions of years ago and integrated as novel modules into their innate immune system. These modules now represent components of a heretofore undescribed defense response and are important for resistance to parasitoid wasps. Phage or bacterially derived eukaryotic toxin genes serve as macromutations that can spur the instantaneous evolution of novelty in animals. LA - English DB - MTMT ER - TY - JOUR AU - Ábrahám, Edit AU - Réthi-Nagy, Zsuzsánna AU - Vilmos, Péter AU - Sinka, Rita AU - Lipinszki, Zoltán TI - Plk4 Is a Novel Substrate of Protein Phosphatase 5 JF - INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES J2 - INT J MOL SCI VL - 24 PY - 2023 IS - 3 PG - 22 SN - 1661-6596 DO - 10.3390/ijms24032033 UR - https://m2.mtmt.hu/api/publication/33723350 ID - 33723350 N1 - Funding Agency and Grant Number: National Research, Development and Innovation Office [K132155]; National Laboratory for Biotechnology Program Grant [2022-2.1.1-NL-2022-00008]; Hungarian Academy of Sciences [BO/00329/15]; Lenduelet Program Grant [LP2017-7/2017] Funding text: This research was funded by the National Research, Development and Innovation Office to R.S. and Z.L. (K132155) and to P.V. and Z.L. (National Laboratory for Biotechnology Program Grant (2022-2.1.1-NL-2022-00008)), and the Hungarian Academy of Sciences (Bolyai Fellowship (BO/00329/15) and Lenduelet Program Grant (LP2017-7/2017)) to Z.L. AB - The conserved Ser/Thr protein phosphatase 5 (PP5) is involved in the regulation of key cellular processes, including DNA damage repair and cell division in eukaryotes. As a co-chaperone of Hsp90, PP5 has been shown to modulate the maturation and activity of numerous oncogenic kinases. Here, we identify a novel substrate of PP5, the Polo-like kinase 4 (Plk4), which is the master regulator of centriole duplication in animal cells. We show that PP5 specifically interacts with Plk4, and is able to dephosphorylate the kinase in vitro and in vivo, which affects the interaction of Plk4 with its partner proteins. In addition, we provide evidence that PP5 and Plk4 co-localize to the centrosomes in Drosophila embryos and cultured cells. We demonstrate that PP5 is not essential; the null mutant flies are viable without a severe mitotic phenotype; however, its loss significantly reduces the fertility of the animals. Our results suggest that PP5 is a novel regulator of the Plk4 kinase in Drosophila. LA - English DB - MTMT ER - TY - JOUR AU - Kartali, Tünde AU - Zsindely, Nóra AU - Nyilasi, Ildikó AU - Németh, Orsolya AU - Sávai, Gergő AU - Kocsubé, Sándor AU - Lipinszki, Zoltán AU - Patai, Roland AU - Spisák, Krisztina AU - Nagy, Gábor AU - Bodai, László AU - Vágvölgyi, Csaba AU - Papp, Tamás TI - Molecular Characterization of Novel Mycoviruses in Seven Umbelopsis Strains JF - VIRUSES J2 - VIRUSES-BASEL VL - 14 PY - 2022 IS - 11 PG - 20 SN - 1999-4915 DO - 10.3390/v14112343 UR - https://m2.mtmt.hu/api/publication/33194158 ID - 33194158 N1 - Funding Agency and Grant Number: NKFIA [K131796, ELKH 2001007]; Hungarian Academy of Sciences-Lendulet Program Grant [LP2017-7/2017]; National Research, Development and Innovation Office [FK-143326]; National Talent Programme; Ministry of Human Capacities [NTP-NFTO-22-B-0027, NTP-NFTO-22-B-0069]; ITM NKFIA [TKP-2021-EGA-28] Funding text: This study was supported by the grants NKFIA K131796, ELKH 2001007 and ITM NKFIA TKP-2021-EGA-28. Z.L. was supported by the Hungarian Academy of Sciences-Lendulet Program Grant (LP2017-7/2017). R.P. was supported by the National Research, Development and Innovation Office through the FK-143326 project; furthermore, R.P. and K.S. were also supported by the "National Talent Programme" with the financial aid of the Ministry of Human Capacities (NTP-NFTO-22-B-0027 and NTP-NFTO-22-B-0069). AB - The presence of viruses is less explored in Mucoromycota as compared to other fungal groups such as Ascomycota and Basidiomycota. Recently, more and more mycoviruses are identified from the early-diverging lineages of fungi. We have determined the genome of 11 novel dsRNA viruses in seven different Umbelopsis strains with next-generation sequencing (NGS). The identified viruses were named Umbelopsis ramanniana virus 5 (UrV5), 6a (UrV6a); 6b (UrV6b); 7 (UrV7); 8a (UrV8a); 8b (UrV8b); Umbelopsis gibberispora virus 1 (UgV1); 2 (UgV2) and Umbelopsis dimorpha virus 1a (UdV1a), 1b (UdV1b) and 2 (UdV2). All the newly identified viruses contain two open reading frames (ORFs), which putatively encode the coat protein (CP) and the RNA-dependent RNA polymerase (RdRp), respectively. Based on the phylogeny inferred from the RdRp sequences, eight viruses (UrV7, UrV8a, UrV8b, UgV1, UgV2, UdV1a, UdV1b and UdV2) belong to the genus Totivirus, while UrV5, UrV6a and UrV6b are placed into a yet unclassified but well-defined Totiviridae-related group. In UrV5, UgV1, UgV2, UrV8b, UdV1a, UdV2 and UdV1b, ORF2 is predicted to be translated as a fusion protein via a rare +1 (or-2) ribosomal frameshift, which is not characteristic to most members of the Totivirus genus. Virus particles 31 to 32 nm in diameter could be detected in the examined fungal strains by transmission electron microscopy. Through the identification and characterization of new viruses of Mucoromycota fungi, we can gain insight into the diversity of mycoviruses, as well as into their phylogeny and genome organization. LA - English DB - MTMT ER - TY - JOUR AU - Réthi-Nagy, Zsuzsánna AU - Ábrahám, Edit AU - Lipinszki, Zoltán TI - RESEARCH PROTOCOL GST-IVTT pull-down: a fast and versatile in vitro method for validating and mapping protein-protein interactions JF - FEBS OPEN BIO J2 - FEBS OPEN BIO VL - 12 PY - 2022 IS - 11 SP - 1988 EP - 1995 PG - 8 SN - 2211-5463 DO - 10.1002/2211-5463.13485 UR - https://m2.mtmt.hu/api/publication/33133587 ID - 33133587 N1 - Funding Agency and Grant Number: Ministry of Human Capacities of Hungary [NTP-NFT_O-22-B-0041]; Aron Marton College; Hungarian Academy of Sciences [BO/00329/15, LP2017-7/2017] Funding text: This research was funded by the Ministry of Human Capacities of Hungary (NTP-NFT_O-22-B-0041) and Aron Marton College to ZR-N, and the Hungarian Academy of Sciences (Bolyai Fellowship (BO/00329/15) and Lend_ulet Program Grant (LP2017-7/2017)) to ZL. AB - Over the past few decades, dozens of in vitro methods have been developed to map, investigate and validate protein-protein interactions. However, most of these approaches are time-consuming and labour-intensive or require specialised equipment or substantial amounts of purified proteins. Here, we describe a fast and versatile research protocol that is suitable for the in vitro analysis of the physical interaction between proteins or for mapping the binding surfaces. The principle of this method is based on the immobilisation of the protein/domain of interest to a carrier followed by its incubation with a labelled putative binding partner, which is generated by a coupled in vitro transcription/translation reaction. Interacting proteins are removed from the carrier, fractionated and visualised by SDS/PAGE autoradiography (or western blotting). This simple and cheap method can be easily carried out in every wet lab. LA - English DB - MTMT ER - TY - JOUR AU - Borkúti, Péter AU - Kristó, Ildikó AU - Szabó, Anikó AU - Bajusz, Csaba AU - Kovács, Zoltán AU - Réthi-Nagy, Zsuzsánna AU - Lipinszki, Zoltán AU - Lukacsovich, Tamas AU - Bogdan, Sven AU - Vilmos, Péter TI - Parallel import mechanisms ensure the robust nuclear localization of actin in Drosophila JF - FRONTIERS IN MOLECULAR BIOSCIENCES J2 - FRONT MOL BIOSCI VL - 9 PY - 2022 PG - 16 SN - 2296-889X DO - 10.3389/fmolb.2022.963635 UR - https://m2.mtmt.hu/api/publication/33133586 ID - 33133586 N1 - Funding Agency and Grant Number: NKFIH (National Research, Development and Innovation Office); Dr. Rollin D. Hotchkiss Foundation [NKFIH-871-3/2020, PD127968]; Hungarian Academy of Sciences Lenduelet Grant; [LP2017-7/2017] Funding text: This work was supported by NKFIH (National Research, Development and Innovation Office) through the National Laboratory for Biotechnology program, grant NKFIH-871-3/2020 (PV), and PD127968 (IK), the Dr. Rollin D. Hotchkiss Foundation (PB), and the Hungarian Academy of Sciences Lenduelet Grant LP2017-7/2017 (ZL). AB - Actin, as an ancient and fundamental protein, participates in various cytoplasmic as well as nuclear functions in eukaryotic cells. Based on its manifold tasks in the nucleus, it is a reasonable assumption that the nuclear presence of actin is essential for the cell, and consequently, its nuclear localization is ensured by a robust system. However, today only a single nuclear import and a single nuclear export pathway is known which maintain the dynamic balance between cytoplasmic and nuclear actin pools. In our work, we tested the robustness of the nuclear import of actin, and investigated whether the perturbations of nuclear localization affect the viability of the whole organism. For this aim, we generated a genetic system in Drosophila, in which we rescued the lethal phenotype of the null mutation of the Actin5C gene with transgenes that express different derivatives of actin, including a Nuclear Export Signal (NES)-tagged isoform which ensures forced nuclear export of the protein. We also disrupted the SUMOylation site of actin, suggested earlier to be responsible for nuclear retention, and eliminated the activity of the single nuclear import factor dedicated to actin. We found that, individually, none of the above mentioned manipulations led to a notable reduction in nuclear actin levels and thus, fully rescued lethality. However, the NES tagging of actin, together with the knock out of its importin, significantly reduced the amount of nuclear actin and induced lethality, confirming that the presence of actin in the nucleus is essential, and thereby, over-secured. Supporting this, we identified novel nuclear importins specific to actin, which sheds light on the mechanism behind the robustness of nuclear localization of actin, and supports the idea of essentiality of its nuclear functions. LA - English DB - MTMT ER -