45 results on '"Bailer SM"'
Search Results
2. TheraVision: Engineering platform technology for the development of oncolytic viruses based on herpes simplex virus type 1.
- Author
-
Funk C, Uhlig N, Ruzsics Z, Baur F, Peindl M, Nietzer S, Epting K, Vacun G, Dandekar G, Botteron C, Werno C, Grunwald T, and Bailer SM
- Abstract
Viruses are able to efficiently penetrate cells, multiply, and eventually kill infected cells, release tumor antigens, and activate the immune system. Therefore, viruses are highly attractive novel agents for cancer therapy. Clinical trials with first generations of oncolytic viruses (OVs) are very promising but show significant need for optimization. The aim of TheraVision was to establish a broadly applicable engineering platform technology for combinatorial oncolytic virus and immunotherapy. Through genetic engineering, an attenuated herpes simplex virus type 1 (HSV1) was generated that showed increased safety compared to the wild-type strain. To demonstrate the modularity and the facilitated generation of new OVs, two transgenes encoding retargeting as well as immunomodulating single-chain variable fragments (scFvs) were integrated into the platform vector. The resulting virus selectively infected epidermal growth factor receptor (EGFR)-expressing cells and produced a functional immune checkpoint inhibitor against programmed cell death protein 1 (PD-1). Thus, both viral-mediated oncolysis and immune-cell-mediated therapy were combined into a single viral vector. Safety and functionality of the armed OVs have been shown in novel preclinical models ranging from patient-derived organoids and tissue-engineered human in vitro 3D tumor models to complex humanized mouse models. Consequently, a novel and proprietary engineering platform vector based on HSV1 is available for the facilitated preclinical development of oncolytic virotherapy., Competing Interests: A patent is pending filed by Funk and Bailer entitled "platform vector based on herpes simplex virus 1 (HSV1) for the modular insertion of transgenes for use in oncolytic virus therapy.”, (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
3. The HSV1 Tail-Anchored Membrane Protein pUL34 Contains a Basic Motif That Supports Active Transport to the Inner Nuclear Membrane Prior to Formation of the Nuclear Egress Complex.
- Author
-
Funk C, Marques da Silveira E Santos D, Ott M, Raschbichler V, and Bailer SM
- Subjects
- Active Transport, Cell Nucleus, Amino Acid Motifs, Animals, Cell Line, Chlorocebus aethiops, HeLa Cells, Herpesvirus 1, Human genetics, Humans, Mutation, Vero Cells, Viral Proteins genetics, Virus Replication, Cell Nucleus metabolism, Herpesvirus 1, Human metabolism, Viral Proteins chemistry, Viral Proteins metabolism, Virus Release
- Abstract
Herpes simplex virus type 1 nucleocapsids are released from the host nucleus by a budding process through the nuclear envelope called nuclear egress. Two viral proteins, the integral membrane proteins pUL34 and pUL31, form the nuclear egress complex at the inner nuclear membrane, which is critical for this process. The nuclear import of both proteins ensues separately from each other: pUL31 is actively imported through the central pore channel, while pUL34 is transported along the peripheral pore membrane. With this study, we identified a functional bipartite NLS between residues 178 and 194 of pUL34. pUL34 lacking its NLS is mislocalized to the TGN but retargeted to the ER upon insertion of the authentic NLS or a mimic NLS, independent of the insertion site. If co-expressed with pUL31, either of the pUL34-NLS variants is efficiently, although not completely, targeted to the nuclear rim where co-localization with pUL31 and membrane budding seem to occur, comparable to the wild-type. The viral mutant HSV1(17
+ )Lox-UL34-NLS mt is modestly attenuated but viable and associated with localization of pUL34-NLS mt to both the nuclear periphery and cytoplasm. We propose that targeting of pUL34 to the INM is facilitated by, but not dependent on, the presence of an NLS, thereby supporting NEC formation and viral replication.- Published
- 2021
- Full Text
- View/download PDF
4. Patterns of Autologous and Nonautologous Interactions Between Core Nuclear Egress Complex (NEC) Proteins of α-, β- and γ-Herpesviruses.
- Author
-
Häge S, Sonntag E, Borst EM, Tannig P, Seyler L, Bäuerle T, Bailer SM, Lee CP, Müller R, Wangen C, Milbradt J, and Marschall M
- Subjects
- Amino Acid Sequence, Animals, Biomarkers, Cell Line, Cell Nucleus metabolism, Disease Models, Animal, Humans, Mice, Models, Biological, Nuclear Envelope metabolism, Protein Binding, Viral Proteins chemistry, Viral Proteins metabolism, Virus Replication, Herpesviridae physiology, Herpesviridae Infections metabolism, Herpesviridae Infections virology, Host-Pathogen Interactions, Virus Release
- Abstract
Nuclear egress is a regulated process shared by α-, β- and γ-herpesviruses. The core nuclear egress complex (NEC) is composed of the membrane-anchored protein homologs of human cytomegalovirus (HCMV) pUL50, murine cytomegalovirus (MCMV) pM50, Epstein-Barr virus (EBV) BFRF1 or varicella zoster virus (VZV) Orf24, which interact with the autologous NEC partners pUL53, pM53, BFLF2 or Orf27, respectively. Their recruitment of additional proteins leads to the assembly of a multicomponent NEC, coordinately regulating viral nucleocytoplasmic capsid egress. Here, the functionality of VZV, HCMV, MCMV and EBV core NECs was investigated by coimmunoprecipitation and confocal imaging analyses. Furthermore, a recombinant MCMV, harboring a replacement of ORF M50 by UL50, was analyzed both in vitro and in vivo. In essence, core NEC interactions were strictly limited to autologous NEC pairs and only included one measurable nonautologous interaction between the homologs of HCMV and MCMV. A comparative analysis of MCMV-WT versus MCMV-UL50-infected murine fibroblasts revealed almost identical phenotypes on the levels of protein and genomic replication kinetics. In infected BALB/c mice, virus spread to lung and other organs was found comparable between these viruses, thus stating functional complementarity. In conclusion, our study underlines that herpesviral core NEC proteins are functionally conserved regarding complementarity of core NEC interactions, which were found either virus-specific or restricted within subfamilies.
- Published
- 2020
- Full Text
- View/download PDF
5. Cold atmospheric plasma as antiviral therapy - effect on human herpes simplex virus type 1.
- Author
-
Bunz O, Mese K, Funk C, Wulf M, Bailer SM, Piwowarczyk A, and Ehrhardt A
- Subjects
- Acyclovir pharmacology, Animals, Antiviral Agents pharmacology, Cell Line, Chlorocebus aethiops, Humans, Vero Cells, Herpes Simplex therapy, Herpesvirus 1, Human drug effects, Plasma Gases pharmacology
- Abstract
In previous studies, cold atmospheric plasma (CAP) was explored as an antibacterial and antiviral agent for the treatment of chronic wounds. The aim of the present study was to investigate whether CAP may also be suitable as an antiviral therapy against herpes simplex virus type 1 (HSV-1). HSV-1 most frequently manifests as recurrent herpes labialis, but it can also cause encephalitis, conjunctivitis or herpes neonatorum as a perinatal infection. HSV-1 encoding the reporter gene GFP was propagated. The CAP dose for HSV-1 treatment was gradually increased, ranging from 0-150 s, and aciclovir was used as a positive control. After CAP treatment, the virus suspension was applied to a standard HSV research cell line (Vero cells) and the neuroblastoma cell line SH-SY5Y as a model for neuronal infection. The results showed that plasma treatment had a negligible antiviral effect on HSV-1 in both Vero- and SH-SY5Y cells at high dose. However, when we lowered the viral load 100-fold, we observed a significantly decreased number of internalized HSV-1 genomes 3 h post-infection for CAP-treated viruses. This difference was less pronounced with respect to GFP expression levels 24 h post-infection, which was in sharp contrast to the acyclovir-treated positive control, for which the viral load was reduced from 95 to 25%. In summary, we observed a low but measurable antiviral effect of CAP on HSV-1.
- Published
- 2020
- Full Text
- View/download PDF
6. Comprehensive analysis of nuclear export of herpes simplex virus type 1 tegument proteins and their Epstein-Barr virus orthologs.
- Author
-
Funk C, Raschbichler V, Lieber D, Wetschky J, Arnold EK, Leimser J, Biggel M, Friedel CC, Ruzsics Z, and Bailer SM
- Subjects
- Animals, Chlorocebus aethiops, HeLa Cells, Herpesvirus 1, Human physiology, Herpesvirus 4, Human physiology, Humans, Vero Cells, Viral Matrix Proteins metabolism, Virus Replication, Herpesvirus 1, Human metabolism, Herpesvirus 4, Human metabolism, Nuclear Export Signals, Viral Matrix Proteins chemistry
- Abstract
Morphogenesis of herpesviral virions is initiated in the nucleus but completed in the cytoplasm. Mature virions contain more than 25 tegument proteins many of which perform both nuclear and cytoplasmic functions suggesting they shuttle between these compartments. While nuclear import of herpesviral proteins was shown to be crucial for viral propagation, active nuclear export and its functional impact are still poorly understood. To systematically analyze nuclear export of tegument proteins present in virions of Herpes simplex virus type 1 (HSV1) and Epstein-Barr virus (EBV), the Nuclear EXport Trapped by RAPamycin (NEX-TRAP) was applied. Nine of the 22 investigated HSV1 tegument proteins including pUL4, pUL7, pUL11, pUL13, pUL21, pUL37d11, pUL47, pUL48 and pUS2 as well as 2 out of 6 EBV orthologs harbor nuclear export activity. A functional leucine-rich nuclear export sequence (NES) recognized by the export factor CRM1/Xpo1 was identified in six of them. The comparison between experimental and bioinformatic data indicates that experimental validation of predicted NESs is required. Mutational analysis of the pUL48/VP16 NES revealed its importance for herpesviral propagation. Together our data suggest that nuclear export is an important feature of the herpesviral life cycle required to co-ordinate nuclear and cytoplasmic processes., (© 2018 The Authors. Traffic published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
7. Nuclear egress of TDP-43 and FUS occurs independently of Exportin-1/CRM1.
- Author
-
Ederle H, Funk C, Abou-Ajram C, Hutten S, Funk EBE, Kehlenbach RH, Bailer SM, and Dormann D
- Subjects
- DNA-Binding Proteins chemistry, Humans, Karyopherins chemistry, Protein Binding, Protein Sorting Signals, Protein Transport, RNA, Messenger genetics, RNA, Messenger metabolism, RNA-Binding Protein FUS chemistry, Receptors, Cytoplasmic and Nuclear chemistry, Exportin 1 Protein, Cell Nucleus metabolism, DNA-Binding Proteins metabolism, Karyopherins metabolism, RNA-Binding Protein FUS metabolism, Receptors, Cytoplasmic and Nuclear metabolism
- Abstract
TDP-43 and FUS are nuclear proteins with multiple functions in mRNA processing. They play key roles in ALS (amyotrophic lateral sclerosis) and FTD (frontotemporal dementia), where they are partially lost from the nucleus and aggregate in the cytoplasm of neurons and glial cells. Defects in nucleocytoplasmic transport contribute to this pathology, hence nuclear import of both proteins has been studied in detail. However, their nuclear export routes remain poorly characterized and it is unclear whether aberrant nuclear export contributes to TDP-43 or FUS pathology. Here we show that predicted nuclear export signals in TDP-43 and FUS are non-functional and that both proteins are exported independently of the export receptor CRM1/Exportin-1. Silencing of Exportin-5 or the mRNA export factor Aly/REF, as well as mutations that abrogate RNA-binding do not impair export of TDP-43 and FUS. However, artificially enlarging TDP-43 or FUS impairs their nuclear egress, suggesting that they could leave the nucleus by passive diffusion. Finally, we found that inhibition of transcription causes accelerated nuclear egress of TDP-43, suggesting that newly synthesized RNA retains TDP-43 in the nucleus, limiting its egress into the cytoplasm. Our findings implicate reduced nuclear retention as a possible factor contributing to mislocalization of TDP-43 in ALS/FTD.
- Published
- 2018
- Full Text
- View/download PDF
8. Venture from the Interior-Herpesvirus pUL31 Escorts Capsids from Nucleoplasmic Replication Compartments to Sites of Primary Envelopment at the Inner Nuclear Membrane.
- Author
-
Bailer SM
- Abstract
Herpesviral capsid assembly is initiated in the nucleoplasm of the infected cell. Size constraints require that newly formed viral nucleocapsids leave the nucleus by an evolutionarily conserved vescular transport mechanism called nuclear egress. Mature capsids released from the nucleoplasm are engaged in a membrane-mediated budding process, composed of primary envelopment at the inner nuclear membrane and de-envelopment at the outer nuclear membrane. Once in the cytoplasm, the capsids receive their secondary envelope for maturation into infectious virions. Two viral proteins conserved throughout the herpesvirus family, the integral membrane protein pUL34 and the phosphoprotein pUL31, form the nuclear egress complex required for capsid transport from the infected nucleus to the cytoplasm. Formation of the nuclear egress complex results in budding of membrane vesicles revealing its function as minimal virus-encoded membrane budding and scission machinery. The recent structural analysis unraveled details of the heterodimeric nuclear egress complex and the hexagonal coat it forms at the inside of budding vesicles to drive primary envelopment. With this review, I would like to present the capsid-escort-model where pUL31 associates with capsids in nucleoplasmic replication compartments for escort to sites of primary envelopment thereby coupling capsid maturation and nuclear egress., Competing Interests: The author declares no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
9. Begomoviral Movement Protein Effects in Human and Plant Cells: Towards New Potential Interaction Partners.
- Author
-
Krapp S, Schuy C, Greiner E, Stephan I, Alberter B, Funk C, Marschall M, Wege C, Bailer SM, Kleinow T, and Krenz B
- Subjects
- Animals, Begomovirus chemistry, Begomovirus growth & development, Biological Transport, COS Cells, Cell Proliferation, Chlorocebus aethiops, Gene Silencing, HeLa Cells, Humans, Intermediate Filaments drug effects, Microtubules drug effects, Nocodazole pharmacology, Peptidyl-Prolyl Cis-Trans Isomerase NIMA-Interacting 4 genetics, Peptidyl-Prolyl Cis-Trans Isomerase NIMA-Interacting 4 metabolism, Plant Cells virology, Plant Proteins metabolism, Plant Viral Movement Proteins chemistry, Plant Viral Movement Proteins genetics, Protein Domains, Viral Proteins chemistry, Viral Proteins genetics, Viral Proteins metabolism, Begomovirus metabolism, Intermediate Filaments metabolism, Microtubules metabolism, Plant Viral Movement Proteins metabolism, Nicotiana virology
- Abstract
Geminiviral single-stranded circular DNA genomes replicate in nuclei so that the progeny DNA has to cross both the nuclear envelope and the plasmodesmata for systemic spread within plant tissues. For intra- and intercellular transport, two proteins are required: a nuclear shuttle protein (NSP) and a movement protein (MP). New characteristics of ectopically produced Abutilon mosaic virus (AbMV) MP (MP
AbMV ), either authentically expressed or fused to a yellow fluorescent protein or epitope tags, respectively, were determined by localization studies in mammalian cell lines in comparison to plant cells. Wild-type MPAbMV and the distinct MPAbMV : reporter protein fusions appeared as curled threads throughout mammalian cells. Co-staining with cytoskeleton markers for actin, intermediate filaments, or microtubules identified these threads as re-organized microtubules. These were, however, not stabilized by the viral MP, as demonstrated by nocodazole treatment. The MP of a related bipartite New World begomovirus, Cleome leaf crumple virus (ClLCrV), resulted in the same intensified microtubule bundling, whereas that of a nanovirus did not. The C-terminal section of MPAbMV , i.e., the protein's oligomerization domain, was dispensable for the effect. However, MP expression in plant cells did not affect the microtubules network. Since plant epidermal cells are quiescent whilst mammalian cells are proliferating, the replication-associated protein RepAbMV protein was then co-expressed with MPAbMV to induce cell progression into S-phase, thereby inducing distinct microtubule bundling without MP recruitment to the newly formed threads. Co-immunoprecipitation of MPAbMV in the presence of RepAbMV , followed by mass spectrometry identified potential novel MPAbMV -host interaction partners: the peptidyl-prolyl cis-trans isomerase NIMA-interacting 4 (Pin4) and stomatal cytokinesis defective 2 (SCD2) proteins. Possible roles of these putative interaction partners in the begomoviral life cycle and cytoskeletal association modes are discussed., Competing Interests: The authors declare no conflict of interest.- Published
- 2017
- Full Text
- View/download PDF
10. Herpesviral vectors and their application in oncolytic therapy, vaccination, and gene transfer.
- Author
-
Bailer SM, Funk C, Riedl A, and Ruzsics Z
- Subjects
- Animals, Genetic Therapy methods, Humans, Vaccination methods, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Genetic Vectors genetics, Genetic Vectors immunology, Herpesviridae genetics, Herpesviridae immunology, Oncolytic Viruses genetics, Oncolytic Viruses immunology
- Abstract
Herpesviruses are enveloped DNA viruses that infect vertebrate cells. Their high potential cloning capacity and the lifelong persistence of their genomes in various host cells make them attractive platforms for vector-based therapy. In this review, we would like to highlight recent advances of three major areas of herpesvirus vector development and application: (i) oncolytic therapy, (ii) recombinant vaccines, and (iii) large capacity gene transfer vehicles.
- Published
- 2017
- Full Text
- View/download PDF
11. Pathogens Inactivated by Low-Energy-Electron Irradiation Maintain Antigenic Properties and Induce Protective Immune Responses.
- Author
-
Fertey J, Bayer L, Grunwald T, Pohl A, Beckmann J, Gotzmann G, Casado JP, Schönfelder J, Rögner FH, Wetzel C, Thoma M, Bailer SM, Hiller E, Rupp S, and Ulbert S
- Subjects
- Antigens, Bacterial immunology, Antigens, Viral immunology, Electrons, Escherichia coli immunology, Vaccines, Inactivated immunology, Viruses immunology, Antigens, Bacterial radiation effects, Antigens, Viral radiation effects, Disinfection methods, Escherichia coli radiation effects, Radiation, Ionizing, Viruses radiation effects
- Abstract
Inactivated vaccines are commonly produced by incubating pathogens with chemicals such as formaldehyde or β-propiolactone. This is a time-consuming process, the inactivation efficiency displays high variability and extensive downstream procedures are often required. Moreover, application of chemicals alters the antigenic components of the viruses or bacteria, resulting in reduced antibody specificity and therefore stimulation of a less effective immune response. An alternative method for inactivation of pathogens is ionizing radiation. It acts very fast and predominantly damages nucleic acids, conserving most of the antigenic structures. However, currently used irradiation technologies (mostly gamma-rays and high energy electrons) require large and complex shielding constructions to protect the environment from radioactivity or X-rays generated during the process. This excludes them from direct integration into biological production facilities. Here, low-energy electron irradiation (LEEI) is presented as an alternative inactivation method for pathogens in liquid solutions. LEEI can be used in normal laboratories, including good manufacturing practice (GMP)- or high biosafety level (BSL)-environments, as only minor shielding is necessary. We show that LEEI efficiently inactivates different viruses (influenza A (H3N8), porcine reproductive and respiratory syndrome virus (PRRSV), equine herpesvirus 1 (EHV-1)) and bacteria ( Escherichia coli ) and maintains their antigenicity. Moreover, LEEI-inactivated influenza A viruses elicit protective immune responses in animals, as analyzed by virus neutralization assays and viral load determination upon challenge. These results have implications for novel ways of developing and manufacturing inactivated vaccines with improved efficacy., Competing Interests: The technology described in this manuscript is part of an IP application (WO 2015011265).
- Published
- 2016
- Full Text
- View/download PDF
12. Asna1/TRC40 that mediates membrane insertion of tail-anchored proteins is required for efficient release of Herpes simplex virus 1 virions.
- Author
-
Ott M, Marques D, Funk C, and Bailer SM
- Subjects
- Animals, Cell Line, Gene Knockdown Techniques, Humans, Microscopy, Fluorescence, Protein Interaction Mapping, Two-Hybrid System Techniques, Arsenite Transporting ATPases metabolism, Herpesvirus 1, Human physiology, Host-Pathogen Interactions, Virus Release
- Abstract
Background: Herpes simplex virus type 1 (HSV1), a member of the alphaherpesvirinae, can cause recurrent facial lesions and encephalitis. Two membrane envelopment processes, one at the inner nuclear membrane and a second at cytoplasmic membranes are crucial for a productive viral infection. Depending on the subfamily, herpesviruses encode more than 11 different transmembrane proteins including members of the tail-anchored protein family. HSV1 encodes three tail-anchored proteins pUL34, pUL56 and pUS9 characterized by a single hydrophobic region positioned at their C-terminal end that needs to be released from the ribosome prior to posttranslational membrane insertion. Asna1/TRC40 is an ATPase that targets tail-anchored proteins to the endoplasmic reticulum in a receptor-dependent manner. Cell biological data point to a critical and general role of Asna1/TRC40 in tail-anchored protein biogenesis. With this study, we aimed to determine the importance of the tail-anchored insertion machinery for HSV1 infection., Methods: To determine protein-protein interactions, the yeast-two hybrid system was applied. Asna1/TRC40 was depleted using RNA interference. Transient transfection and virus infection experiments followed by indirect immunofluorescence analysis were applied to analyse the localization of viral proteins as well as the impact of Asna1/TRC40 depletion on virus infection., Results: All HSV1 tail-anchored proteins specifically bound to Asna1/TRC40 but independently localized to their target membranes. While non-essential for cell viability, Asna1/TRC40 is required for efficient HSV1 replication. We show that early events of the replication cycle like virion entry and overall viral gene expression were unaffected by depletion of Asna1/TRC40. Furthermore, equal amounts of infectious virions were formed and remained cell-associated. This indicated that both nuclear egress of capsids that requires the essential tail-anchored protein pUL34, and secondary envelopment to form infectious virions were successfully completed. Despite large part of the virus life cycle proceeding normally, viral propagation was more than 10 fold reduced. We show that depletion of Asna1/TRC40 specifically affected a step late in infection during release of infectious virions to the extracellular milieu., Conclusions: Asna1/TRC40 is required at a late step of herpesviral infection for efficient release of mature virions to the extracellular milieu. This study reveals novel tools to decipher exocytosis of newly formed virions as well as hitherto unknown cellular targets for antiviral therapy.
- Published
- 2016
- Full Text
- View/download PDF
13. Subcellular Trafficking and Functional Relationship of the HSV-1 Glycoproteins N and M.
- Author
-
Striebinger H, Funk C, Raschbichler V, and Bailer SM
- Subjects
- Endoplasmic Reticulum metabolism, Gene Deletion, Golgi Apparatus metabolism, HeLa Cells, Herpesvirus 1, Human genetics, Humans, Membrane Glycoproteins genetics, Protein Binding, Protein Transport, Viral Matrix Proteins genetics, Viral Plaque Assay, Viral Proteins genetics, Herpesvirus 1, Human physiology, Membrane Glycoproteins metabolism, Viral Matrix Proteins metabolism, Viral Proteins metabolism, Virus Assembly, Virus Internalization
- Abstract
The herpes simplex virus type 1 (HSV-1) glycoprotein N (gN/UL49.5) is a type I transmembrane protein conserved throughout the herpesvirus family. gN is a resident of the endoplasmic reticulum that in the presence of gM is translocated to the trans Golgi network. gM and gN are covalently linked by a single disulphide bond formed between cysteine 46 of gN and cysteine 59 of gM. Exit of gN from the endoplasmic reticulum requires the N-terminal core of gM composed of eight transmembrane domains but is independent of the C-terminal extension of gM. Co-transport of gN and gM to the trans Golgi network also occurs upon replacement of conserved cysteines in gM and gN, suggesting that their physical interaction is mediated by covalent and non-covalent forces. Deletion of gN/UL49.5 using bacterial artificial chromosome (BAC) mutagenesis generated mutant viruses with wild-type growth behaviour, while full deletion of gM/UL10 resulted in an attenuated phenotype. Deletion of gN/UL49.5 in conjunction with various gM/UL10 mutants reduced average plaque sizes to the same extent as either single gM/UL10 mutant, indicating that gN is nonessential for the function performed by gM. We propose that gN functions in gM-dependent as well as gM-independent processes during which it is complemented by other viral factors.
- Published
- 2016
- Full Text
- View/download PDF
14. Subcellular trafficking and functional importance of herpes simplex virus type 1 glycoprotein M domains.
- Author
-
Striebinger H, Zhang J, Ott M, Funk C, Radtke K, Duron J, Ruzsics Z, Haas J, Lippé R, and Bailer SM
- Subjects
- Amino Acid Motifs, Herpesvirus 1, Human chemistry, Herpesvirus 1, Human genetics, Humans, Membrane Glycoproteins genetics, Protein Structure, Tertiary, Protein Transport, Viral Proteins genetics, Herpes Simplex virology, Herpesvirus 1, Human metabolism, Membrane Glycoproteins chemistry, Membrane Glycoproteins metabolism, Viral Proteins chemistry, Viral Proteins metabolism, trans-Golgi Network virology
- Abstract
Herpes simplex virus type 1 (HSV-1) glycoprotein M (gM/UL10) is a 473 aa type III transmembrane protein that resides in various membrane compartments. HSV-1 gM contains several putative trafficking motifs, but their functional relevance remains to be elucidated. We show here that transiently expressed gM 19–343 was sufficient for transport to the trans-Golgi network (TGN), whilst gM 133–473, where the first two transmembrane domains were deleted, and gM 1–342, which lacked the final residue of the last transmembrane domain, were retained in the endoplasmic reticulum (ER), indicating that all transmembrane domains are required for proper folding and ER exit. A series of bacterial artificial chromosome mutants revealed that in addition to the authentic start codon, translation of gM can be initiated at methionine 19 and 133/135. Whilst a protein lacking the first 18 residues supported WT-like growth, gM 133/135–473 resulted in reduced plaque diameters resembling a UL10 deletion mutant. An HSV-1 mutant encoding gM 1–342 showed similar growth characteristics and accumulated non-enveloped cytoplasmic particles, whilst gM 1–343 resulted in a gain of function, indicating that all transmembrane domains of the protein are important for viral growth. A C-terminal extension further supported viral propagation; however, the C-terminal trafficking motifs (residues 423–473) were completely dispensable. We propose a functional core within gM 19–343 comprised of all transmembrane domains that is sufficient to target the protein to the TGN, a favoured site for envelopment, and to support viral functions.
- Published
- 2015
- Full Text
- View/download PDF
15. The Herpes Simplex Virus Protein pUL31 Escorts Nucleocapsids to Sites of Nuclear Egress, a Process Coordinated by Its N-Terminal Domain.
- Author
-
Funk C, Ott M, Raschbichler V, Nagel CH, Binz A, Sodeik B, Bauerfeind R, and Bailer SM
- Subjects
- Animals, Chlorocebus aethiops, HeLa Cells, Humans, Microscopy, Fluorescence, Mutagenesis, Site-Directed, Vero Cells, Active Transport, Cell Nucleus physiology, Herpesvirus 1, Human physiology, Nucleocapsid Proteins metabolism, Virus Assembly physiology
- Abstract
Progeny capsids of herpesviruses leave the nucleus by budding through the nuclear envelope. Two viral proteins, the membrane protein pUL34 and the nucleo-phosphoprotein pUL31 form the nuclear egress complex that is required for capsid egress out of the nucleus. All pUL31 orthologs are composed of a diverse N-terminal domain with 1 to 3 basic patches and a conserved C-terminal domain. To decipher the functions of the N-terminal domain, we have generated several Herpes simplex virus mutants and show here that the N-terminal domain of pUL31 is essential with basic patches being critical for viral propagation. pUL31 and pUL34 entered the nucleus independently of each other via separate routes and the N-terminal domain of pUL31 was required to prevent their premature interaction in the cytoplasm. Unexpectedly, a classical bipartite nuclear localization signal embedded in this domain was not required for nuclear import of pUL31. In the nucleus, pUL31 associated with the nuclear envelope and newly formed capsids. Viral mutants lacking the N-terminal domain or with its basic patches neutralized still associated with nucleocapsids but were unable to translocate them to the nuclear envelope. Replacing the authentic basic patches with a novel artificial one resulted in HSV1(17+)Lox-UL31-hbpmp1mp2, that was viable but delayed in nuclear egress and compromised in viral production. Thus, while the C-terminal domain of pUL31 is sufficient for the interaction with nucleocapsids, the N-terminal domain was essential for capsid translocation to sites of nuclear egress and a coordinated interaction with pUL34. Our data indicate an orchestrated sequence of events with pUL31 binding to nucleocapsids and escorting them to the inner nuclear envelope. We propose a common mechanism for herpesviral nuclear egress: pUL31 is required for intranuclear translocation of nucleocapsids and subsequent interaction with pUL34 thereby coupling capsid maturation with primary envelopment.
- Published
- 2015
- Full Text
- View/download PDF
16. HCMV pUL135 remodels the actin cytoskeleton to impair immune recognition of infected cells.
- Author
-
Stanton RJ, Prod'homme V, Purbhoo MA, Moore M, Aicheler RJ, Heinzmann M, Bailer SM, Haas J, Antrobus R, Weekes MP, Lehner PJ, Vojtesek B, Miners KL, Man S, Wilkie GS, Davison AJ, Wang ECY, Tomasec P, and Wilkinson GWG
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes virology, Cytoskeletal Proteins metabolism, Host-Pathogen Interactions, Humans, Immunological Synapses virology, Immunomodulation, Killer Cells, Natural immunology, Killer Cells, Natural virology, Talin metabolism, Wiskott-Aldrich Syndrome Protein Family metabolism, Actin Cytoskeleton metabolism, Cytomegalovirus immunology, Viral Proteins physiology
- Abstract
Immune evasion genes help human cytomegalovirus (HCMV) establish lifelong persistence. Without immune pressure, laboratory-adapted HCMV strains have undergone genetic alterations. Among these, the deletion of the UL/b' domain is associated with loss of virulence. In a screen of UL/b', we identified pUL135 as a protein responsible for the characteristic cytopathic effect of clinical HCMV strains that also protected from natural killer (NK) and T cell attack. pUL135 interacted directly with abl interactor 1 (ABI1) and ABI2 to recruit the WAVE2 regulatory complex to the plasma membrane, remodel the actin cytoskeleton and dramatically reduce the efficiency of immune synapse (IS) formation. An intimate association between F-actin filaments in target cells and the IS was dispelled by pUL135 expression. Thus, F-actin in target cells plays a critical role in synaptogenesis, and this can be exploited by pathogens to protect against cytotoxic immune effector cells. An independent interaction between pUL135 and talin disrupted cell contacts with the extracellular matrix., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
17. Single-stranded DNA catalyzes hybridization of PCR-products to microarray capture probes.
- Author
-
Dally S, Rupp S, Lemuth K, Hartmann SC, Hiller E, Bailer SM, Knabbe C, and Weile J
- Subjects
- Biocatalysis, Nucleic Acid Hybridization, DNA, Single-Stranded metabolism, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction methods
- Abstract
Since its development, microarray technology has evolved to a standard method in the biotechnological and medical field with a broad range of applications. Nevertheless, the underlying mechanism of the hybridization process of PCR-products to microarray capture probes is still not completely understood, and several observed phenomena cannot be explained with current models. We investigated the influence of several parameters on the hybridization reaction and identified ssDNA to play a major role in the process. An increase of the ssDNA content in a hybridization reaction strongly enhanced resulting signal intensities. A strong influence could also be observed when unlabeled ssDNA was added to the hybridization reaction. A reduction of the ssDNA content resulted in a massive decrease of the hybridization efficiency. According to these data, we developed a novel model for the hybridization mechanism. This model is based on the assumption that single stranded DNA is necessary as catalyst to induce the hybridization of dsDNA. The developed hybridization model is capable of giving explanations for several yet unresolved questions regarding the functionality of microarrays. Our findings not only deepen the understanding of the hybridization process, but also have immediate practical use in data interpretation and the development of new microarrays.
- Published
- 2014
- Full Text
- View/download PDF
18. Comprehensive analysis of varicella-zoster virus proteins using a new monoclonal antibody collection.
- Author
-
Lenac Roviš T, Bailer SM, Pothineni VR, Ouwendijk WJ, Šimić H, Babić M, Miklić K, Malić S, Verweij MC, Baiker A, Gonzalez O, von Brunn A, Zimmer R, Früh K, Verjans GM, Jonjić S, and Haas J
- Subjects
- Animals, Blotting, Western, Cell Line, Cell Line, Tumor, Chickenpox virology, Epithelial Cells virology, Fluorescent Antibody Technique, Indirect, Herpes Zoster virology, Herpesvirus 3, Human immunology, Humans, Mice, Mice, Inbred BALB C, Proteomics, Skin immunology, Skin virology, Antibodies, Monoclonal immunology, Herpesvirus 3, Human metabolism, Viral Proteins immunology, Viral Proteins metabolism
- Abstract
Varicella-zoster virus (VZV) is the etiological agent of chickenpox and shingles. Due to the virus's restricted host and cell type tropism and the lack of tools for VZV proteomics, it is one of the least-characterized human herpesviruses. We generated 251 monoclonal antibodies (MAbs) against 59 of the 71 (83%) currently known unique VZV proteins to characterize VZV protein expression in vitro and in situ. Using this new set of MAbs, 44 viral proteins were detected by Western blotting (WB) and indirect immunofluorescence (IF); 13 were detected by WB only, and 2 were detected by IF only. A large proportion of viral proteins was analyzed for the first time in the context of virus infection. Our study revealed the subcellular localization of 46 proteins, 14 of which were analyzed in detail by confocal microscopy. Seven viral proteins were analyzed in time course experiments and showed a cascade-like temporal gene expression pattern similar to those of other herpesviruses. Furthermore, selected MAbs tested positive on human skin lesions by using immunohistochemistry, demonstrating the wide applicability of the MAb collection. Finally, a significant portion of the VZV-specific antibodies reacted with orthologs of simian varicella virus (SVV), thus enabling the systematic analysis of varicella in a nonhuman primate model system. In summary, this study provides insight into the potential function of numerous VZV proteins and novel tools to systematically study VZV and SVV pathogenesis.
- Published
- 2013
- Full Text
- View/download PDF
19. Mouse cytomegalovirus egress protein pM50 interacts with cellular endophilin-A2.
- Author
-
Lemnitzer F, Raschbichler V, Kolodziejczak D, Israel L, Imhof A, Bailer SM, Koszinowski U, and Ruzsics Z
- Subjects
- Acyltransferases antagonists & inhibitors, Acyltransferases genetics, Animals, Cytosol metabolism, Cytosol virology, Gene Expression, Host-Pathogen Interactions, Mass Spectrometry, Mice, Mutant Chimeric Proteins genetics, Nuclear Envelope metabolism, Nuclear Envelope virology, Protein Binding, Protein Interaction Mapping, RNA, Small Interfering genetics, Two-Hybrid System Techniques, Viral Proteins genetics, Virus Release, Acyltransferases metabolism, Muromegalovirus physiology, Mutant Chimeric Proteins metabolism, Viral Proteins metabolism
- Abstract
The herpesvirus replication cycle comprises maturation processes in the nucleus and cytoplasm of the infected cells. After their nuclear assembly viral capsids translocate via primary envelopment towards the cytoplasm. This event is mediated by the nuclear envelopment complex, which is composed by two conserved viral proteins belonging to the UL34 and UL31 protein families. Here, we generated recombinant viruses, which express affinity-tagged pM50 and/or pM53, the pUL34 and pUL31 homologues of the murine cytomegalovirus. We extracted pM50- and pM53-associated protein complexes from infected cells and analysed their composition after affinity purification by mass spectrometry. We observed reported interaction partners and identified new putative protein-protein interactions for both proteins. Endophilin-A2 was observed as the most prominent cellular partner of pM50. We found that endophilin-A2 binds to pM50 directly, and this interaction seems to be conserved in the pUL34 family., (© 2012 Blackwell Publishing Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
20. Determination of HSV-1 infectivity by plaque assay and a luciferase reporter cell line.
- Author
-
Lieber D and Bailer SM
- Subjects
- Animals, Antiviral Agents pharmacology, Cell Line, Herpesvirus 1, Human drug effects, Humans, Luciferases metabolism, Gene Expression, Genes, Reporter, Herpesvirus 1, Human physiology, Luciferases genetics, Viral Plaque Assay
- Abstract
Quantification of infectious virus is crucial to many experimental approaches in virological research. A broadly used and facile technique is the so-called "plaque assay" which provides precise information on the absolute quantity of infectivity in a given volume. Due to advances in the understanding of viral gene expression, transactivator-promoter pairs have been identified which can be used in transgenic cell lines as reporters of viral infection. Even though such "cellular reporter assay" systems are mostly restricted to relative quantification, they are attractive tools which can complement or replace the conventional plaque assay. Cellular reporter assays become especially interesting in state-of-the-art high-throughput screening approaches, as for instance RNAi and compound library screens, since they are often compatible with small-scale and automated experimentation. In this chapter, a regular plaque assay as well as a cellular reporter assay employing a luciferase reporter gene are described. As an example, HSV-1 infectivity is assessed with both methods yielding complementary information. Advantages and disadvantages of the two techniques and possible applications are discussed.
- Published
- 2013
- Full Text
- View/download PDF
21. A high-throughput yeast two-hybrid protocol to determine virus-host protein interactions.
- Author
-
Striebinger H, Koegl M, and Bailer SM
- Subjects
- Gene Library, Host-Pathogen Interactions, Viral Proteins genetics, Viral Proteins metabolism, Viruses genetics, High-Throughput Screening Assays methods, Protein Interaction Mapping methods, Two-Hybrid System Techniques, Viruses metabolism
- Abstract
The yeast two-hybrid (Y2H) system is a powerful method to identify and analyze binary protein interactions. In the field of virology, the Y2H system has significantly increased our knowledge of structure and function of viral proteins by systematically assessing intraviral protein interactions. Several comprehensive approaches to determine virus-host interactions have provided insight into viral strategies to manipulate the host for efficient replication and to escape host-derived countermeasures. To expand our knowledge of intraviral and virus-host protein interactions, we here present a Y2H protocol that is well suited for high-throughput screening. Yeast mating followed by liquid handling in a 96-well format as well as fluorescent readout of the reporter system provides a highly standardized and fully automated screening situation. The protocol can either be applied to screen complex host cDNA libraries or protein pairs arrayed for cross-testing. The ease of use, the cost-effectiveness as well as the robotic handling allows for extensive and multiple rounds of screening providing high coverage of protein-protein interactions. Thus, this protocol represents an improved "deep" screening method for high-throughput Y2H assays.
- Published
- 2013
- Full Text
- View/download PDF
22. NEX-TRAP, a novel method for in vivo analysis of nuclear export of proteins.
- Author
-
Raschbichler V, Lieber D, and Bailer SM
- Subjects
- Cell Nucleus metabolism, Cytological Techniques, Dimerization, Green Fluorescent Proteins genetics, HeLa Cells, Humans, Nuclear Localization Signals, Nuclear Proteins genetics, Nuclear Proteins metabolism, Recombinant Proteins, Sirolimus chemistry, Sirolimus pharmacology, Tacrolimus Binding Proteins genetics, Tacrolimus Binding Proteins metabolism, Viral Proteins genetics, Viral Proteins metabolism, trans-Golgi Network drug effects, trans-Golgi Network metabolism, Active Transport, Cell Nucleus
- Abstract
Transport of proteins between cytoplasm and nucleus is mediated by transport factors of the importin α- and β-families and occurs along a gradient of the small GTPase Ran. To date, in vivo analysis as well as prediction of protein nuclear export remain tedious and difficult. We generated a novel bipartite assay called NEX-TRAP (Nuclear EXport Trapped by RAPamycin) for in vivo analysis of protein nuclear export. The assay is based on the rapamycin-induced dimerization of the modules FRB (FK506-rapamycin (FR)-binding domain) and FKBP (FK506-binding protein-12): a potential nuclear export cargo is fused to FRB, to EYFP for direct visualization as well as to an SV40-derived nuclear localization signal (NLS) for constitutive nuclear import. An integral membrane protein that resides at the trans Golgi network (TGN) is fused to a cytoplasmically exposed FKBP and serves as reporter. EYFP-NLS-FRB fusion proteins with export activity accumulate in the nucleus at steady state but continuously shuttle between nucleus and cytoplasm. Rapamycin-induced dimerization of FRB and FKBP at the TGN traps the shuttling protein outside of the nucleus, making nuclear export permanent. Using several example cargoes, we show that the NEX-TRAP is superior to existing assays owing to its ease of use, its sensitivity and accuracy. Analysis of large numbers of export cargoes is facilitated by recombinational cloning. The NEX-TRAP holds the promise of applicability in automated fluorescence imaging for systematic analysis of nuclear export, thereby improving in silico prediction of nuclear export sequences., (© 2012 John Wiley & Sons A/S.)
- Published
- 2012
- Full Text
- View/download PDF
23. Cross-presentation and genome-wide screening reveal candidate T cells antigens for a herpes simplex virus type 1 vaccine.
- Author
-
Jing L, Haas J, Chong TM, Bruckner JJ, Dann GC, Dong L, Marshak JO, McClurkan CL, Yamamoto TN, Bailer SM, Laing KJ, Wald A, Verjans GM, and Koelle DM
- Subjects
- Adult, Antigens, Viral genetics, CD4-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes cytology, Cells, Cultured, Cytotoxicity, Immunologic immunology, Female, HLA Antigens genetics, HLA Antigens immunology, Herpes Simplex prevention & control, Herpesvirus 1, Human genetics, Humans, Interferon-gamma immunology, Male, Middle Aged, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology, Young Adult, Antigens, Viral immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cross-Priming immunology, Herpesvirus 1, Human immunology, Viral Vaccines immunology
- Abstract
Herpes simplex virus type 1 (HSV-1) not only causes painful recurrent oral-labial infections, it can also cause permanent brain damage and blindness. There is currently no HSV-1 vaccine. An effective vaccine must stimulate coordinated T cell responses, but the large size of the genome and the low frequency of HSV-1-specific T cells have hampered the search for the most effective T cell antigens for inclusion in a candidate vaccine. We have now developed what we believe to be novel methods to efficiently generate a genome-wide map of the responsiveness of HSV-1-specific T cells, and demonstrate the applicability of these methods to a second complex microbe, vaccinia virus. We used cross-presentation and CD137 activation-based FACS to enrich for polyclonal CD8+ T effector T cells. The HSV-1 proteome was prepared in a flexible format for analyzing both CD8+ and CD4+ T cells from study participants. Scans with participant-specific panels of artificial APCs identified an oligospecific response in each individual. Parallel CD137-based CD4+ T cell research showed discrete oligospecific recognition of HSV-1 antigens. Unexpectedly, the two HSV-1 proteins not previously considered as vaccine candidates elicited both CD8+ and CD4+ T cell responses in most HSV-1-infected individuals. In this era of microbial genomics, our methods - also demonstrated in principle for vaccinia virus for both CD8+ and CD4+ T cells - should be broadly applicable to the selection of T cell antigens for inclusion in candidate vaccines for many pathogens.
- Published
- 2012
- Full Text
- View/download PDF
24. Functional characterization of the essential tail anchor of the herpes simplex virus type 1 nuclear egress protein pUL34.
- Author
-
Ott M, Tascher G, Haßdenteufel S, Zimmermann R, Haas J, and Bailer SM
- Subjects
- Animals, Fluorescent Antibody Technique, Indirect methods, Gene Deletion, Herpesvirus 1, Human metabolism, Humans, Mutation, Nuclear Envelope metabolism, Nuclear Proteins genetics, Plasmids, Vero Cells, Viral Proteins genetics, Virus Replication, Herpesvirus 1, Human genetics, Nuclear Envelope chemistry, Nuclear Envelope virology, Nuclear Proteins metabolism, Viral Proteins metabolism
- Abstract
Release of herpes simplex virus type 1 (HSV-1) nucleocapsids from the host nucleus relies on the nuclear egress complex consisting of the two essential proteins pUL34 and pUL31. The cytoplasmically exposed N-terminal region of pUL34 interacts with pUL31, while a hydrophobic region followed by a short luminal part mediates membrane association. Based on its domain organization, pUL34 was postulated to be a tail-anchor (TA) protein. We performed a coupled in vitro transcription/translation assay to show that membrane insertion of pUL34 occurs post-translationally. Transient transfection and localization experiments in mammalian cells were combined with HSV-1 bacterial artificial chromosome mutagenesis to reveal the functional properties of the essential pUL34 TA. Our data show that a minimal tail length of 15 residues is sufficient for nuclear envelope targeting and pUL34 function. Permutations of the pUL34 TA with orthologous regions of human cytomegalovirus pUL50 or Epstein-Barr virus pBFRF1 as well as the heterologous HSV-1 TA proteins pUL56 or pUS9 or the cellular TA proteins Bcl-2 and Vamp2 revealed that nuclear egress tolerates TAs varying in sequence and hydrophobicity, while a non-α-helical membrane anchor failed to complement the pUL34 function. In conclusion, this study provides the first mechanistic insights into the particular role of the TA of pUL34 in membrane curving and capsid egress from the host nucleus.
- Published
- 2011
- Full Text
- View/download PDF
25. The Saccharomyces cerevisiae ubiquitin E3 ligase Asr1p targets calmodulin for ubiquitylation.
- Author
-
Fries T, Frank R, and Bailer SM
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Amino Acid Sequence, Molecular Sequence Data, Saccharomyces cerevisiae Proteins genetics, Stress, Physiological, Adaptor Proteins, Signal Transducing metabolism, Calcium metabolism, Calmodulin metabolism, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae Proteins metabolism, Ubiquitination
- Abstract
Yeast calmodulin known to be ubiquitylated in vivo in a Ca(2+) dependent manner has long remained an orphan substrate. Here we identify Saccharomyces cerevisiae Asr1p as an ubiquitin E3 ligase for yeast calmodulin, a protein involved in calcium signaling. A short region within Asr1p-C harboring two putative calmodulin-binding motifs is sufficient and necessary for interaction with calmodulin. The interaction is direct, occurs in vivo and depends on physiological concentrations of Ca(2+). A minimal set of purified proteins including Asr1p E3 ligase was sufficient for in vitro ubiquitylation of calmodulin, a reaction that required a functional Asr1p Ring domain. We propose a role of the Asr1p E3 ligase activity in coping with stress., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
26. The heterogeneous nuclear ribonucleoprotein K is important for Herpes simplex virus-1 propagation.
- Author
-
Schmidt T, Striebinger H, Haas J, and Bailer SM
- Subjects
- Animals, Blotting, Western, Chlorocebus aethiops, DNA, Viral genetics, HeLa Cells, Herpesvirus 1, Human genetics, Herpesvirus 1, Human metabolism, Heterogeneous-Nuclear Ribonucleoprotein K genetics, Humans, Microscopy, Fluorescence, Polymerase Chain Reaction, Vero Cells, Viral Proteins genetics, Viral Proteins metabolism, Virus Replication genetics, Herpesvirus 1, Human growth & development, Heterogeneous-Nuclear Ribonucleoprotein K metabolism, RNA Interference physiology, Virus Replication physiology
- Abstract
The heterogeneous nuclear ribonucleoprotein (hnRNP) K is an evolutionarily conserved protein with roles in signal transduction and gene expression. An impact of hnRNP K on the life cycle of a broad range of viral pathogens was reported while functional data for herpesviruses were lacking. In this study we show that hnRNP K is important for Herpes simplex virus 1 egress. In absence of hnRNP K, viral entry, gene expression, viral DNA replication, and maturation of nuclear particles appear normal whereas release of infectious virions to the extracellular space was significantly affected. Our results indicate that hnRNP K has an impact on a late step of herpesviral propagation making it a potential antiviral target., (Copyright © 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
27. Systematic analysis of the IgG antibody immune response against varicella zoster virus (VZV) using a self-assembled protein microarray.
- Author
-
Ceroni A, Sibani S, Baiker A, Pothineni VR, Bailer SM, LaBaer J, Haas J, and Campbell CJ
- Subjects
- Animals, Antigens, Viral genetics, Blotting, Western, Herpesvirus 3, Human genetics, Humans, Mice, Open Reading Frames genetics, Open Reading Frames immunology, Viral Proteins genetics, Antibodies, Viral immunology, Antigens, Viral immunology, Herpesvirus 3, Human immunology, Immunoglobulin G immunology, Protein Array Analysis methods, Viral Proteins immunology
- Abstract
Varicella zoster virus (VZV) is a human herpesvirus encoding at least 69 distinct viral proteins which causes chickenpox after primary infection and shingles during reactivation and which is particularly important in pregnancy and immunocompromised patients. Current serodiagnostic tests are either based on whole cell lysates or glycoprotein preparations. In order to investigate the humoral immune response to VZV infection or vaccination in more detail, and to improve the currently available diagnostic assays, we developed a nucleic acid programmable protein array (NAPPA) containing all 69 VZV proteins and performed a detailed analysis of 68 sera from individuals with either no, a previous or an acute VZV infection. In addition to the known reactive glycoprotein antigens (ORF 5, ORF 14, ORF 31, ORF 37, ORF 68), we discovered IgG antibodies against a variety of other membrane (ORF 2, ORF 24), capsid (ORF 20, ORF 23, ORF 43) and tegument (ORF 53, ORF 9, ORF 11) proteins, as well as other proteins involved in virus replication and assembly (ORF 25, ORF 26, ORF 28) and the transactivator proteins ORF 12, ORF 62 and ORF 63. All of these antigens were only reactive in a subset of VZV-positive individuals. A subset of the newly identified VZV antigens was validated by western blot analysis. Using these seroreactive new VZV antigens, more sensitive assays and tests distinguishing between different clinical entities may be developed.
- Published
- 2010
- Full Text
- View/download PDF
28. Systematic analysis of viral and cellular microRNA targets in cells latently infected with human gamma-herpesviruses by RISC immunoprecipitation assay.
- Author
-
Dölken L, Malterer G, Erhard F, Kothe S, Friedel CC, Suffert G, Marcinowski L, Motsch N, Barth S, Beitzinger M, Lieber D, Bailer SM, Hoffmann R, Ruzsics Z, Kremmer E, Pfeffer S, Zimmer R, Koszinowski UH, Grässer F, Meister G, and Haas J
- Subjects
- Cell Line, Cells, Cultured, Gene Expression Regulation, Gene Expression Regulation, Viral, Humans, MicroRNAs genetics, Microarray Analysis, RNA, Viral metabolism, Chromatin Immunoprecipitation methods, Herpesvirus 4, Human genetics, Herpesvirus 8, Human genetics, Host-Pathogen Interactions, MicroRNAs metabolism, Virology methods
- Abstract
The mRNA targets of microRNAs (miRNAs) can be identified by immunoprecipitation of Argonaute (Ago) protein-containing RNA-induced silencing complexes (RISCs) followed by microarray analysis (RIP-Chip). Here we used Ago2-based RIP-Chip to identify transcripts targeted by Kaposi's sarcoma-associated herpesvirus (KSHV) miRNAs (n = 114), Epstein-Barr virus (EBV) miRNAs (n = 44), and cellular miRNAs (n = 2337) in six latently infected or stably transduced human B cell lines. Of the six KSHV miRNA targets chosen for validation, four showed regulation via their 3'UTR, while two showed regulation via binding sites within coding sequences. Two genes governing cellular transport processes (TOMM22 and IPO7) were confirmed to be targeted by EBV miRNAs. A significant number of viral miRNA targets were upregulated in infected cells, suggesting that viral miRNAs preferentially target cellular genes induced upon infection. Transcript half-life both of cellular and viral miRNA targets negatively correlated with recruitment to RISC complexes, indicating that RIP-Chip offers a quantitative estimate of miRNA function., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
29. Evolutionarily conserved herpesviral protein interaction networks.
- Author
-
Fossum E, Friedel CC, Rajagopala SV, Titz B, Baiker A, Schmidt T, Kraus T, Stellberger T, Rutenberg C, Suthram S, Bandyopadhyay S, Rose D, von Brunn A, Uhlmann M, Zeretzke C, Dong YA, Boulet H, Koegl M, Bailer SM, Koszinowski U, Ideker T, Uetz P, Zimmer R, and Haas J
- Subjects
- Cluster Analysis, Evolution, Molecular, HeLa Cells, Herpesviridae metabolism, Herpesvirus 1, Human genetics, Herpesvirus 3, Human genetics, Herpesvirus 4, Human genetics, Herpesvirus 8, Human genetics, Humans, Immunohistochemistry, Muromegalovirus genetics, Phylogeny, Signal Transduction, Viral Core Proteins genetics, Viral Core Proteins metabolism, Virion metabolism, Herpesviridae genetics, Protein Interaction Mapping methods, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
Herpesviruses constitute a family of large DNA viruses widely spread in vertebrates and causing a variety of different diseases. They possess dsDNA genomes ranging from 120 to 240 kbp encoding between 70 to 170 open reading frames. We previously reported the protein interaction networks of two herpesviruses, varicella-zoster virus (VZV) and Kaposi's sarcoma-associated herpesvirus (KSHV). In this study, we systematically tested three additional herpesvirus species, herpes simplex virus 1 (HSV-1), murine cytomegalovirus and Epstein-Barr virus, for protein interactions in order to be able to perform a comparative analysis of all three herpesvirus subfamilies. We identified 735 interactions by genome-wide yeast-two-hybrid screens (Y2H), and, together with the interactomes of VZV and KSHV, included a total of 1,007 intraviral protein interactions in the analysis. Whereas a large number of interactions have not been reported previously, we were able to identify a core set of highly conserved protein interactions, like the interaction between HSV-1 UL33 with the nuclear egress proteins UL31/UL34. Interactions were conserved between orthologous proteins despite generally low sequence similarity, suggesting that function may be more conserved than sequence. By combining interactomes of different species we were able to systematically address the low coverage of the Y2H system and to extract biologically relevant interactions which were not evident from single species.
- Published
- 2009
- Full Text
- View/download PDF
30. Rtr1 is the Saccharomyces cerevisiae homolog of a novel family of RNA polymerase II-binding proteins.
- Author
-
Gibney PA, Fries T, Bailer SM, and Morano KA
- Subjects
- Amino Acid Sequence, Carrier Proteins chemistry, Carrier Proteins genetics, Cell Nucleus metabolism, Cytoplasm metabolism, Galactokinase genetics, Humans, Molecular Sequence Data, Promoter Regions, Genetic, Protein Transport, RNA Polymerase II chemistry, Saccharomyces cerevisiae chemistry, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Sequence Alignment, Temperature, Transcription Factors chemistry, Transcription Factors genetics, Transcription, Genetic, Zinc Fingers, Carrier Proteins metabolism, RNA Polymerase II metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Transcription Factors metabolism
- Abstract
Cells must rapidly sense and respond to a wide variety of potentially cytotoxic external stressors to survive in a constantly changing environment. In a search for novel genes required for stress tolerance in Saccharomyces cerevisiae, we identified the uncharacterized open reading frame YER139C as a gene required for growth at 37 degrees C in the presence of the heat shock mimetic formamide. YER139C encodes the closest yeast homolog of the human RPAP2 protein, recently identified as a novel RNA polymerase II (RNAPII)-associated factor. Multiple lines of evidence support a role for this gene family in transcription, prompting us to rename YER139C RTR1 (regulator of transcription). The core RNAPII subunits RPB5, RPB7, and RPB9 were isolated as potent high-copy-number suppressors of the rtr1Delta temperature-sensitive growth phenotype, and deletion of the nonessential subunits RPB4 and RPB9 hypersensitized cells to RTR1 overexpression. Disruption of RTR1 resulted in mycophenolic acid sensitivity and synthetic genetic interactions with a number of genes involved in multiple phases of transcription. Consistently, rtr1Delta cells are defective in inducible transcription from the GAL1 promoter. Rtr1 constitutively shuttles between the cytoplasm and nucleus, where it physically associates with an active RNAPII transcriptional complex. Taken together, our data reveal a role for members of the RTR1/RPAP2 family as regulators of core RNAPII function.
- Published
- 2008
- Full Text
- View/download PDF
31. A novel conserved nuclear localization signal is recognized by a group of yeast importins.
- Author
-
Fries T, Betz C, Sohn K, Caesar S, Schlenstedt G, and Bailer SM
- Subjects
- Active Transport, Cell Nucleus drug effects, Active Transport, Cell Nucleus genetics, Adaptor Proteins, Signal Transducing, Animals, Carrier Proteins genetics, Connectin, Ethanol pharmacology, Histones genetics, Histones metabolism, Karyopherins genetics, Mammals genetics, Mammals metabolism, Muscle Proteins genetics, Muscle Proteins metabolism, Mutation, Nuclear Localization Signals genetics, Protein Kinases genetics, Protein Kinases metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Solvents pharmacology, Stress, Physiological metabolism, Carrier Proteins metabolism, Cell Nucleus metabolism, Karyopherins metabolism, Nuclear Localization Signals metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
Nucleo-cytoplasmic transport of proteins is mostly mediated by specific interaction between transport receptors of the importin beta family and signal sequences present in their cargo. While several signal sequences, in particular the classical nuclear localization signal (NLS) recognized by the heterodimeric importin alpha/beta complex are well known, the signals recognized by other importin beta-like transport receptors remain to be characterized in detail. Here we present the systematic analysis of the nuclear import of Saccharomyces cerevisiae Asr1p, a nonessential alcohol-responsive Ring/PHD finger protein that shuttles between nucleus and cytoplasm but accumulates in the nucleus upon alcohol stress. Nuclear import of Asr1p is constitutive and mediated by its C-terminal domain. A short sequence comprising residues 243-280 is sufficient and necessary for active targeting to the nucleus. Moreover, the nuclear import signal is conserved from yeast to mammals. In vitro, the nuclear localization signal of Asr1p directly interacts with the importins Kap114p, Kap95p, Pse1p, Kap123p, or Kap104p, interactions that are sensitive to the presence of RanGTP. In vivo, these importins cooperate in nuclear import. Interestingly, the same importins mediate nuclear transport of histone H2A. Based on mutational analysis and sequence comparison with a region mediating nuclear import of histone H2A, we identified a novel type of NLS with the consensus sequence R/KxxL(x)(n)V/YxxV/IxK/RxxxK/R that is recognized by five yeast importins and connects them into a highly efficient network for nuclear import of proteins.
- Published
- 2007
- Full Text
- View/download PDF
32. Crystal structure of the archaeal A1Ao ATP synthase subunit B from Methanosarcina mazei Gö1: Implications of nucleotide-binding differences in the major A1Ao subunits A and B.
- Author
-
Schäfer IB, Bailer SM, Düser MG, Börsch M, Bernal RA, Stock D, and Grüber G
- Subjects
- ATP Synthetase Complexes genetics, ATP Synthetase Complexes isolation & purification, Conserved Sequence, Crystallography, X-Ray, Gene Expression, Models, Molecular, Protein Structure, Quaternary, Protein Structure, Tertiary, Protein Subunits chemistry, Protein Subunits genetics, Protein Subunits isolation & purification, Protein Subunits metabolism, Spectrometry, Fluorescence, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Structural Homology, Protein, Substrate Specificity, ATP Synthetase Complexes chemistry, ATP Synthetase Complexes metabolism, Methanosarcina enzymology, Nucleotides chemistry, Nucleotides metabolism
- Abstract
The A1Ao ATP synthase from archaea represents a class of chimeric ATPases/synthases, whose function and general structural design share characteristics both with vacuolar V1Vo ATPases and with F1Fo ATP synthases. The primary sequences of the two large polypeptides A and B, from the catalytic part, are closely related to the eukaryotic V1Vo ATPases. The chimeric nature of the A1Ao ATP synthase from the archaeon Methanosarcina mazei Gö1 was investigated in terms of nucleotide interaction. Here, we demonstrate the ability of the overexpressed A and B subunits to bind ADP and ATP by photoaffinity labeling. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry was used to map the peptide of subunit B involved in nucleotide interaction. Nucleotide affinities in both subunits were determined by fluorescence correlation spectroscopy, indicating a weaker binding of nucleotide analogues to subunit B than to A. In addition, the nucleotide-free crystal structure of subunit B is presented at 1.5 A resolution, providing the first view of the so-called non-catalytic subunit of the A1Ao ATP synthase. Superposition of the A-ATP synthase non-catalytic B subunit and the F-ATP synthase non-catalytic alpha subunit provides new insights into the similarities and differences of these nucleotide-binding ATPase subunits in particular, and into nucleotide binding in general. The arrangement of subunit B within the intact A1Ao ATP synthase is presented.
- Published
- 2006
- Full Text
- View/download PDF
33. Structural analysis of the stalk subunit Vma5p of the yeast V-ATPase in solution.
- Author
-
Armbrüster A, Svergun DI, Coskun U, Juliano S, Bailer SM, and Grüber G
- Subjects
- Amino Acid Sequence, Animals, Chromatography, Gel, Circular Dichroism, DNA chemistry, Escherichia coli metabolism, Hydrolysis, Manduca, Models, Molecular, Models, Statistical, Molecular Sequence Data, Protein Conformation, Protein Structure, Secondary, Protein Structure, Tertiary, Saccharomyces cerevisiae enzymology, Saccharomyces cerevisiae metabolism, Scattering, Radiation, Sequence Homology, Amino Acid, Water metabolism, X-Rays, Saccharomyces cerevisiae Proteins chemistry, Vacuolar Proton-Translocating ATPases chemistry
- Abstract
Vma5p (subunit C) of the yeast V-ATPase was produced in Escherichia coli and purified to homogeneity. Analysis of secondary structure by circular dichroism spectroscopy showed that Vma5p comprises 64% alpha-helix and 17% beta-sheet content. The molecular mass of this subunit, determined by gel filtration analysis and small angle X-ray scattering (SAXS), was approximately 51+/-4 kDa, indicating a high hydration level of the protein in solution. The radius of gyration and the maximum size of Vma5p were determined to be 3.74+/-0.03 and 12.5+/-0.1 nm, respectively. Using two independent ab initio approaches, the first low-resolution shape of the protein was determined. Vma5p is an elongated boot-shaped particle consisting of two distinct domains. Co-reconstitution of Vma5p to V1 without C from Manduca sexta resulted in a V1-Vma5p hybrid complex and a 20% increase in ATPase hydrolysis activity.
- Published
- 2004
- Full Text
- View/download PDF
34. Asr1p, a novel yeast ring/PHD finger protein, signals alcohol stress to the nucleus.
- Author
-
Betz C, Schlenstedt G, and Bailer SM
- Subjects
- Adaptor Proteins, Signal Transducing, Amino Acid Sequence, Carrier Proteins chemistry, Cytoplasm metabolism, Fungal Proteins chemistry, Green Fluorescent Proteins, Karyopherins metabolism, Luminescent Proteins metabolism, Microscopy, Fluorescence, Molecular Sequence Data, Monomeric GTP-Binding Proteins metabolism, Nuclear Proteins metabolism, Oxygen metabolism, Plasmids metabolism, Protein Binding, Protein Structure, Tertiary, Recombinant Fusion Proteins metabolism, Saccharomyces cerevisiae Proteins chemistry, Signal Transduction, Temperature, Time Factors, Exportin 1 Protein, Alcohols metabolism, Carrier Proteins metabolism, Cell Nucleus metabolism, Fungal Proteins physiology, Receptors, Cytoplasmic and Nuclear, Saccharomyces cerevisiae Proteins metabolism
- Abstract
During fermentation, yeast cells are exposed to increasing amounts of alcohol, which is stressful and affects both growth and viability. On the molecular level, numerous aspects of alcohol stress signaling remain unresolved. We have identified a novel yeast Ring/PHD finger protein that constitutively shuttles between nucleus and cytoplasm but accumulates in the nucleus upon exposure to ethanol, 2-propanol, or 1-butanol. Subcellular localization of this protein is not altered by osmotic, oxidative, or heat stress or during nitrogen or glucose starvation. Because of its exclusive sensitivity to environmental alcohol, the protein was called Asr1p for Alcohol Sensitive Ring/PHD finger 1 protein. Nuclear accumulation of Asr1p is rapid, reversible, and requires a functional Ran/Gsp1p gradient. Asr1p contains two N terminally located leucine-rich nuclear export sequences (NES) required for nuclear export. Consistently, it accumulates in the nucleus of xpo1-1 cells at restrictive temperature and forms a trimeric complex with the exportin Xpo1p and Ran-GTP. Deletion of ASR1 leads to sensitivity in growth on medium containing alcohol or detergent, consistent with a function of Asr1p in alcohol-related signaling. Asr1p is the first reported protein that changes its subcellular localization specifically upon exposure to alcohol and therefore represents a key element in the analysis of alcohol-responsive signaling.
- Published
- 2004
- Full Text
- View/download PDF
35. Nuclear accumulation of the small GTPase Gsp1p depends on nucleoporins Nup133p, Rat2p/Nup120p, Nup85p, Nic96p, and the acetyl-CoA carboxylase Acc1p.
- Author
-
Gao H, Sumanaweera N, Bailer SM, and Stochaj U
- Subjects
- Cell Division, Cytoplasm metabolism, Green Fluorescent Proteins, Hot Temperature, Luminescent Proteins metabolism, Microscopy, Fluorescence, Mutation, Plasmids metabolism, Protein Structure, Tertiary, Saccharomyces cerevisiae metabolism, Temperature, Time Factors, Acetyltransferases physiology, Cell Nucleus metabolism, Membrane Proteins physiology, Monomeric GTP-Binding Proteins metabolism, Nuclear Pore Complex Proteins physiology, Nuclear Proteins metabolism, Nuclear Proteins physiology, Saccharomyces cerevisiae Proteins physiology
- Abstract
The small GTPase Ran/Gsp1p plays an essential role in nuclear trafficking of macromolecules, as Ran/Gsp1p regulates many transport processes across the nuclear pore complex (NPC). To determine the role of nucleoporins in the generation of the nucleocytoplasmic Gsp1p concentration gradient, mutations in various nucleoporin genes were analyzed in the yeast Saccharomyces cerevisiae. We show that the nucleoporins Nup133p, Rat2p/Nup120p, Nup85p, Nic96p, and the enzyme acetyl-CoA carboxylase (MTR7) control the distribution and cellular concentration of Gsp1p. At the restrictive temperature the reporter protein GFP-Gsp1p, which is too large to diffuse across the nuclear envelope, fails to concentrate in nuclei of nup133delta, rat2-1, nup85delta, nic96deltaC, and mtr7-1 cells, demonstrating that GFP-Gsp1p nuclear import is deficient. In addition, the concentration of Gsp1p is severely reduced in mutants nup133Delta and mtr7-1 under these conditions. We have now identified the molecular mechanisms that contribute to the dissipation of the Gsp1p concentration gradient in these mutants. Loss of the Gsp1p gradient in nup133delta and rat2-1 can be explained by reduced binding of the Gsp1p nuclear carrier Ntf2p to NPCs. Likewise, nup85delta cells that mislocalize GFP-Gsp1p at the permissive as well as non-permissive temperature have a diminished association of Ntf2p-GFP with nuclear envelopes under both conditions. Moreover, under restrictive conditions Prp20p, the guanine nucleotide exchange factor for Gsp1p, mislocalizes to the cytoplasm in nup85delta, nic96deltaC, and mtr7-1 cells, thereby contributing to a collapse of the Gsp1p gradient. Taken together, components of the NPC subcomplex containing Rat2p/Nup120p, Nup133p, and Nup85p, in addition to proteins Nic96p and Mtr7p, are shown to be crucial for the formation of a nucleocytoplasmic Gsp1p gradient.
- Published
- 2003
- Full Text
- View/download PDF
36. Dimer formation of subunit G of the yeast V-ATPase.
- Author
-
Armbrüster A, Bailer SM, Koch MH, Godovac-Zimmermann J, and Grüber G
- Subjects
- Base Sequence, Chromatography, Gel, DNA Primers, Dimerization, Electrophoresis, Polyacrylamide Gel, Molecular Weight, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, Scattering, Radiation, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Vacuolar Proton-Translocating ATPases isolation & purification, Vacuolar Proton-Translocating ATPases metabolism, Saccharomyces cerevisiae enzymology, Vacuolar Proton-Translocating ATPases chemistry
- Abstract
The G subunit of the vacuolar ATPase (V-ATPase) is a component of the stalk connecting the V(1) and V(O) sectors of the enzyme and is essential for normal assembly and function. Subunit G (Vma10p) of the yeast V-ATPase was expressed in Escherichia coli as a soluble protein and was purified to homogeneity. The molecular mass of subunit G, determined by Native-polyacrylamide gel electrophoresis, gel filtration analysis and small-angle X-ray scattering, was approximately 28+/-2 kDa, indicating that this protein is dimeric. With a radius of gyration (R(g)) and a maximum size (D(max)) of 2.7+/-0.2 nm and 8.0+/-0.3 nm, respectively, the G-dimer is rather elongated. To understand which region of subunit G is required to mediate dimerization, a G(38-144) form (the carboxyl-terminus) was expressed and purified. G(38-144) is homogeneous, with a molecular mass of approximately 12+/-3 kDa, indicating a monomeric form in solution.
- Published
- 2003
- Full Text
- View/download PDF
37. Expression, purification, and characterization of subunit E, an essential subunit of the vacuolar ATPase.
- Author
-
Grüber G, Godovac-Zimmermann J, Link TA, Coskun U, Rizzo VF, Betz C, and Bailer SM
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Circular Dichroism, DNA Primers, Electrophoresis, Polyacrylamide Gel, Manduca enzymology, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments isolation & purification, Peptide Fragments metabolism, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Trypsin metabolism, Peptide Fragments genetics, Vacuolar Proton-Translocating ATPases chemistry
- Abstract
A recombinant form of subunit E (Vma4p) from yeast vacuolar ATPases (V-ATPases) has been overexpressed in Escherichia coli, purified to homogeneity, and explored by mass spectrometry. Analysis of the secondary structure of Vma4p by circular dichroism spectroscopy indicated 32% alpha-helix and 23% beta-sheet content. Vma4p formed a hybrid-complex with the nucleotide-binding subunits alpha and beta of the closely related F(1) ATPase of the thermophilic bacterium PS3 (TF(1)). The alpha(3)beta(3)E-hybrid-complex had 56% of the ATPase activity of the native TF(1). By comparison, an alpha(3)beta(3)-formation without Vma4p showed about 24% of total TF(1) ATPase activity. This is the first demonstration of a hydrolytically active hybrid-complex consisting of F(1) and V(1) subunits. The arrangement of subunit E in V(1) has been probed using the recombinant Vma4p, the alpha(3)beta(3)E-hybrid-complex together with V(1) and an A(3)B(3)HEG-subcomplex of the V(1) ATPase from Manduca sexta, respectively, indicating that subunit E is shielded in V(1).
- Published
- 2002
- Full Text
- View/download PDF
38. The Nsp1p carboxy-terminal domain is organized into functionally distinct coiled-coil regions required for assembly of nucleoporin subcomplexes and nucleocytoplasmic transport.
- Author
-
Bailer SM, Balduf C, and Hurt E
- Subjects
- Active Transport, Cell Nucleus physiology, Amino Acid Motifs physiology, Macromolecular Substances, Mutagenesis, Site-Directed, Nuclear Pore metabolism, Nuclear Pore Complex Proteins metabolism, Protein Binding physiology, Protein Structure, Secondary, Protein Structure, Tertiary physiology, Saccharomyces cerevisiae, Saccharomyces cerevisiae Proteins metabolism, Structure-Activity Relationship, Calcium-Binding Proteins, Cell Nucleus metabolism, Cytoplasm metabolism, Fungal Proteins chemistry, Fungal Proteins metabolism, Membrane Proteins, Nuclear Pore Complex Proteins biosynthesis, Nuclear Proteins chemistry, Nuclear Proteins metabolism
- Abstract
Nucleoporin Nsp1p, which has four predicted coiled-coil regions (coils 1 to 4) in the essential carboxy-terminal domain, is unique in that it is part of two distinct nuclear pore complex (NPC) subcomplexes, Nsp1p-Nup57p-Nup49p-Nic96p and Nsp1p-Nup82p-Nup159p. As shown by in vitro reconstitution, coiled-coil region 2 (residues 673 to 738) is sufficient to form heterotrimeric core complexes and can bind either Nup57p or Nup82p. Accordingly, interaction of Nup82p with Nsp1p coil 2 is competed by excess Nup57p. Strikingly, coil 3 and 4 mutants are still assembled into the core Nsp1p-Nup57p-Nup49p complex but no longer associate with Nic96p. Consistently, the Nsp1p-Nup57p-Nup49p core complex dissociates from the nuclear pores in nsp1 coil 3 and 4 mutant cells, and as a consequence, defects in nuclear protein import are observed. Finally, the nsp1-L640S temperature-sensitive mutation, which maps in coil 1, leads to a strong nuclear mRNA export defect. Thus, distinct coiled-coil regions within Nsp1p-C have separate functions that are related to the assembly of different NPC subcomplexes, nucleocytoplasmic transport, and incorporation into the nuclear pores.
- Published
- 2001
- Full Text
- View/download PDF
39. A sensitive homologous radioimmunoassay for human relaxin-2 (h-RLX-2) based on antibodies characterized by epitope mapping studies.
- Author
-
Armbruster FP, Grön HJ, Maier I, Becker S, Bailer SM, Lippert TH, Seeger H, Klöppinger M, Tampe J, Stoeva S, and Voelter W
- Subjects
- Amino Acid Sequence, Estradiol administration & dosage, Estradiol therapeutic use, Estrogen Replacement Therapy, Female, Humans, Male, Models, Molecular, Molecular Sequence Data, Pregnancy, Protein Conformation, Relaxin blood, Relaxin immunology, Relaxin urine, Semen chemistry, Sensitivity and Specificity, Antibodies immunology, Epitope Mapping, Radioimmunoassay methods, Relaxin analysis
- Abstract
We present a sensitive homologous radioimmunoassay (RIA) for the quantitative determination of human relaxin (hRLX) in human serum, plasma, seminal plasma, and urine. This assay is based on a rabbit antiserum which was generated using recombinant hRLX-2 as immunogen. Using 125I-hRLX-2 as tracer and a total incubation time of 20 - 24 hours the radioimmunoassay showed linearity in a range of 60 - 4000 ng/l, a lower detection limit of 38 ng/l and a mean recovery rate of 98.5%. Intraassay variation was 4.0% (mean = 526 ng/l) and 11.9% (mean = 2368 ng/l), and interassay variation 10.7% (mean = 256 ng/l) and 13.1% (mean = 2368 ng/l). Using hRLX-2 hexapeptides on polystyrene pins, epitopes recognized by the hRLX-2 specific rabbit antiserum were determined experimentally, and compared to predicted epitopes. Both methods led to comparable results. The antiserum, recognizing different epitopes, showed no cross-reactivity with human insulin, hZn-insulin, hIGF-I, hIGF-II, human inhibin alpha-subunit, two different forms of seminal plasma inhibin like peptide, spermolaxin, ubiquitin, prolactin, LH, FSH and hCG.
- Published
- 2001
40. Nup116p associates with the Nup82p-Nsp1p-Nup159p nucleoporin complex.
- Author
-
Bailer SM, Balduf C, Katahira J, Podtelejnikov A, Rollenhagen C, Mann M, Pante N, and Hurt E
- Subjects
- Cell Polarity, Cytoplasm, Humans, Membrane Proteins genetics, Nuclear Envelope ultrastructure, Nuclear Proteins genetics, Protein Binding, Recombinant Fusion Proteins metabolism, Staphylococcal Protein A genetics, Staphylococcal Protein A metabolism, Yeasts, Calcium-Binding Proteins, Fungal Proteins metabolism, Membrane Proteins metabolism, Nuclear Envelope metabolism, Nuclear Pore Complex Proteins, Nuclear Proteins metabolism, Saccharomyces cerevisiae Proteins
- Abstract
Nup116p is a GLFG nucleoporin involved in RNA export processes. We show here that Nup116p physically interacts with the Nup82p-Nsp1p-Nup159p nuclear pore subcomplex, which plays a central role in nuclear mRNA export. For this association, a sequence within the C-terminal domain of Nup116p that includes the conserved nucleoporin RNA-binding motif was sufficient and necessary. Consistent with this biochemical interaction, protein A-Nup116p and the protein A-tagged Nup116p C-terminal domain, like the members of the Nup82p complex, localized to the cytoplasmic side of the nuclear pore complex, as revealed by immunogold labeling. Finally, synthetic lethal interactions were found between mutant alleles of NUP116 and all members of the Nup82p complex. Thus, Nup116p consists of three independent functional domains: 1) the C-terminal part interacts with the Nup82p complex; 2) the Gle2p-binding sequence interacts with Gle2p/Rae1p; and 3) the GLFG domain interacts with shuttling transport receptors such as karyopherin-beta family members.
- Published
- 2000
- Full Text
- View/download PDF
41. Nup116p and nup100p are interchangeable through a conserved motif which constitutes a docking site for the mRNA transport factor gle2p.
- Author
-
Bailer SM, Siniossoglou S, Podtelejnikov A, Hellwig A, Mann M, and Hurt E
- Subjects
- Amino Acid Sequence, Biological Transport genetics, Cell Nucleus genetics, Cell Nucleus metabolism, Fungal Proteins genetics, Membrane Proteins genetics, Molecular Sequence Data, Mutagenesis, Insertional, Nuclear Envelope genetics, Nuclear Proteins genetics, Protein Structure, Tertiary, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Saccharomyces cerevisiae, Signal Transduction, Staphylococcal Protein A genetics, Conserved Sequence, Fungal Proteins metabolism, Membrane Proteins metabolism, Nuclear Matrix-Associated Proteins, Nuclear Pore Complex Proteins, Nuclear Proteins metabolism, Nucleocytoplasmic Transport Proteins, RNA, Messenger metabolism, Saccharomyces cerevisiae Proteins, Schizosaccharomyces pombe Proteins
- Abstract
Nup116p and Nup100p are highly related yeast GLFG nucleoporins, but only Nup116p is stoichiometrically bound to Gle2p, a previously identified mRNA export factor. A short Gle2p-binding sequence within Nup116p (GLEBS; residues 110-166) is sufficient and necessary to anchor Gle2p at the nuclear pores, whereas the carboxy-terminal domain of Nup116p mediates its own nuclear pore complex (NPC) association. The GLEBS is evolutionarily conserved and found in rat/Xenopus Nup98 and an uncharacterized Caenorhabditis elegans ORF, but is absent from Nup100p. When the GLEBS is deleted from Nup116p, Gle2p dissociates from the nuclear envelope and clusters of herniated nuclear pores form. When the GLEBS is inserted into Nup100p, Nup100p-GLEBS complements both the thermosensitive and NPC-herniated phenotype of nup116- cells, and Gle2p is retargeted concomitantly to the NPCs. Thus, the in vivo function of Gle2p is strictly coupled to the short GLEBS within Nup116p which links this putative mRNA transport factor to the nuclear pores.
- Published
- 1998
- Full Text
- View/download PDF
42. Characterization of nuclear pore protein p62 produced using baculovirus.
- Author
-
Bailer SM, Berlin WK, Starr CM, and Hanover JA
- Subjects
- Acetylglucosamine analysis, Amino Acid Sequence, Animals, Cell Line, Escherichia coli genetics, Gene Expression, Membrane Glycoproteins isolation & purification, Molecular Sequence Data, Molecular Structure, Nuclear Pore Complex Proteins, Nuclear Proteins isolation & purification, Rats, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Recombinant Proteins isolation & purification, Spodoptera, Membrane Glycoproteins biosynthesis, Membrane Glycoproteins genetics, Nuclear Proteins biosynthesis, Nuclear Proteins genetics, Nucleopolyhedroviruses genetics
- Abstract
Nuclear pore glycoproteins are essential components of the nuclear import apparatus in eukaryotes. In vertebrates, the most abundant of these glycoproteins is a molecule called p62. Like other O-linked N-acetylglucosamine glycoproteins, p62 is normally modified in the cytoplasm and cannot be overexpressed and conveniently collected in a secreted form. We devised an efficient scheme for expression and purification of recombinant p62 from Sf9 cells that may have general applicability for this class of glycoproteins. The purified rat p62 bound to wheat germ agglutinin, consistent with modification by O-linked N-acetylglucosamine. Carbohydrate analysis, in conjunction with amino acid analysis, revealed that baculovirus-expressed rat p62 contains 5-6 mol of N-acetylglucosamine/mol of p62. As observed by circular dichroism, purified p62 expressed in the baculovirus system or in Escherichia coli share essentially the same secondary structure. Purified glycosylated rat p62 will be critical in determining the role of N-acetylglucosamine in both nuclear transport and assembly of the nuclear pore complex.
- Published
- 1995
- Full Text
- View/download PDF
43. Role of different domains in the self-association of rat nucleoporin p62.
- Author
-
Buss F, Kent H, Stewart M, Bailer SM, and Hanover JA
- Subjects
- Amino Acid Sequence, Animals, Circular Dichroism, DNA, Complementary genetics, Escherichia coli genetics, Gene Expression, Macromolecular Substances, Membrane Glycoproteins genetics, Microscopy, Electron, Models, Chemical, Molecular Sequence Data, Molecular Structure, Nuclear Pore Complex Proteins, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments isolation & purification, Protein Conformation, Protein Structure, Secondary, Rats, Recombinant Proteins chemistry, Recombinant Proteins genetics, Solubility, Membrane Glycoproteins chemistry
- Abstract
We have expressed rat nucleoporin p62 cDNA in Escherichia coli to obtain material for structural and self-association studies. Electron microscopy and circular dichroism spectroscopy are consistent with a rod-shaped molecule with an alpha-helical coiled-coil domain at its C terminus and a cross-beta structure at its N terminus, separated by a threonine-rich linker, which has a less-defined secondary structure. Electron microscopy and the solubility properties of fragments produced using thrombin and CNBr digestion indicate that p62 molecules associate to form linear chains and that a small region near the C terminus is an important determinant of assembly. This association may have important consequences for pore structure and function; for example, one way p62 could associate would be to form rings in nuclear pores that could function like barrel hoops.
- Published
- 1994
- Full Text
- View/download PDF
44. Characterization of A 54-kD protein of the inner nuclear membrane: evidence for cell cycle-dependent interaction with the nuclear lamina.
- Author
-
Bailer SM, Eppenberger HM, Griffiths G, and Nigg EA
- Subjects
- Animals, Antibodies, Monoclonal, CDC2 Protein Kinase metabolism, Cell Fractionation, Chick Embryo, Detergents, Fluorescent Antibody Technique, Immunoblotting, Lamins, Membrane Proteins ultrastructure, Microscopy, Immunoelectron, Mitosis, Nuclear Envelope ultrastructure, Nuclear Proteins ultrastructure, Phosphorylation, Receptors, Cell Surface metabolism, Solubility, Cell Cycle, Membrane Proteins metabolism, Nuclear Envelope metabolism, Nuclear Proteins metabolism
- Abstract
Using a mAb (R-7), we have characterized a 54-kD protein of the chicken nuclear envelope. Based on its biochemical properties and subnuclear distribution p54 is likely to be an integral membrane component specific to the inner nuclear membrane. Fractionation experiments indicate that p54 interacts, directly or indirectly, with the nuclear lamina, and analysis of p54 in cultured cells suggests that this interaction is controlled by cell cycle-dependent posttranslational modification, most likely phosphorylation. Modification of p54 results in a slightly reduced electrophoretic mobility, and it converts the protein from a detergent-resistant to a detergent-extractable form. Detergent solubilization of p54 can be induced in vivo by treating isolated nuclei or nuclear envelopes with highly purified cdc2 kinase, one of the most prominent kinases active in mitotic cells. These results suggest that mitotic phosphorylation of p54 might contribute to control nuclear envelope dynamics during mitosis in vivo.
- Published
- 1991
- Full Text
- View/download PDF
45. Cloning and sequencing of cDNA clones encoding chicken lamins A and B1 and comparison of the primary structures of vertebrate A- and B-type lamins.
- Author
-
Peter M, Kitten GT, Lehner CF, Vorburger K, Bailer SM, Maridor G, and Nigg EA
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, DNA, Circular isolation & purification, Lamins, Molecular Sequence Data, Protein Conformation, Restriction Mapping, Chickens genetics, DNA, Circular genetics, Lamin Type B, Nuclear Proteins genetics, Vertebrates genetics
- Abstract
Nuclear lamins are intermediate-filament-type proteins forming a fibrillar meshwork underlying the inner nuclear membrane. The existence of multiple isoforms of lamin proteins in vertebrates is believed to reflect functional specializations during cell division and differentiation. Although biochemical criteria may be used to classify many lamin isoforms into A- and B-type subfamilies, the structural features distinguishing the members of these subfamilies remain to be characterized fully. Here, we report the complete primary structures of chicken lamins A and B1, as they are deduced from cloned cDNAs; in the accompanying paper we present the complete sequence of lamin B2, a second avian B-type lamin. Comparisons of the chicken lamin sequences with each other and with those of other lamins allow us to establish structural features that are common to members of both subfamilies. Conversely, multiple sequence alignments make it possible to identify a number of structural motifs that clearly differentiate B-type lamins from A-type lamins. With this information at hand, we attempt to correlate different biochemical properties of A- and B-type lamins with the presence or absence of specific sequence motifs.
- Published
- 1989
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.