Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 418(2): 307-22, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27339293

ABSTRACT

RacGAP1 is one of the two components of the centralspindlin complex essential for orchestrating cytokinesis in all animal cells. We report here that the early arrest mutant ogre is a maternal and zygotic loss of function mutation in the zebrafish homolog of racgap1. Like the other model organisms in which racgap1 is mutated, cells in the mutant stop dividing. In vivo cell recordings reveal that gradual loss of wild-type RacGAP1 leads progressively from a failure of abscission, then to cleavage furrow ingression, and finally complete absence of furrow formation. Despite the lack of cytokinesis, gross patterning occurs overtly normally in ogre mutants and cells continue to cycle slowly, some even attaining four or eight nuclei. Many multinucleate cells differentiate and survive, but the majority of cells enter apoptosis that we demonstrate is due to cumulative rounds of defective cytokinesis. Investigation of the cells that differentiate in the mutant indicate that RacGAP1 is also needed for long-term survival of motoneurons and the cytoskeletal organization of sensory axons. We conclude that while RacGAP1 function is crucial for cytokinesis and its activity at different levels controls different aspects of cytokinesis, these defects have occluded other critical roles of this interesting protein.


Subject(s)
Cytokinesis/physiology , GTPase-Activating Proteins/deficiency , Zebrafish Proteins/deficiency , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Apoptosis/physiology , Cell Cycle/genetics , Cell Cycle/physiology , Cytokinesis/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Microtubules/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation , Neurogenesis/genetics , Neurogenesis/physiology , RNA, Antisense/genetics , Time-Lapse Imaging , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
J Virol ; 89(20): 10247-59, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223647

ABSTRACT

UNLABELLED: The vaccinia virus B1R gene encodes a highly conserved protein kinase that is essential for the poxviral life cycle. As demonstrated in many cell types, B1 plays a critical role during viral DNA replication when it inactivates the cellular host defense effector barrier to autointegration factor (BAF or BANF1). To better understand the role of B1 during infection, we have characterized the growth of a B1-deficient temperature-sensitive mutant virus (Cts2 virus) in U2OS osteosarcoma cells. In contrast to all other cell lines tested to date, we found that in U2OS cells, Cts2 viral DNA replication is unimpaired at the nonpermissive temperature. However, the Cts2 viral yield in these cells was reduced more than 10-fold, thus indicating that B1 is required at another stage of the vaccinia virus life cycle. Our results further suggest that the host defense function of endogenous BAF may be absent in U2OS cells but can be recovered through either overexpression of BAF or fusion of U2OS cells with mouse cells in which the antiviral function of BAF is active. Interestingly, examination of late viral proteins during Cts2 virus infection demonstrated that B1 is required for optimal processing of the L4 protein. Finally, execution point analyses as well as electron microscopy studies uncovered a role for B1 during maturation of poxviral virions. Overall, this work demonstrates that U2OS cells are a novel model system for studying the cell type-specific regulation of BAF and reveals a role for B1 beyond DNA replication during the late stages of the viral life cycle. IMPORTANCE: The most well characterized role for the vaccinia virus B1 kinase is to facilitate viral DNA replication by phosphorylating and inactivating BAF, a cellular host defense responsive to foreign DNA. Additional roles for B1 later in the viral life cycle have been postulated for decades but are difficult to examine directly due to the importance of B1 during DNA replication. Here, we demonstrate that in U2OS cells, a B1 mutant virus escapes the block in DNA replication observed in other cell types and, instead, this mutant virus exhibits impaired late protein accumulation and incomplete maturation of new virions. These data provide the clearest evidence to date that B1 is needed for multiple critical junctures in the poxviral life cycle in a manner that is both dependent on and independent of BAF.


Subject(s)
DNA Replication , DNA, Viral/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Osteoblasts/virology , Protein Kinases/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Animals , Cell Fusion , Cell Line, Tumor , DNA, Viral/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Osteoblasts/immunology , Protein Kinases/immunology , Signal Transduction , Vaccinia virus/immunology , Viral Proteins/immunology , Virion/genetics , Virion/growth & development , Virion/immunology , Virus Replication/genetics
3.
PLoS One ; 9(6): e100511, 2014.
Article in English | MEDLINE | ID: mdl-24945635

ABSTRACT

BAF (Barrier to Autointegration Factor) is a highly conserved DNA binding protein that senses poxviral DNA in the cytoplasm and tightly binds to the viral genome to interfere with DNA replication and transcription. To counteract BAF, a poxviral-encoded protein kinase phosphorylates BAF, which renders BAF unable to bind DNA and allows efficient viral replication to occur. Herein, we examined how BAF phosphorylation is affected by herpes simplex virus type 1 (HSV-1) infection and tested the ability of BAF to interfere with HSV-1 productive infection. Interestingly, we found that BAF phosphorylation decreases markedly following HSV-1 infection. To determine whether dephosphorylated BAF impacts HSV-1 productive infection, we employed cell lines stably expressing a constitutively unphosphorylated form of BAF (BAF-MAAAQ) and cells overexpressing wild type (wt) BAF for comparison. Although HSV-1 production in cells overexpressing wtBAF was similar to that in cells expressing no additional BAF, viral growth was reduced approximately 80% in the presence of BAF-MAAAQ. Experiments were also performed to determine the mechanism of the antiviral activity of BAF with the following results. BAF-MAAAQ was localized to the nucleus, whereas wtBAF was dispersed throughout cells prior to infection. Following infection, wtBAF becomes dephosphorylated and relocalized to the nucleus. Additionally, BAF was associated with the HSV-1 genome during infection, with BAF-MAAAQ associated to a greater extent than wtBAF. Importantly, unphosphorylated BAF inhibited both viral DNA replication and gene expression. For example, expression of two regulatory proteins, ICP0 and VP16, were substantially reduced in cells expressing BAF-MAAAQ. However, other viral genes were not dramatically affected suggesting that expression of certain viral genes can be differentially regulated by unphosphorylated BAF. Collectively, these results suggest that BAF can act in a phosphorylation-regulated manner to impair HSV-1 transcription and/or DNA replication, which is similar to the antiviral activity of BAF during vaccinia infection.


Subject(s)
DNA Replication , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/metabolism , DNA/metabolism , DNA-Binding Proteins/chemistry , Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Proteins/chemistry , Phosphorylation , Protein Binding , Protein Transport , Subcellular Fractions/metabolism , Viral Proteins/metabolism , Virus Replication
4.
J Virol ; 88(10): 5342-55, 2014 May.
Article in English | MEDLINE | ID: mdl-24600006

ABSTRACT

UNLABELLED: Barrier-to-autointegration factor (BAF) is a DNA binding protein with multiple cellular functions, including the ability to act as a potent defense against vaccinia virus infection. This antiviral function involves BAF's ability to condense double-stranded DNA and subsequently prevent viral DNA replication. In recent years, it has become increasingly evident that dynamic phosphorylation involving the vaccinia virus B1 kinase and cellular enzymes is likely a key regulator of multiple BAF functions; however, the precise mechanisms are poorly understood. Here we analyzed how phosphorylation impacts BAF's DNA binding, subcellular localization, dimerization, and antipoxviral activity through the characterization of BAF phosphomimetic and unphosphorylatable mutants. Our studies demonstrate that increased phosphorylation enhances BAF's mobilization from the nucleus to the cytosol, while dephosphorylation restricts BAF to the nucleus. Phosphorylation also impairs both BAF's dimerization and its DNA binding activity. Furthermore, our studies of BAF's antiviral activity revealed that hyperphosphorylated BAF is unable to suppress viral DNA replication or virus production. Interestingly, the unphosphorylatable BAF mutant, which is capable of binding DNA but localizes predominantly to the nucleus, was also incapable of suppressing viral replication. Thus, both DNA binding and localization are important determinants of BAF's antiviral function. Finally, our examination of how phosphatases are involved in regulating BAF revealed that PP2A dephosphorylates BAF during vaccinia infection, thus counterbalancing the activity of the B1 kinase. Altogether, these data demonstrate that phosphoregulation of BAF by viral and cellular enzymes modulates this protein at multiple molecular levels, thus determining its effectiveness as an antiviral factor and likely other functions as well. IMPORTANCE: The barrier-to-autointegration factor (BAF) contributes to cellular genomic integrity in multiple ways, the best characterized of which are as a host defense against cytoplasmic DNA and as a regulator of mitotic nuclear reassembly. Although dynamic phosphorylation involving both viral and cellular enzymes is likely a key regulator of multiple BAF functions, the precise mechanisms involved are poorly understood. Here we demonstrate that phosphorylation coordinately regulates BAF's DNA binding, subcellular localization, dimerization, and antipoxviral activity. Overall, our findings provide new insights into how phosphoregulation of BAF modulates this protein at multiple levels and governs its effectiveness as an antiviral factor against foreign DNA.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Vaccinia virus/immunology , Amino Acid Substitution , Animals , Cell Line , Cell Nucleus/chemistry , Chlorocebus aethiops , Cytosol/chemistry , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins/genetics , Phosphorylation , Protein Binding
5.
Virology ; 444(1-2): 363-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23891157

ABSTRACT

Barrier to autointegration factor (BAF/BANF1) is a cellular DNA-binding protein found in the nucleus and cytoplasm. Cytoplasmic BAF binds to foreign DNA and can act as a defense against vaccinia DNA replication. To evade BAF, vaccinia expresses the B1 kinase, which phosphorylates BAF and blocks its ability to bind DNA. Interestingly, B1 is also needed for viral intermediate gene expression via an unknown mechanism. Therefore, we evaluated the impact of B1-BAF signaling on vaccinia transcription. Strikingly, the decrease in vaccinia transcription caused by loss of B1 can be rescued by depletion of BAF. The repressive action of BAF is greatest on a viral promoter, and is more modest when non-vaccinia promoters are employed, which suggests BAF acts in a gene specific manner. These studies expand our understanding of the role of the B1 kinase during infection and provide the first evidence that BAF is a defense against viral gene expression.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Host-Pathogen Interactions , Immune Evasion , Nuclear Proteins/antagonists & inhibitors , Transcription, Genetic , Vaccinia virus/physiology , Viral Proteins/metabolism , Virus Replication , Animals , Cell Line , DNA-Binding Proteins/immunology , Humans , Nuclear Proteins/immunology , Vaccinia virus/immunology
6.
J Virol ; 85(22): 11588-600, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21880762

ABSTRACT

The barrier to autointegration factor (BAF) is an essential cellular protein with functions in mitotic nuclear reassembly, retroviral preintegration complex stability, and transcriptional regulation. Molecular properties of BAF include the ability to bind double-stranded DNA in a sequence-independent manner, homodimerize, and bind proteins containing a LEM domain. These capabilities allow BAF to compact DNA and assemble higher-order nucleoprotein complexes, the nature of which is poorly understood. Recently, it was revealed that BAF also acts as a potent host defense against poxviral DNA replication in the cytoplasm. Here, we extend these observations by examining the molecular mechanism through which BAF acts as a host defense against vaccinia virus replication and cytoplasmic DNA in general. Interestingly, BAF rapidly relocalizes to transfected DNA from a variety of sources, demonstrating that BAF's activity as a host defense factor is not limited to poxviral infection. BAF's relocalization to cytoplasmic foreign DNA is highly dependent upon its DNA binding and dimerization properties but does not appear to require its LEM domain binding activity. However, the LEM domain protein emerin is recruited to cytoplasmic DNA in a BAF-dependent manner during both transfection and vaccinia virus infection. Finally, we demonstrate that the DNA binding and dimerization capabilities of BAF are essential for its function as an antipoxviral effector, while the presence of emerin is not required. Together, these data provide further mechanistic insight into which of BAF's molecular properties are employed by cells to impair the replication of poxviruses or respond to foreign DNA in general.


Subject(s)
DNA-Binding Proteins/immunology , Host-Pathogen Interactions , Nuclear Proteins/immunology , Vaccinia virus/immunology , Virus Integration/immunology , Animals , Cell Line , Humans , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Protein Binding , Protein Multimerization
SELECTION OF CITATIONS
SEARCH DETAIL
...