Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Nutr Biochem ; 56: 48-54, 2018 06.
Article in English | MEDLINE | ID: mdl-29454998

ABSTRACT

The mitochondrial proteome comprises 1000 to 1500 proteins, in addition to proteins for which the mitochondrial localization is uncertain. About 800 diseases have been linked with mutations in mitochondrial proteins. We devised a cell survival assay for assessing the mitochondrial localization in a high-throughput format. This protocol allows us to assess the mitochondrial localization of proteins and their mutants, and to identify drugs and nutrients that modulate the mitochondrial targeting of proteins. The assay works equally well for proteins directed to the outer mitochondrial membrane, inner mitochondrial membrane mitochondrial and mitochondrial matrix, as demonstrated by assessing the mitochondrial targeting of the following proteins: carnitine palmitoyl transferase 1 (consensus sequence and R123C mutant), acetyl-CoA carboxylase 2, uncoupling protein 1 and holocarboxylase synthetase. Our screen may be useful for linking the mitochondrial proteome with rare diseases and for devising drug- and nutrition-based strategies for altering the mitochondrial targeting of proteins.


Subject(s)
Cell Death , Cytological Techniques , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteome , Cell Survival , HEK293 Cells , Humans , Microscopy, Confocal , Mutation , Proteomics , Xenobiotics/chemistry
2.
J Biol Chem ; 292(48): 19814-19825, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28972144

ABSTRACT

The pyridine-based multimerization selective HIV-1 integrase (IN) inhibitors (MINIs) are a distinct subclass of allosteric IN inhibitors. MINIs potently inhibit HIV-1 replication during virion maturation by inducing hyper- or aberrant IN multimerization but are largely ineffective during the early steps of viral replication. Here, we investigated the mechanism for the evolution of a triple IN substitution (T124N/V165I/T174I) that emerges in cell culture with a representative MINI, KF116. We show that HIV-1 NL4-3(IN T124N/V165I/T174I) confers marked (>2000-fold) resistance to KF116. Two IN substitutions (T124N/T174I) directly weaken inhibitor binding at the dimer interface of the catalytic core domain but at the same time markedly impair HIV-1 replication capacity. Unexpectedly, T124N/T174I IN substitutions inhibited proteolytic processing of HIV-1 polyproteins Gag and Gag-Pol, resulting in immature virions. Strikingly, the addition of the third IN substitution (V165I) restored polyprotein processing, virus particle maturation, and significant levels of replication capacity. These results reveal an unanticipated role of IN for polyprotein proteolytic processing during virion morphogenesis. The complex evolutionary pathway for the emergence of resistant viruses, which includes the need for the compensatory V165I IN substitution, highlights a relatively high genetic barrier exerted by MINI KF116. Additionally, we have solved the X-ray structure of the drug-resistant catalytic core domain protein, which provides means for rational development of second-generation MINIs.


Subject(s)
Gene Products, gag/metabolism , Gene Products, pol/metabolism , HIV Integrase Inhibitors/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , Pyridines/chemistry , Crystallography, X-Ray , Drug Resistance, Viral , HEK293 Cells , HIV Integrase Inhibitors/chemistry , HIV-1/metabolism , Humans , Proteolysis , Surface Plasmon Resonance
3.
J Virol ; 91(9)2017 05 01.
Article in English | MEDLINE | ID: mdl-28202766

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) capsid protein is an attractive therapeutic target, owing to its multifunctionality in virus replication and the high fitness cost of amino acid substitutions in capsids to HIV-1 infectivity. To date, small-molecule inhibitors have been identified that inhibit HIV-1 capsid assembly and/or impair its function in target cells. Here, we describe the mechanism of action of the previously reported capsid-targeting HIV-1 inhibitor, Boehringer-Ingelheim compound 1 (C1). We show that C1 acts during HIV-1 maturation to prevent assembly of a mature viral capsid. However, unlike the maturation inhibitor bevirimat, C1 did not significantly affect the kinetics or fidelity of Gag processing. HIV-1 particles produced in the presence of C1 contained unstable capsids that lacked associated electron density and exhibited impairments in early postentry stages of infection, most notably reverse transcription. C1 inhibited assembly of recombinant HIV-1 CA in vitro and induced aberrant cross-links in mutant HIV-1 particles capable of spontaneous intersubunit disulfide bonds at the interhexamer interface in the capsid lattice. Resistance to C1 was conferred by a single amino acid substitution within the compound-binding site in the N-terminal domain of the CA protein. Our results demonstrate that the binding site for C1 represents a new pharmacological vulnerability in the capsid assembly stage of the HIV-1 life cycle.IMPORTANCE The HIV-1 capsid protein is an attractive but unexploited target for clinical drug development. Prior studies have identified HIV-1 capsid-targeting compounds that display different mechanisms of action, which in part reflects the requirement for capsid function at both the efferent and afferent phases of viral replication. Here, we show that one such compound, compound 1, interferes with assembly of the conical viral capsid during virion maturation and results in perturbations at a specific protein-protein interface in the capsid lattice. We also identify and characterize a mutation in the capsid protein that confers resistance to the inhibitor. This study reveals a novel mechanism by which a capsid-targeting small molecule can inhibit HIV-1 replication.


Subject(s)
Anti-HIV Agents/pharmacology , Benzimidazoles/pharmacology , Capsid Proteins/metabolism , Capsid/metabolism , HIV-1/growth & development , Virus Assembly/drug effects , Amino Acid Substitution/genetics , Binding Sites/genetics , Capsid Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Viral/genetics , HEK293 Cells , HIV-1/genetics , Humans , Protein Structure, Tertiary , Reverse Transcription/drug effects
4.
J Virol ; 90(8): 3806-3809, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26842478

ABSTRACT

Intrinsic defenses targeting foreign DNA are one facet of the cellular armament tasked with protecting host genomic integrity. The DNA binding protein BAF (barrier to autointegration factor) contributes to multiple aspects of genome maintenance and intercepts retrovirus, poxvirus, and herpesvirus genomes during infection. In this gem, we discuss the unique position BAF occupies at the virus-host interface and how both viral and cellular mechanisms may regulate its capacity to act as a pro- or antiviral effector targeting viral DNA.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Genome, Human , Nuclear Proteins/metabolism , Virus Integration , Humans
5.
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
6.
Curr Opin Cell Biol ; 34: 61-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26072104

ABSTRACT

The Barrier to Autointegration Factor (BAF or BANF1) is an abundant, highly conserved DNA binding protein. BAF is involved in multiple pathways including mitosis, nuclear assembly, viral infection, chromatin and gene regulation and the DNA damage response. BAF is also essential for early development in metazoans and relevant to human physiology; BANF1 mutations cause a progeroid syndrome, placing BAF within the laminopathy disease spectrum. This review summarizes previous knowledge about BAF in the context of recent discoveries about its protein partners, posttranslational regulation, dynamic subcellular localizations and roles in disease, innate immunity, transposable elements and genome integrity.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Genome , Immunity, Innate , Nuclear Proteins/metabolism , Progeria/genetics , Progeria/metabolism , Stress, Physiological , Animals , DNA-Binding Proteins/genetics , Humans , Nuclear Proteins/genetics
7.
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
8.
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
9.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...