Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.446
1.
J Virol ; 98(3): e0151523, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38323812

Adeno-associated virus (AAV) requires co-infection with helper virus for efficient replication. We previously reported that Human Bocavirus 1 (HBoV1) genes, including NP1, NS2, and BocaSR, were critical for AAV2 replication. Here, we first demonstrate the essential roles of the NP1 protein in AAV2 DNA replication and protein expression. We show that NP1 binds to single-strand DNA (ssDNA) at least 30 nucleotides (nt) in length in a sequence-independent manner. Furthermore, NP1 colocalized with the BrdU-labeled AAV2 DNA replication center, and the loss of the ssDNA-binding ability of NP1 by site-directed mutation completely abolished AAV2 DNA replication. We used affinity-tagged NP1 protein to identify host cellular proteins associated with NP1 in cells cotransfected with the HBoV1 helper genes and AAV2 duplex genome. Of the identified proteins, we demonstrate that NP1 directly binds to the DBD-F domain of the RPA70 subunit with a high affinity through the residues 101-121. By reconstituting the heterotrimer protein RPA in vitro using gel filtration, we demonstrate that NP1 physically associates with RPA to form a heterologous complex characterized by typical fast-on/fast-off kinetics. Following a dominant-negative strategy, we found that NP1-RPA complex mainly plays a role in expressing AAV2 capsid protein by enhancing the transcriptional activity of the p40 promoter. Our study revealed a novel mechanism by which HBoV1 NP1 protein supports AAV2 DNA replication and capsid protein expression through its ssDNA-binding ability and direct interaction with RPA, respectively.IMPORTANCERecombinant adeno-associated virus (rAAV) vectors have been extensively used in clinical gene therapy strategies. However, a limitation of these gene therapy strategies is the efficient production of the required vectors, as AAV alone is replication-deficient in the host cells. HBoV1 provides the simplest AAV2 helper genes consisting of NP1, NS2, and BocaSR. An important question regarding the helper function of HBoV1 is whether it provides any direct function that supports AAV2 DNA replication and protein expression. Also of interest is how HBoV1 interplays with potential host factors to constitute a permissive environment for AAV2 replication. Our studies revealed that the multifunctional protein NP1 plays important roles in AAV2 DNA replication via its sequence-independent ssDNA-binding ability and in regulating AAV2 capsid protein expression by physically interacting with host protein RPA. Our findings present theoretical guidance for the future application of the HBoV1 helper genes in the rAAV vector production.


Capsid Proteins , Capsid , DNA, Single-Stranded , DNA, Viral , DNA-Binding Proteins , Dependovirus , Human bocavirus , Viral Proteins , Humans , Capsid/metabolism , Capsid Proteins/biosynthesis , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Dependovirus/genetics , Dependovirus/growth & development , Dependovirus/metabolism , DNA, Single-Stranded/biosynthesis , DNA, Single-Stranded/metabolism , DNA, Viral/biosynthesis , DNA, Viral/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Human bocavirus/genetics , Human bocavirus/metabolism , Kinetics , Mutagenesis, Site-Directed , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Domains , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
2.
J Mol Biol ; 434(7): 167507, 2022 04 15.
Article En | MEDLINE | ID: mdl-35217069

In retroviruses, strand displacement DNA-dependent DNA polymerization catalyzed by the viral reverse transcriptase (RT) is required to synthesize double-stranded proviral DNA. In addition, strand displacement during RNA-dependent DNA synthesis is critical to generate high-quality cDNA for use in molecular biology and biotechnology. In this work, we show that the loss of RNase H activity due to inactivating mutations in HIV-1 RT (e.g. D443N or E478Q) has no significant effect on strand displacement while copying DNA templates, but has a large impact on DNA polymerization in reactions carried out with RNA templates. Similar effects were observed with ß-thujaplicinol and other RNase H active site inhibitors, including compounds with dual activity (i.e., characterized also as inhibitors of HIV-1 integrase and/or the RT DNA polymerase). Among them, dual inhibitors of HIV-1 RT DNA polymerase/RNase H activities, containing a 7-hydroxy-6-nitro-2H-chromen-2-one pharmacophore were found to be very potent and effective strand displacement inhibitors in RNA-dependent DNA polymerization reactions. These findings might be helpful in the development of transcriptomics technologies to obtain more uniform read coverages when copying long RNAs and for the construction of more representative libraries avoiding biases towards 5' and 3' ends, while providing valuable information for the development of novel antiretroviral agents.


DNA, Viral , HIV Reverse Transcriptase , Ribonuclease H, Human Immunodeficiency Virus , Anti-Retroviral Agents/chemistry , Anti-Retroviral Agents/pharmacology , DNA, Viral/biosynthesis , Drug Development , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Reverse Transcriptase Inhibitors/pharmacology , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Ribonuclease H, Human Immunodeficiency Virus/metabolism , Tropolone/analogs & derivatives , Tropolone/pharmacology
3.
Viruses ; 14(2)2022 02 10.
Article En | MEDLINE | ID: mdl-35215961

A modified SELEX (Systematic Evolution of Ligands by Exponential Enrichment) pr,otocol (referred to as PT SELEX) was used to select primer-template (P/T) sequences that bound to the vaccinia virus polymerase catalytic subunit (E9) with enhanced affinity. A single selected P/T sequence (referred to as E9-R5-12) bound in physiological salt conditions with an apparent equilibrium dissociation constant (KD,app) of 93 ± 7 nM. The dissociation rate constant (koff) and binding half-life (t1/2) for E9-R5-12 were 0.083 ± 0.019 min-1 and 8.6 ± 2.0 min, respectively. The values indicated a several-fold greater binding ability compared to controls, which bound too weakly to be accurately measured under the conditions employed. Loop-back DNA constructs with 3'-recessed termini derived from E9-R5-12 also showed enhanced binding when the hybrid region was 21 nucleotides or more. Although the sequence of E9-R5-12 matched perfectly over a 12-base-pair segment in the coding region of the virus B20 protein, there was no clear indication that this sequence plays any role in vaccinia virus biology, or a clear reason why it promotes stronger binding to E9. In addition to E9, five other polymerases (HIV-1, Moloney murine leukemia virus, and avian myeloblastosis virus reverse transcriptases (RTs), and Taq and Klenow DNA polymerases) have demonstrated strong sequence binding preferences for P/Ts and, in those cases, there was biological or potential evolutionary relevance. For the HIV-1 RT, sequence preferences were used to aid crystallization and study viral inhibitors. The results suggest that several other DNA polymerases may have P/T sequence preferences that could potentially be exploited in various protocols.


DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase/metabolism , Vaccinia virus/enzymology , Viral Proteins/metabolism , Avian Myeloblastosis Virus/genetics , Avian Myeloblastosis Virus/metabolism , Base Sequence , DNA-Directed DNA Polymerase/genetics , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/metabolism , Protein Binding , SELEX Aptamer Technique , Vaccinia virus/genetics , Viral Proteins/genetics , Virus Replication
5.
J Virol ; 96(4): e0184021, 2022 02 23.
Article En | MEDLINE | ID: mdl-34878919

Human bocavirus 1 (HBoV1), an autonomous human parvovirus, causes acute respiratory tract infections in young children. HBoV1 infects well-differentiated (polarized) human airway epithelium cultured at an air-liquid interface (HAE-ALI). HBoV1 expresses a large nonstructural protein, NS1, that is essential for viral DNA replication. HBoV1 infection of polarized human airway epithelial cells induces a DNA damage response (DDR) that is critical to viral DNA replication involving DNA repair with error-free Y-family DNA polymerases. HBoV1 NS1 or the isoform NS1-70 per se induces a DDR. In this study, using the second-generation proximity-dependent biotin identification (BioID2) approach, we identified that Ku70 is associated with the NS1-BioID2 pulldown complex through a direct interaction with NS1. Biolayer interferometry (BLI) assay determined a high binding affinity of NS1 with Ku70, which has an equilibrium dissociation constant (KD) value of 0.16 µM and processes the strongest interaction at the C-terminal domain. The association of Ku70 with NS1 was also revealed during HBoV1 infection of HAE-ALI. Knockdown of Ku70 and overexpression of the C-terminal domain of Ku70 significantly decreased HBoV1 replication in HAE-ALI. Thus, our study provides, for the first time, a direct interaction of parvovirus large nonstructural protein NS1 with Ku70. IMPORTANCE Parvovirus infection induces a DNA damage response (DDR) that plays a pivotal role in viral DNA replication. The DDR includes activation of ATM (ataxia telangiectasia mutated), ATR (ATM- and RAD3-related), and DNA-PKcs (DNA-dependent protein kinase catalytic subunit). The large nonstructural protein (NS1) often plays a role in the induction of DDR; however, how the DDR is induced during parvovirus infection or simply by the NS1 is not well studied. Activation of DNA-PKcs has been shown as one of the key DDR pathways in DNA replication of HBoV1. We identified that HBoV1 NS1 directly interacts with Ku70, but not Ku80, of the Ku70/Ku80 heterodimer at high affinity. This interaction is also important for HBoV1 replication in HAE-ALI. We propose that the interaction of NS1 with Ku70 recruits the Ku70/Ku80 complex to the viral DNA replication center, which activates DNA-PKcs and facilitates viral DNA replication.


Human bocavirus/physiology , Ku Autoantigen/metabolism , Respiratory Mucosa/virology , Viral Nonstructural Proteins/metabolism , Virus Replication , DNA Damage , DNA Replication , DNA, Viral/biosynthesis , Genome, Viral , HEK293 Cells , Human bocavirus/metabolism , Humans , Ku Autoantigen/genetics , Protein Binding , Protein Domains , Respiratory Mucosa/metabolism , Viral Nonstructural Proteins/genetics , Viral Replication Compartments/metabolism
6.
J Gen Virol ; 102(12)2021 12.
Article En | MEDLINE | ID: mdl-34904939

Reverse transcriptases (RTs) are typically assayed using optimized Mg2+ concentrations (~5-10 mM) several-fold higher than physiological cellular free Mg2+ (~0.5 mM). Recent analyses demonstrated that HIV-1, but not Moloney murine leukaemia (MuLV) or avain myeloblastosis (AMV) virus RTs has higher fidelity in low Mg2+. In the current report, lacZα-based α-complementation assays were used to measure the fidelity of several RTs including HIV-1 (subtype B and A/E), several drug-resistant HIV-1 derivatives, HIV-2, and prototype foamy virus (PFV), all which showed higher fidelity using physiological Mg2+, while MuLV and AMV RTs demonstrated equivalent fidelity in low and high Mg2+. In 0.5 mM Mg2+, all RTs demonstrated approximately equal fidelity, except for PFV which showed higher fidelity. A Next Generation Sequencing (NGS) approach that used barcoding to determine mutation profiles was used to examine the types of mutations made by HIV-1 RT (type B) in low (0.5 mM) and high (6 mM) Mg2+ on a lacZα template. Unlike α-complementation assays which are dependent on LacZα activity, the NGS assay scores mutations at all positions and of every type. Consistent with α-complementation assays, a ~four-fold increase in mutations was observed in high Mg2+. These findings help explain why HIV-1 RT displays lower fidelity in vitro (with high Mg2+ concentrations) than other RTs (e.g. MuLV and AMV), yet cellular fidelity for these viruses is comparable. Establishing in vitro conditions that accurately represent RT's activity in cells is pivotal to determining the contribution of RT and other factors to the mutation profile observed with HIV-1.


Magnesium/metabolism , RNA-Directed DNA Polymerase/genetics , Retroviridae/genetics , DNA, Viral/biosynthesis , DNA, Viral/genetics , Drug Resistance, Viral/genetics , Magnesium/analysis , Mutation , Mutation Rate , RNA-Directed DNA Polymerase/metabolism , Retroviridae/classification , Retroviridae/enzymology
7.
Viruses ; 13(11)2021 11 04.
Article En | MEDLINE | ID: mdl-34835025

Pseudorabies virus (PRV) is the causative agent of Aujeszky's disease, which still causes large economic losses for the swine industry. Therefore, it is urgent to find a new strategy to prevent and control PRV infection. Previous studies have proven that guanine (G)-rich DNA or RNA sequences in some other viruses' genomes have the potential to form G-quadruplex (G4), which serve as promising antivirus targets. In this study, we identified two novel G4-forming sequences, OriL-A and OriL-S, which are located at the upstream origin of replication (OriL) in the PRV genome and conserved across 32 PRV strains. Circular dichroism (CD) spectroscopy and a gel electrophoresis assay showed that the two G-rich sequences can fold into parallel G4 structures in vitro. Moreover, fluorescence resonance energy transfer (FRET) melting and a Taq polymerase stop assay indicated that the G4 ligand PhenDC3 has the capacity to bind and stabilize the G4. Notably, the treatment of PRV-infected cells with G4-stabilizer PhenDC3 significantly inhibited PRV DNA replication in host cells but did not affect PRV's attachment and entry. These results not only expand our knowledge about the G4 characteristics in the PRV genome but also suggest that G4 may serve as an innovative therapeutic target against PRV.


Antiviral Agents/pharmacology , G-Quadruplexes , Herpesvirus 1, Suid/genetics , Replication Origin/genetics , Animals , Antiviral Agents/chemistry , Cell Line , DNA Replication/drug effects , DNA, Viral/biosynthesis , DNA, Viral/chemistry , DNA, Viral/drug effects , Fused-Ring Compounds/chemistry , Fused-Ring Compounds/pharmacology , G-Quadruplexes/drug effects , Genome, Viral/drug effects , Genome, Viral/genetics , Herpesvirus 1, Suid/drug effects , Herpesvirus 1, Suid/physiology , Replication Origin/drug effects , Swine , Virus Replication/drug effects
8.
Viruses ; 13(9)2021 09 14.
Article En | MEDLINE | ID: mdl-34578405

Papillomavirus L1 and L2, the major and minor capsid proteins, play significant roles in viral assembly, entry, and propagation. In the current study, we investigate the impact of L1 and L2 on viral life cycle and tumor growth with a newly established mouse papillomavirus (MmuPV1) infection model. MmuPV1 L1 knockout, L2 knockout, and L1 plus L2 knockout mutant genomes (designated as L1ATGko-4m, L2ATGko, and L1-L2ATGko respectively) were generated. The mutants were examined for their ability to generate lesions in athymic nude mice. Viral activities were examined by qPCR, immunohistochemistry (IHC), in situ hybridization (ISH), and transmission electron microscopy (TEM) analyses. We demonstrated that viral DNA replication and tumor growth occurred at both cutaneous and mucosal sites infected with each of the mutants. Infections involving L1ATGko-4m, L2ATGko, and L1-L2ATGko mutant genomes generally resulted in smaller tumor sizes compared to infection with the wild type. The L1 protein was absent in L1ATGko-4m and L1-L2ATGko mutant-treated tissues, even though viral transcripts and E4 protein expression were robust. Therefore, L1 is not essential for MmuPV1-induced tumor growth, and this finding parallels our previous observations in the rabbit papillomavirus model. Very few viral particles were detected in L2ATGko mutant-infected tissues. Interestingly, the localization of L1 in lesions induced by L2ATGko was primarily cytoplasmic rather than nuclear. The findings support the hypothesis that the L2 gene influences the expression, location, transport, and assembly of the L1 protein in vivo.


Capsid Proteins/physiology , Mucous Membrane/virology , Oncogene Proteins, Viral/physiology , Papillomaviridae/physiology , Skin/virology , Animals , Capsid Proteins/genetics , Cell Transformation, Viral , DNA, Viral/biosynthesis , Female , Genome, Viral , Mice , Mice, Nude , Mutation , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Virus Replication
9.
Viruses ; 13(9)2021 09 18.
Article En | MEDLINE | ID: mdl-34578444

The localization of viral nucleic acids in the cell is essential for understanding the infectious cycle. One of the strategies developed for this purpose is the use of nucleotide analogs such as bromodeoxyuridine (BrdU, analog to thymine) or bromouridine (BrU, analog of uridine), which are incorporated into the nucleic acids during replication or transcription. In adenovirus infections, BrdU has been used to localize newly synthesized viral genomes in the nucleus, where it is key to distinguish between host and viral DNA. Here, we describe our experience with methodological variations of BrdU labeling to localize adenovirus genomes in fluorescence and electron microscopy. We illustrate the need to define conditions in which most of the newly synthesized DNA corresponds to the virus and not the host, and the amount of BrdU provided is enough to incorporate to the new DNA molecules without hampering the cell metabolism. We hope that our discussion of problems encountered and solutions implemented will help other researches interested in viral genome localization in infected cells.


Adenoviruses, Human/genetics , Bromodeoxyuridine , DNA, Viral/analysis , Genome, Viral , Microscopy, Electron , Microscopy, Fluorescence , Adenoviruses, Human/physiology , Cell Nucleus/virology , DNA Replication , DNA, Viral/biosynthesis , HEK293 Cells , Humans , Microscopy, Confocal , Staining and Labeling , Virus Replication
10.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article En | MEDLINE | ID: mdl-34244425

Virus infection causes major rearrangements in the subcellular architecture of eukaryotes, but its impact in prokaryotic cells was much less characterized. Here, we show that infection of the bacterium Bacillus subtilis by bacteriophage SPP1 leads to a hijacking of host replication proteins to assemble hybrid viral-bacterial replisomes for SPP1 genome replication. Their biosynthetic activity doubles the cell total DNA content within 15 min. Replisomes operate at several independent locations within a single viral DNA focus positioned asymmetrically in the cell. This large nucleoprotein complex is a self-contained compartment whose boundaries are delimited neither by a membrane nor by a protein cage. Later during infection, SPP1 procapsids localize at the periphery of the viral DNA compartment for genome packaging. The resulting DNA-filled capsids do not remain associated to the DNA transactions compartment. They bind to phage tails to build infectious particles that are stored in warehouse compartments spatially independent from the viral DNA. Free SPP1 structural proteins are recruited to the dynamic phage-induced compartments following an order that recapitulates the viral particle assembly pathway. These findings show that bacteriophages restructure the crowded host cytoplasm to confine at different cellular locations the sequential processes that are essential for their multiplication.


Bacillus subtilis/virology , Cell Compartmentation , Virus Diseases/pathology , Bacillus subtilis/ultrastructure , Bacteriophages/physiology , Bacteriophages/ultrastructure , Capsid/metabolism , DNA Replication , DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase , Host-Pathogen Interactions , Multienzyme Complexes , Time Factors , Virion/metabolism
11.
J Mater Chem B ; 9(28): 5636-5644, 2021 07 21.
Article En | MEDLINE | ID: mdl-34196647

DNA polymerization is of high specificity in vivo. However, its specificity is much lower in vitro, which limits advanced applications of DNA polymerization in ultrasensitive nucleic acid detection. Herein, we report a unique mechanism of single selenium-atom modified dNTP (dNTPαSe) to enhance polymerization specificity. We have found that both dNTPαSe (approximately 660 fold) and Se-DNA (approximately 2.8 fold) have lower binding affinity to DNA polymerase than canonical ones, and the Se-DNA duplex has much lower melting-temperature (Tm) than the corresponding canonical DNA duplex. The reduced affinity and Tm can destabilize the substrate-primer-template-enzyme assembly, thereby largely slowing down the mismatch of DNA polymerization and enhancing the amplification specificity and in turn detection sensitivity. Furthermore, the Se-strategy enables us to develop the selenium enhanced specific isothermal amplification (SEA) for nucleic acid detection with high specificity and sensitivity (up to detection of single-digit copies), allowing convenient detection of clinical HPV and COVID-19 viruses in the low-copy number. Clearly, we have discovered the exciting mechanism for enhancing DNA polymerization accuracy, amplification specificity and detection sensitivity by SEA, up to two orders of magnitude higher.


DNA, Viral/analysis , Human papillomavirus 16/isolation & purification , Phosphates/chemistry , SARS-CoV-2/isolation & purification , Selenium/chemistry , DNA, Viral/biosynthesis , Human papillomavirus 16/metabolism , Humans , Nucleic Acid Amplification Techniques , Polymerization , SARS-CoV-2/metabolism , Temperature
12.
Nat Commun ; 12(1): 2780, 2021 05 13.
Article En | MEDLINE | ID: mdl-33986276

Geminiviruses are causal agents of devastating diseases in crops. Geminiviruses have circular single-stranded (ss) DNA genomes that are replicated in the nucleus of the infected plant cell through double-stranded (ds) DNA intermediates by the plant DNA replication machinery. Which host DNA polymerase mediates geminiviral multiplication, however, has so far remained elusive. Here, we show that subunits of the nuclear replicative DNA polymerases α and δ physically interact with the geminivirus-encoded replication enhancer protein, C3, and that these polymerases are required for viral replication. Our results suggest that, while DNA polymerase α is essential to generate the viral dsDNA intermediate, DNA polymerase δ mediates the synthesis of new copies of the geminiviral ssDNA genome, and that the virus-encoded C3 may act selectively, recruiting DNA polymerase δ over ε to favour productive replication.


DNA Polymerase III/metabolism , DNA Polymerase I/metabolism , DNA Replication/genetics , DNA, Viral/biosynthesis , Geminiviridae/genetics , Virus Replication/genetics , Genome, Viral/genetics , Plants/virology , Protein Subunits/metabolism , Viral Proteins/metabolism
13.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article En | MEDLINE | ID: mdl-33919712

The unfolded protein response (UPR) is an intracellular signaling pathway essential for alleviating the endoplasmic reticulum (ER) stress. To support the productive infection, many viruses are known to use different strategies to manipulate the UPR signaling network. However, it remains largely unclear whether the UPR signaling pathways are modulated in the lytic cycle of Epstein-Barr virus (EBV), a widely distributed human pathogen. Herein, we show that the expression of GRP78, a central UPR regulator, is up-regulated during the EBV lytic cycle. Our data further revealed that knockdown of GRP78 in EBV-infected cell lines did not substantially affect lytic gene expression; however, GRP78 knockdown in these cells markedly reduced the production of virus particles. Importantly, we identified that the early lytic protein BMLF1 is the key regulator critically contributing to the activation of the grp78 gene promoter. Mechanistically, we found that BMLF1 can trigger the proteolytic cleavage and activation of the UPR senor ATF6, which then transcriptionally activates the grp78 promoter through the ER stress response elements. Our findings therefore provide evidence for the connection between the EBV lytic cycle and the UPR, and implicate that the BMLF1-mediated ATF6 activation may play critical roles in EBV lytic replication.


Activating Transcription Factor 6/metabolism , Heat-Shock Proteins/genetics , Phosphoproteins/metabolism , Trans-Activators/metabolism , Up-Regulation , Base Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , DNA, Viral/biosynthesis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Models, Biological , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Signal Transduction , Transcriptional Activation/genetics , Unfolded Protein Response , Up-Regulation/genetics , eIF-2 Kinase/metabolism
14.
Elife ; 102021 04 27.
Article En | MEDLINE | ID: mdl-33904396

HIV-1 replication commences inside the cone-shaped viral capsid, but timing, localization, and mechanism of uncoating are under debate. We adapted a strategy to visualize individual reverse-transcribed HIV-1 cDNA molecules and their association with viral and cellular proteins using fluorescence and correlative-light-and-electron-microscopy (CLEM). We specifically detected HIV-1 cDNA inside nuclei, but not in the cytoplasm. Nuclear cDNA initially co-localized with a fluorescent integrase fusion (IN-FP) and the viral CA (capsid) protein, but cDNA-punctae separated from IN-FP/CA over time. This phenotype was conserved in primary HIV-1 target cells, with nuclear HIV-1 complexes exhibiting strong CA-signals in all cell types. CLEM revealed cone-shaped HIV-1 capsid-like structures and apparently broken capsid-remnants at the position of IN-FP signals and elongated chromatin-like structures in the position of viral cDNA punctae lacking IN-FP. Our data argue for nuclear uncoating by physical disruption rather than cooperative disassembly of the CA-lattice, followed by physical separation from the pre-integration complex.


When viruses infect human cells, they hijack the cell's machinery to produce the proteins they need to replicate. Retroviruses like HIV-1 do this by entering the nucleus and inserting their genetic information into the genome of the infected cell. This requires HIV-1 to convert its genetic material into DNA, which is then released from the protective shell surrounding it (known as the capsid) via a process called uncoating. The nucleus is enclosed within an envelope containing pores that molecules up to a certain size can pass through. Until recently these pores were thought to be smaller than the viral capsid, which led scientists to believe that the HIV-1 genome must shed this coat before penetrating the nucleus. However, recent studies have found evidence for HIV-1 capsid proteins and capsid structures inside the nucleus of some infected cells. This suggests that the capsid may not be removed before nuclear entry or that it may even play a role in helping the virus get inside the nucleus. To investigate this further, Müller et al. attached fluorescent labels to the newly made DNA of HIV-1 and some viral and cellular proteins. Powerful microscopy tools were then used to monitor the uncoating process in various cells that had been infected with the virus. Müller et al. found large amounts of capsid protein inside the nuclei of all the infected cells studied. During the earlier stages of infection, the capsid proteins were mostly associated with viral DNA and the capsid structure appeared largely intact. At later time points, the capsid structure had been broken down and the viral DNA molecules were gradually separating themselves from these remnants. These findings suggest that the HIV-1 capsid helps the virus get inside the nucleus and may protect its genetic material during conversion into DNA until right before integration into the cell's genome. Further experiments studying this process could lead to new therapeutic approaches that target the capsid as a way to prevent or treat HIV-1.


Cell Nucleus/virology , DNA Replication , DNA, Viral/biosynthesis , HIV Infections/virology , HIV-1/growth & development , Virus Internalization , Virus Replication , Virus Uncoating , CD4-Positive T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/virology , Capsid Proteins/metabolism , Cell Nucleus/ultrastructure , DNA, Viral/genetics , DNA, Viral/ultrastructure , HEK293 Cells , HIV Infections/pathology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , HIV-1/ultrastructure , HeLa Cells , Host-Pathogen Interactions , Humans , Macrophages/ultrastructure , Macrophages/virology , Microscopy, Electron , Microscopy, Fluorescence , Time Factors
15.
Science ; 372(6541): 512-516, 2021 04 30.
Article En | MEDLINE | ID: mdl-33926954

DNA modifications vary in form and function but generally do not alter Watson-Crick base pairing. Diaminopurine (Z) is an exception because it completely replaces adenine and forms three hydrogen bonds with thymine in cyanophage S-2L genomic DNA. However, the biosynthesis, prevalence, and importance of Z genomes remain unexplored. Here, we report a multienzyme system that supports Z-genome synthesis. We identified dozens of globally widespread phages harboring such enzymes, and we further verified the Z genome in one of these phages, Acinetobacter phage SH-Ab 15497, by using liquid chromatography with ultraviolet and mass spectrometry. The Z genome endows phages with evolutionary advantages for evading the attack of host restriction enzymes, and the characterization of its biosynthetic pathway enables Z-DNA production on a large scale for a diverse range of applications.


2-Aminopurine/metabolism , Adenylosuccinate Synthase/chemistry , Bacteriophages/chemistry , Bacteriophages/enzymology , DNA, Viral/chemistry , DNA, Z-Form/chemistry , Viral Nonstructural Proteins/chemistry , 2-Aminopurine/chemistry , Adenylosuccinate Lyase/chemistry , Adenylosuccinate Lyase/genetics , Adenylosuccinate Lyase/metabolism , Adenylosuccinate Synthase/genetics , Adenylosuccinate Synthase/metabolism , Bacteriophages/genetics , Base Pairing , Biosynthetic Pathways , DNA, Viral/biosynthesis , DNA, Viral/genetics , DNA, Z-Form/biosynthesis , DNA, Z-Form/genetics , Genome, Viral , Hydrogen Bonding , Protein Domains , Substrate Specificity , Thymine/chemistry , Thymine/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
16.
Science ; 372(6541): 520-524, 2021 04 30.
Article En | MEDLINE | ID: mdl-33926956

Bacteriophage genomes harbor the broadest chemical diversity of nucleobases across all life forms. Certain DNA viruses that infect hosts as diverse as cyanobacteria, proteobacteria, and actinobacteria exhibit wholesale substitution of aminoadenine for adenine, thereby forming three hydrogen bonds with thymine and violating Watson-Crick pairing rules. Aminoadenine-encoded DNA polymerases, homologous to the Klenow fragment of bacterial DNA polymerase I that includes 3'-exonuclease but lacks 5'-exonuclease, were found to preferentially select for aminoadenine instead of adenine in deoxynucleoside triphosphate incorporation templated by thymine. Polymerase genes occur in synteny with genes for a biosynthesis enzyme that produces aminoadenine deoxynucleotides in a wide array of Siphoviridae bacteriophages. Congruent phylogenetic clustering of the polymerases and biosynthesis enzymes suggests that aminoadenine has propagated in DNA alongside adenine since archaic stages of evolution.


2-Aminopurine/analogs & derivatives , DNA Replication , DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase/chemistry , Polymerization , Siphoviridae/chemistry , Siphoviridae/enzymology , Viral Nonstructural Proteins/chemistry , 2-Aminopurine/chemistry , DNA-Directed DNA Polymerase/classification , DNA-Directed DNA Polymerase/genetics , Genome, Viral , Phylogeny , Siphoviridae/genetics , Viral Nonstructural Proteins/classification , Viral Nonstructural Proteins/genetics
17.
FASEB J ; 35(5): e21607, 2021 05.
Article En | MEDLINE | ID: mdl-33908664

Strand displacement DNA synthesis (SDDS) is an essential step in DNA replication. With magnetic tweezers, we investigated SDDS kinetics of wild-type gp90 and its exonuclease-deficient polymerase gp90 exo- at single-molecule level. A novel binding state of gp90 to the fork flap was confirmed prior to SDDS, suggesting an intermediate in the initiation of SDDS. The rate and processivity of SDDS by gp90 exo- or wt-gp90 are increased with force and dNTP concentration. The rate and processivity of exonuclease by wt-gp90 are decreased with force. High GC content decreases SDDS and exonuclease processivity but increases exonuclease rate for wt-gp90. The high force and dNTP concentration and low GC content facilitate the successive SDDS but retard the successive exonuclease for wt-gp90. Furthermore, increasing GC content accelerates the transition from SDDS or exonuclease to exonuclease. This work reveals the kinetics of SDDS in detail and offers a broader cognition on the regulation of various factors on SDDS at single-polymerase level.


Bacteriophages/physiology , DNA Replication , DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase/metabolism , Pseudomonas aeruginosa/virology , Single-Cell Analysis/methods , DNA-Directed DNA Polymerase/genetics , Recombination, Genetic
18.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article En | MEDLINE | ID: mdl-33753499

Hepadnaviruses, with the human hepatitis B virus as prototype, are small, enveloped hepatotropic DNA viruses which replicate by reverse transcription of an RNA intermediate. Replication is initiated by a unique protein-priming mechanism whereby a hydroxy amino acid side chain of the terminal protein (TP) domain of the viral polymerase (P) is extended into a short DNA oligonucleotide, which subsequently serves as primer for first-strand synthesis. A key component in the priming of reverse transcription is the viral RNA element epsilon, which contains the replication origin and serves as a template for DNA primer synthesis. Here, we show that recently discovered non-enveloped fish viruses, termed nackednaviruses [C. Lauber et al., Cell Host Microbe 22, 387-399 (2017)], employ a fundamentally similar replication mechanism despite their huge phylogenetic distance and major differences in genome organization and viral lifestyle. In vitro cross-priming studies revealed that few strategic nucleotide substitutions in epsilon enable site-specific protein priming by heterologous P proteins, demonstrating that epsilon is functionally conserved since the two virus families diverged more than 400 Mya. In addition, other cis elements crucial for the hepadnavirus-typical replication of pregenomic RNA into relaxed circular double-stranded DNA were identified at conserved positions in the nackednavirus genomes. Hence, the replication mode of both hepadnaviruses and nackednaviruses was already established in their Paleozoic common ancestor, making it a truly ancient and evolutionary robust principle of genome replication that is more widespread than previously thought.


DNA Replication , DNA, Viral/biosynthesis , Evolution, Molecular , Hepadnaviridae/physiology , Reverse Transcription , Viral Proteins/metabolism , Virus Replication , Conserved Sequence , Hepadnaviridae/classification , Hepadnaviridae/genetics , Hepatitis B virus/classification , Hepatitis B virus/genetics , Phylogeny , RNA, Viral/genetics , Replication Origin , Viral Proteins/genetics
19.
Antiviral Res ; 189: 105057, 2021 05.
Article En | MEDLINE | ID: mdl-33716051

Emergence of drug resistance and adverse effects often affect the efficacy of nucleoside analogues in the therapy of Herpes simplex type 1 (HSV-1) and type 2 (HSV-2) infections. Host-targeting antivirals could therefore be considered as an alternative or complementary strategy in the management of HSV infections. To contribute to this advancement, here we report on the ability of a new generation inhibitor of a key cellular enzyme of de novo pyrimidine biosynthesis, the dihydroorotate dehydrogenase (DHODH), to inhibit HSV-1 and HSV-2 in vitro replication, with a potency comparable to that of the reference drug acyclovir. Analysis of the HSV replication cycle in MEDS433-treated cells revealed that it prevented the accumulation of viral genomes and reduced late gene expression, thus suggesting an impairment at a stage prior to viral DNA replication consistent with the ability of MEDS433 to inhibit DHODH activity. In fact, the anti-HSV activity of MEDS433 was abrogated by the addition of exogenous uridine or of the product of DHODH, the orotate, thus confirming DHODH as the MEDS433 specific target in HSV-infected cells. A combination of MEDS433 with dipyridamole (DPY), an inhibitor of the pyrimidine salvage pathway, was then observed to be effective in inhibiting HSV replication even in the presence of exogenous uridine, thus mimicking in vivo conditions. Finally, when combined with acyclovir and DPY in checkerboard experiments, MEDS433 exhibited highly synergistic antiviral activity. Taken together, these findings suggest that MEDS433 is a promising candidate as either single agent or in combination regimens with existing direct-acting anti-HSV drugs to develop new strategies for treatment of HSV infections.


Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Virus Replication/drug effects , Acyclovir/pharmacology , Animals , Cell Line, Tumor , Chlorocebus aethiops , DNA Replication/drug effects , DNA, Viral/biosynthesis , Dihydroorotate Dehydrogenase , Drug Synergism , Drug Therapy, Combination , Gene Expression Regulation, Viral/drug effects , Herpes Simplex/virology , Humans , Pyrimidines/biosynthesis , Vero Cells
20.
Sci Rep ; 11(1): 5025, 2021 03 03.
Article En | MEDLINE | ID: mdl-33658544

Bioconversion of organic materials is the foundation of many applications in chemical engineering, microbiology and biochemistry. Herein, we introduce a new methodology to quantitatively determine conversion of biomass in viral infections while simultaneously imaging morphological changes of the host cell. As proof of concept, the viral replication of an unidentified giant DNA virus and the cellular response of an amoebal host are studied using soft X-ray microscopy, titration dilution measurements and thermal gravimetric analysis. We find that virions produced inside the cell are visible from 18 h post infection and their numbers increase gradually to a burst size of 280-660 virions. Due to the large size of the virion and its strong X-ray absorption contrast, we estimate that the burst size corresponds to a conversion of 6-12% of carbonaceous biomass from amoebal host to virus. The occurrence of virion production correlates with the appearance of a possible viral factory and morphological changes in the phagosomes and contractile vacuole complex of the amoeba, whereas the nucleus and nucleolus appear unaffected throughout most of the replication cycle.


Acanthamoeba/virology , DNA Viruses/ultrastructure , DNA, Viral/genetics , Genome, Viral , Giant Viruses/ultrastructure , Virion/ultrastructure , Acanthamoeba/ultrastructure , Biomass , DNA Viruses/genetics , DNA Viruses/growth & development , DNA Viruses/isolation & purification , DNA, Viral/biosynthesis , Giant Viruses/genetics , Giant Viruses/growth & development , Giant Viruses/isolation & purification , Host-Pathogen Interactions/genetics , Phagosomes/ultrastructure , Phagosomes/virology , Soil Microbiology , Thermogravimetry , Vacuoles/ultrastructure , Vacuoles/virology , Virion/genetics , Virion/growth & development , Virus Replication , X-Ray Microtomography
...