Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36093836

ABSTRACT

Intracellular mature viruses (IMVs) are the first and most abundant infectious form of vaccinia virus to assemble during its replication cycle. IMVs can undergo microtubule-based motility, but their directionality and the motor involved in their transport remain unknown. Here, we demonstrate that IMVs, like intracellular enveloped viruses (IEVs), the second form of vaccinia that are wrapped in Golgi-derived membranes, recruit kinesin-1 and undergo anterograde transport. In vitro reconstitution of virion transport in infected cell extracts revealed that IMVs and IEVs move toward microtubule plus ends with respective velocities of 0.66 and 0.56 µm/s. Quantitative imaging established that IMVs and IEVs recruit an average of 139 and 320 kinesin-1 motor complexes, respectively. In the absence of kinesin-1, there was a near-complete loss of in vitro motility and reduction in the intracellular spread of both types of virions. Our observations demonstrate that kinesin-1 transports two morphologically distinct forms of vaccinia. Reconstitution of vaccinia-based microtubule motility in vitro provides a new model to elucidate how motor number and regulation impacts transport of a bona fide kinesin-1 cargo.


Subject(s)
Kinesins , Vaccinia , Cell Extracts , Humans , Microtubules/metabolism , Vaccinia/metabolism , Vaccinia virus , Virion/physiology
2.
Nat Commun ; 13(1): 668, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115540

ABSTRACT

DENR and MCTS1 have been identified as oncogenes in several different tumor entities. The heterodimeric DENR·MCTS1 protein complex promotes translation of mRNAs containing upstream Open Reading Frames (uORFs). We show here that DENR is phosphorylated on Serine 73 by Cyclin B/CDK1 and Cyclin A/CDK2 at the onset of mitosis, and then dephosphorylated as cells exit mitosis. Phosphorylation of Ser73 promotes mitotic stability of DENR protein and prevents its cleavage at Asp26. This leads to enhanced translation of mRNAs involved in mitosis. Indeed, we find that roughly 40% of all mRNAs with elevated translation in mitosis are DENR targets. In the absence of DENR or of Ser73 phosphorylation, cells display elevated levels of aberrant mitoses and cell death. This provides a mechanism how the cell cycle regulates translation of a subset of mitotically relevant mRNAs during mitosis.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin A/metabolism , Cyclin B/metabolism , Cyclin-Dependent Kinase 2/metabolism , Eukaryotic Initiation Factors/metabolism , Blotting, Western , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/genetics , Cell Line, Tumor , Cyclin A/genetics , Cyclin B/genetics , Cyclin-Dependent Kinase 2/genetics , Eukaryotic Initiation Factors/genetics , HeLa Cells , Humans , MCF-7 Cells , Mitosis/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Open Reading Frames/genetics , Phosphorylation , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine/genetics , Serine/metabolism
3.
PLoS Biol ; 16(6): e2005160, 2018 06.
Article in English | MEDLINE | ID: mdl-29889857

ABSTRACT

The succession of molecular events leading to eukaryotic translation reinitiation-whereby ribosomes terminate translation of a short open reading frame (ORF), resume scanning, and then translate a second ORF on the same mRNA-is not well understood. Density-regulated reinitiation and release factor (DENR) and multiple copies in T-cell lymphoma-1 (MCTS1) are implicated in promoting translation reinitiation both in vitro in translation extracts and in vivo. We present here the crystal structure of MCTS1 bound to a fragment of DENR. Based on this structure, we identify and experimentally validate that DENR residues Glu42, Tyr43, and Tyr46 are important for MCTS1 binding and that MCTS1 residue Phe104 is important for tRNA binding. Mutation of these residues reveals that DENR-MCTS1 dimerization and tRNA binding are both necessary for DENR and MCTS1 to promote translation reinitiation in human cells. These findings thereby link individual residues of DENR and MCTS1 to specific molecular functions of the complex. Since DENR-MCTS1 can bind tRNA in the absence of the ribosome, this suggests the DENR-MCTS1 complex could recruit tRNA to the ribosome during reinitiation analogously to the eukaryotic initiation factor 2 (eIF2) complex in cap-dependent translation.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Eukaryotic Initiation Factors/chemistry , Eukaryotic Initiation Factors/metabolism , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , RNA, Transfer/metabolism , Amino Acid Substitution , Cell Cycle Proteins/genetics , Crystallography, X-Ray , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factors/genetics , HeLa Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Oncogene Proteins/genetics , Open Reading Frames , Peptide Chain Initiation, Translational , Protein Interaction Domains and Motifs , Protein Multimerization , RNA, Transfer/genetics , Ribosomes/metabolism
4.
Sci Rep ; 7(1): 3722, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623304

ABSTRACT

The non-canonical initiation factors DENR and MCTS1 have been linked to cancer and autism. We recently showed in Drosophila that DENR and MCTS1 regulate translation re-initiation on transcripts containing upstream Open Reading Frames (uORFs) with strong Kozak sequences (stuORFs). Due to the medical relevance of DENR and MCTS1, it is worthwhile identifying the transcripts in human cells that depend on DENR and MCTS1 for their translation. We show here that in humans, as in Drosophila, transcripts with short stuORFs require DENR and MCTS1 for their optimal expression. In contrast to Drosophila, however, the dependence on stuORF length in human cells is very strong, so that only transcripts with very short stuORFs coding for 1 amino acid are dependent on DENR and MCTS1. This identifies circa 100 genes as putative DENR and MCTS1 translational targets. These genes are enriched for neuronal genes and G protein-coupled receptors. The identification of DENR and MCTS1 target transcripts will serve as a basis for future studies aimed at understanding the mechanistic involvement of DENR and MCTS1 in cancer and autism.


Subject(s)
Cell Cycle Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Profiling , Oncogene Proteins/metabolism , Open Reading Frames , Protein Biosynthesis , RNA, Messenger/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism
5.
Cell Rep ; 15(10): 2251-2265, 2016 06 07.
Article in English | MEDLINE | ID: mdl-27239039

ABSTRACT

Disruptions to neuronal mRNA translation are hypothesized to underlie human neurodevelopmental syndromes. Notably, the mRNA translation re-initiation factor DENR is a regulator of eukaryotic translation and cell growth, but its mammalian functions are unknown. Here, we report that Denr influences the migration of murine cerebral cortical neurons in vivo with its binding partner Mcts1, whereas perturbations to Denr impair the long-term positioning, dendritic arborization, and dendritic spine characteristics of postnatal projection neurons. We characterized de novo missense mutations in DENR (p.C37Y and p.P121L) detected in two unrelated human subjects diagnosed with brain developmental disorder to find that each variant impairs the function of DENR in mRNA translation re-initiation and disrupts the migration and terminal branching of cortical neurons in different ways. Thus, our findings link human brain disorders to impaired mRNA translation re-initiation through perturbations in DENR (OMIM: 604550) function in neurons.


Subject(s)
Eukaryotic Initiation Factors/genetics , Mutation/genetics , Nervous System Diseases/congenital , Nervous System Diseases/genetics , Neurogenesis/genetics , Neurons/metabolism , Peptide Chain Initiation, Translational/genetics , Animals , Cell Differentiation , Cell Movement , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Gene Knockdown Techniques , Humans , Mice, Inbred C57BL , Mutant Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Nature ; 512(7513): 208-212, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25043021

ABSTRACT

During cap-dependent eukaryotic translation initiation, ribosomes scan messenger RNA from the 5' end to the first AUG start codon with favourable sequence context. For many mRNAs this AUG belongs to a short upstream open reading frame (uORF), and translation of the main downstream ORF requires re-initiation, an incompletely understood process. Re-initiation is thought to involve the same factors as standard initiation. It is unknown whether any factors specifically affect translation re-initiation without affecting standard cap-dependent translation. Here we uncover the non-canonical initiation factors density regulated protein (DENR) and multiple copies in T-cell lymphoma-1 (MCT-1; also called MCTS1 in humans) as the first selective regulators of eukaryotic re-initiation. mRNAs containing upstream ORFs with strong Kozak sequences selectively require DENR-MCT-1 for their proper translation, yielding a novel class of mRNAs that can be co-regulated and that is enriched for regulatory proteins such as oncogenic kinases. Collectively, our data reveal that cells have a previously unappreciated translational control system with a key role in supporting proliferation and tissue growth.


Subject(s)
Drosophila Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation/genetics , Protein Biosynthesis/genetics , Animals , Cell Proliferation , Cells, Cultured , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eukaryotic Initiation Factors/genetics , Open Reading Frames , Signal Transduction
7.
Cell Host Microbe ; 10(3): 210-23, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21925109

ABSTRACT

Many viruses deliver their genomes into the host cell nucleus for replication. However, the size restrictions of the nuclear pore complex (NPC), which regulates the passage of proteins, nucleic acids, and solutes through the nuclear envelope, require virus capsid uncoating before viral DNA can access the nucleus. We report a microtubule motor kinesin-1-mediated and NPC-supported mechanism of adenovirus uncoating. The capsid binds to the NPC filament protein Nup214 and kinesin-1 light-chain Klc1/2. The nucleoporin Nup358, which is bound to Nup214/Nup88, interacts with the kinesin-1 heavy-chain Kif5c to indirectly link the capsid to the kinesin motor. Kinesin-1 disrupts capsids docked at Nup214, which compromises the NPC and dislocates nucleoporins and capsid fragments into the cytoplasm. NPC disruption increases nuclear envelope permeability as indicated by the nuclear influx of large cytoplasmic dextran polymers. Thus, kinesin-1 uncoats viral DNA and compromises NPC integrity, allowing viral genomes nuclear access to promote infection.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae/physiology , Capsid/metabolism , Kinesins/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Virus Assembly , Adenoviridae/genetics , Adenoviridae Infections/virology , Cell Line , HeLa Cells , Humans , Kinesins/genetics , Nuclear Pore/genetics , Nuclear Pore Complex Proteins/genetics , Protein Binding
8.
PLoS One ; 4(7): e6305, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19609361

ABSTRACT

Akt, an essential component of the insulin pathway, is a potent inducer of tissue growth. One of Akt's phosphorylation targets is Tsc2, an inhibitor of the anabolic kinase TOR. This could account for part of Akt's growth promoting activity. Although phosphorylation of Tsc2 by Akt does occur in vivo, and under certain circumstances can lead to reduced Tsc2 activity, the functional significance of this event is unclear since flies lacking Akt phosphorylation sites on Tsc2 are viable and normal in size and growth rate. Since Drosophila Tsc1, the obligate partner of Tsc2, has an Akt phosphorylation motif that is not conserved in mammals, we investigate here whether Akt redundantly phosphorylates the Tsc complex on Tsc1 and Tsc2. We provide evidence that Akt phosphorylates Tsc1 at Ser533. We show that flies lacking Akt phosphorylation sites on Tsc1 alone, or on both Tsc1 and Tsc2 concurrently, are viable and normal in size. This shows that phosphorylation of the Tsc1/2 complex by Akt is not required for Akt to activate TORC1 and to promote tissue growth in Drosophila.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/growth & development , Proto-Oncogene Proteins c-akt/metabolism , Animals , Base Sequence , DNA Primers , Phosphorylation , Polymerase Chain Reaction
9.
Nature ; 458(7234): 87-91, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19262673

ABSTRACT

Understanding cell motility will require detailed knowledge not only of the localization of signalling networks regulating actin polymerization, but also of their dynamics. Unfortunately, many signalling networks are not amenable to such analysis, as they are frequently transient and dispersed. By contrast, the signalling pathways used by pathogens undergoing actin-based motility are highly localized and operate in a constitutive fashion. Taking advantage of this, we have analysed the dynamics of neuronal Wiskott-Aldrich syndrome protein (N-WASP), WASP-interacting protein (WIP), GRB2 and NCK, which are required to stimulate actin-related protein (ARP)2/3-complex-dependent actin-based motility of vaccinia virus, using fluorescence recovery after photobleaching. Here we show that all four proteins are rapidly exchanging, albeit at different rates, and that the turnover of N-WASP depends on its ability to stimulate ARP2/3-complex-mediated actin polymerization. Conversely, disruption of the interaction of N-WASP with GRB2 and/or the barbed ends of actin filaments increases its exchange rate and results in a faster rate of virus movement. We suggest that the exchange rate of N-WASP controls the rate of ARP2/3-complex-dependent actin-based motility by regulating the extent of actin polymerization by antagonizing filament capping.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actins/chemistry , Adaptor Proteins, Signal Transducing , Biopolymers/chemistry , Biopolymers/metabolism , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Vaccinia virus/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
10.
Cell Host Microbe ; 1(3): 227-40, 2007 May 17.
Article in English | MEDLINE | ID: mdl-18005701

ABSTRACT

Prior to being released from the infected cell, intracellular enveloped vaccinia virus particles are transported from their perinuclear assembly site to the plasma membrane along microtubules by the motor kinesin-1. After fusion with the plasma membrane, stimulation of actin tails beneath extracellular virus particles acts to enhance cell-to-cell virus spread. However, we lack molecular understanding of events that occur at the cell periphery just before and during the liberation of virus particles. Using live cell imaging, we show that virus particles move in the cell cortex, independently of actin tail formation. These cortical movements and the subsequent release of virus particles, which are both actin dependent, require F11L-mediated inhibition of RhoA-mDia signaling. We suggest that the exit of vaccinia virus from infected cells has strong parallels to exocytosis, as it is dependent on the assembly and organization of actin in the cell cortex.


Subject(s)
Actins/metabolism , Vaccinia virus/physiology , Viral Proteins/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , Exocytosis/physiology , HeLa Cells , Humans
11.
Traffic ; 7(10): 1352-67, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16899087

ABSTRACT

Poxviruses, such as vaccinia virus (VV), replicate their DNA in endoplasmic-reticulum-enclosed cytoplasmic sites. Here, we compare the dynamics of the VV replication sites with those of the attenuated strain, modified VV Ankara (MVA). By live-cell imaging, small, early replication sites of both viruses undergo motility typical of microtubule (MT)-motor-mediated movement. Over time, growing replication sites of VV collect around the nucleus in a MT-dependent fashion, whereas those of MVA remain mostly scattered in the cytoplasm. Surprisingly, blocking the dynein function does not impair the perinuclear accumulation of large VV replication sites. Live-cell imaging demonstrates that in contrast to small replication sites, large sites do not display MT-motor-mediated motility. Instead, VV infection induces cellular contractility that facilitates the collection of growing replication sites around the nucleus. In a subset of cells (30-40%), this VV-induced contractility is alternated by phases of directed cell migration, suggesting that the two processes may be linked. The MVA-infected cells do not display contractility or cell migration, supporting the idea that these cellular activities facilitate the efficient accumulation of the VV replication sites around the nucleus. We propose that the recently described cytoskeletal rearrangements induced by VV are a prerequisite for the observed cell contractility and migration activities that apparently contribute to the organization of the complex cytoplasmic life cycle of VV.


Subject(s)
Cell Movement/physiology , Cell Nucleus/virology , Cytoplasm/virology , Vaccinia virus/physiology , Virus Replication , Animals , Cell Line , Dynactin Complex , Dyneins/genetics , Dyneins/metabolism , Humans , Microscopy, Video , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Motor Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
12.
Cell Microbiol ; 8(3): 427-37, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16469055

ABSTRACT

The assembly of the intracellular mature virus (IMV) of vaccinia virus (VV), the prototype member of the poxviridae, is poorly understood and controversial. We have previously proposed that the IMV is composed of a continuous double-membraned cisterna derived from the smooth ER, whereby the genome-containing core is enwrapped by a part of this cisterna. In the present study we characterize a mutant virus in which the synthesis of the major core protein A10L can be conditionally expressed. Without A10L, IMVs are not made; immature viruses (IVs) and regularly stacked membrane structures that contain viral DNA, accumulate instead. By immunolabelling of thawed cryo-sections these stacks contain most of the viral core proteins and low levels of viral membrane proteins. Importantly, the stacked membranes could be labelled with antibodies to an ER marker protein, implying that they are derived from this cellular compartment. By electron tomography (ET) on semi-thin cryo-sections we show that the membranes of the stacks are continuous with the membranes of the IVs. Direct continuities with ER cisternae, to which the stacks are tightly apposed, were, however, not unequivocally seen. Finally, ET revealed how the IV membranes separated to become two-membrane profiles. Taken together, this study shows that VV core proteins and the viral DNA can coassemble onto ER-derived membranes that are continuous with the membranes of the IVs.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Mutation , Vaccinia virus/metabolism , Viral Core Proteins/metabolism , Virion/metabolism , Virus Assembly , Animals , Cell Line , DNA, Viral/metabolism , Humans , Microscopy, Electron , Tomography , Vaccinia virus/genetics , Vaccinia virus/ultrastructure , Viral Core Proteins/genetics
13.
Science ; 311(5759): 377-81, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16424340

ABSTRACT

RhoA signaling plays a critical role in many cellular processes, including cell migration. Here we show that the vaccinia F11L protein interacts directly with RhoA, inhibiting its signaling by blocking the interaction with its downstream effectors Rho-associated kinase (ROCK) and mDia. RNA interference-mediated depletion of F11L during infection resulted in an absence of vaccinia-induced cell motility and inhibition of viral morphogenesis. Disruption of the RhoA binding site in F11L, which resembles that of ROCK, led to an identical phenotype. Thus, inhibition of RhoA signaling is required for both vaccinia morphogenesis and virus-induced cell motility.


Subject(s)
Cell Movement , Signal Transduction , Vaccinia virus/physiology , Viral Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Animals , Cell Line , Cytoskeletal Proteins , Enzyme Inhibitors/pharmacology , Genes, Viral , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Morphogenesis , Phosphoproteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering , Recombinant Fusion Proteins/metabolism , Transfection , Vaccinia virus/genetics , Vaccinia virus/growth & development , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Assembly , rho-Associated Kinases
14.
Mol Biol Cell ; 16(6): 2822-35, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15800065

ABSTRACT

Vaccinia virus (VV), the prototype member of the Poxviridae, a family of large DNA viruses, carries out DNA replication in specialized cytoplasmic sites that are enclosed by the rough endoplasmic reticulum (ER). We show that the VV gene product of A40R is quantitatively modified by SUMO-1, which is required for its localization to the ER-enclosed replication sites. Expression of A40R lacking SUMO-1 induced the formation of rod-shaped cytoplasmic aggregates. The latter likely consisted of polymers of nonsumoylated protein, because unmodified A40R interacted with itself, but not with the SUMO-1-conjugated protein. Using a bacterial sumoylation system, we furthermore show that unmodified A40R is mostly insoluble, whereas the modified form is completely soluble. By electron microscopy, the A40R rods seen in cells were associated with the cytosolic side of the ER and induced the apposition of several ER cisternae. A40R is the first example of a poxvirus protein to acquire SUMO-1. Its quantitative SUMO-1 modification is required for its proper localization to the viral "mini-nuclei" and prevents its self-association. The ability of the nonsumoylated A40R to bring ER membranes close together could suggest a role in the fusion of ER cisternae when these coalesce to enclose the VV replication sites.


Subject(s)
Membrane Glycoproteins/metabolism , SUMO-1 Protein/metabolism , Vaccinia virus/metabolism , Vaccinia virus/physiology , Viral Nonstructural Proteins/metabolism , Blotting, Western , DNA, Viral/genetics , DNA, Viral/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Lysine/chemistry , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/ultrastructure , Molecular Weight , Mutagenesis, Site-Directed , Precipitin Tests , Protein Binding , Protein Biosynthesis , SUMO-1 Protein/chemistry , Vaccinia virus/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/ultrastructure , Virus Assembly , Virus Replication
15.
J Virol ; 77(10): 6014-28, 2003 May.
Article in English | MEDLINE | ID: mdl-12719593

ABSTRACT

The vaccinia virus (VV) I3L gene product is a single-stranded DNA-binding protein made early in infection that localizes to the cytoplasmic sites of viral DNA replication (S. C. Rochester and P. Traktman, J. Virol. 72:2917-2926, 1998). Surprisingly, when replication was blocked, the protein localized to distinct cytoplasmic spots (A. Domi and G. Beaud, J. Gen. Virol. 81:1231-1235, 2000). Here these I3L-positive spots were characterized in more detail. By using an anti-I3L peptide antibody we confirmed that the protein localized to the cytoplasmic sites of viral DNA replication by both immunofluorescence and electron microscopy (EM). Before replication had started or when replication was inhibited with hydroxyurea or cytosine arabinoside, I3L localized to distinct cytoplasmic punctate structures of homogeneous size. We show that these structures are not incoming cores or cytoplasmic sites of VV early mRNA accumulation. Instead, morphological and quantitative data indicate that they are specialized sites where the parental DNA accumulates after its release from incoming viral cores. By EM, these sites appeared as complex, electron-dense structures that were intimately associated with the cellular endoplasmic reticulum (ER). By double labeling of cryosections we show that they contain DNA and a viral early protein, the gene product of E8R. Since E8R is a membrane protein that is able to bind to DNA, the localization of this protein to the I3L puncta suggests that they are composed of membranes. The results are discussed in relation to our previous data showing that the process of viral DNA replication also occurs in close association with the ER.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/ultrastructure , Vaccinia virus/metabolism , Viral Proteins/metabolism , Virus Replication , Cytoplasm/metabolism , Cytoplasm/ultrastructure , DNA, Viral/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Microscopy, Electron , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Proteins/genetics
16.
J Virol ; 76(19): 9773-86, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12208956

ABSTRACT

Vaccinia virus (VV), a member of the poxvirus family, is unique among most other DNA viruses in that both transcription and DNA replication occur in the cytoplasm of the host cell. It was recently shown by electron microscopy (EM) that soon after viral DNA synthesis is initiated in HeLa cells, the replication sites become enwrapped by the membrane of the endoplasmic reticulum (ER). In the same study, a novel VV membrane protein, the E8R gene product, that may play a role in the ER wrapping process was identified (N. Tolonen, L. Doglio, S. Schleich, and J. Krijnse Locker, Mol. Biol. Cell 12:2031-2046, 2001). In the present study, the gene product of E8R was characterized both biochemically and morphologically. We show that E8R is made predominantly early in infection but is packaged into the virion. On two-dimensional gel electrophoresis, the protein appeared as a single spot throughout the VV life cycle; however, in the assembled virion, the protein underwent several modifications which resulted in a change in its molecular weight and its isoelectric point. EM of labeled cryosections of infected HeLa cells showed that the protein localized to the ER and to membranes located on one side of the Golgi complex as early as 1 h postinfection. Late in infection, E8R was additionally associated with membranes of immature virions and with intracellular mature viruses. Although E8R is predominantly associated with membranes, we show that the protein is associated with viral cores; the protein is present in cores made with NP-40-dithiothreitol as well as in incoming cores, the result of the viral entry process, early in infection. Finally, we show that E8R can be phosphorylated in vitro by the viral kinase F10L. It is able to bind DNA in vitro, and this binding may be modulated by phosphorylation by F10L. A putative role of the E8R gene product throughout the VV life cycle is discussed.


Subject(s)
Membrane Proteins/analysis , Vaccinia virus/chemistry , Viral Proteins/analysis , Virus Assembly , Cell Membrane/chemistry , DNA/metabolism , HeLa Cells , Humans , Membrane Proteins/physiology , Microscopy, Electron , Microscopy, Fluorescence , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Viral Proteins/physiology
17.
J Virol ; 76(16): 8318-34, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12134037

ABSTRACT

It has previously been shown that upon infection of HeLa cells with modified vaccinia virus Ankara (MVA), assembly is blocked at a late stage of infection and immature virions (IVs) accumulate (G. Sutter and B. Moss, Proc. Natl. Acad. Sci. USA 89:10847-10851, 1992). In the present study the morphogenesis of MVA in HeLa cells was studied in more detail and compared to that under two conditions that permit the production of infectious particles: infection of HeLa cells with the WR strain of vaccinia virus (VV) and infection of BHK cells with MVA. Using several quantitative and qualitative assays, we show that early in infection, MVA in HeLa cells behaves in a manner identical to that under the permissive conditions. By immunofluorescence microscopy (IF) at late times of infection, the labelings for an abundant membrane protein of the intracellular mature virus, p16/A14L, and the viral DNA colocalize under permissive conditions, whereas in HeLa cells infected with MVA these two structures do not colocalize to the same extent. In both permissive and nonpermissive infection, p16-labeled IVs first appear at 5 h postinfection. In HeLa cells infected with MVA, IVs accumulated predominantly outside the DNA regions, whereas under permissive conditions they were associated with the viral DNA. At 4 h 30 min, the earliest time at which p16 is detected, the p16 labeling was found predominantly in a small number of distinct puncta by IF, which were distinct from the sites of DNA in both permissive and nonpermissive infection. By electron microscopy, no crescents or IVs were found at this time, and the p16-labeled structures were found to consist of membrane-rich vesicles that were in continuity with the cellular endoplasmic reticulum. Over the next 30 min of infection, a large number of p16-labeled crescents and IVs appeared abruptly under both permissive and nonpermissive conditions. Under permissive conditions, these IVs were in close association with the sites of DNA, and a significant amount of these IVs engulfed the viral DNA. In contrast, under nonpermissive conditions, the IVs and DNA were mostly in separate locations and relatively few IVs acquired DNA. Our data show that in HeLa cells MVA forms normal DNA replication sites and normal viral precursor membranes but the transport between these two structures is inhibited.


Subject(s)
Vaccinia virus/growth & development , Animals , Cell Line , Cricetinae , DNA Replication , DNA, Viral/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Microscopy, Immunoelectron , Vaccinia virus/physiology , Vaccinia virus/ultrastructure , Viral Envelope Proteins/metabolism , Virus Assembly
18.
J Virol ; 76(10): 5167-83, 2002 May.
Article in English | MEDLINE | ID: mdl-11967332

ABSTRACT

Virus assembly, a late event in the life cycle of vaccinia virus (VV), is preceded by a number of steps that all occur in the cytoplasm of the infected host cell: virion entry, delivery of the viral core into the cytoplasm, and transcription from these cores of early mRNAs, followed by the process of DNA replication. In the present study the quantitative and structural relationships between these distinct steps of VV morphogenesis were investigated. We show that viral RNA and DNA synthesis increases linearly with increasing amounts of incoming cores. Moreover, at multiplicities of infection that result in 10 to 40 cores per cell, an approximately 1:1 ratio between cores and sites of DNA replication exists, suggesting that each core is infectious. We have shown previously that VV early mRNAs collect in distinct granular structures that recruit components of the host cell translation machinery. Strikingly, these structures appeared to form some distance away from intracellular cores (M. Mallardo, S. Schleich, and J. Krijnse Locker, Mol. Biol. Cell 12:3875-3891, 2001). In the present study the intracellular locations of the sites of early mRNA accumulation and those of the subsequent process of DNA replication were compared. We show that these are distinct structures that have different intracellular locations. Finally, we study the fate of the parental DNA after core uncoating. By electron microscopy, cores were found close to membranes of the endoplasmic reticulum (ER) and the parental DNA, once it had left the core, appeared to associate preferentially with the cytosolic side of those membranes. Since we have previously shown that the process of DNA replication occurs in an ER-enclosed cytosolic "subcompartment" (N. Tolonen, L. Doglio, S. Schleich, and J. Krijnse Locker, Mol. Biol. Cell 12:2031-2046, 2001), the present data suggest that the parental DNA is released into the cytosol and associates with the same membranes where DNA replication is subsequently initiated. The combined data are discussed with respect to the cytosolic organization of VV morphogenesis.


Subject(s)
DNA Replication , DNA, Viral/metabolism , RNA, Messenger/metabolism , RNA, Viral/metabolism , Vaccinia virus/physiology , Viral Core Proteins/metabolism , Cytoplasm/metabolism , Cytoplasm/ultrastructure , HeLa Cells , Humans , Vaccinia virus/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...