Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(8): 1481-1494, 2021 08.
Article in English | MEDLINE | ID: mdl-34045231

ABSTRACT

Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.


Subject(s)
Apoptosis , Cytokines/metabolism , Immunity , Neuroendocrine Tumors/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Vaccinia virus/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/virology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Tumor Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 114(51): E11001-E11009, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203656

ABSTRACT

The long-standing inability to visualize connections between poxvirus membranes and cellular organelles has led to uncertainty regarding the origin of the viral membrane. Indeed, there has been speculation that viral membranes form de novo in cytoplasmic factories. Another possibility, that the connections are too short-lived to be captured by microscopy during a normal infection, motivated us to identify and characterize virus mutants that are arrested in assembly. Five conserved vaccinia virus proteins, referred to as Viral Membrane Assembly Proteins (VMAPs), that are necessary for formation of immature virions were found. Transmission electron microscopy studies of two VMAP deletion mutants had suggested retention of connections between viral membranes and the endoplasmic reticulum (ER). We now analyzed cells infected with each of the five VMAP deletion mutants by electron tomography, which is necessary to validate membrane continuity, in addition to conventional transmission electron microscopy. In all cases, connections between the ER and viral membranes were demonstrated by 3D reconstructions, supporting a role for the VMAPs in creating and/or stabilizing membrane scissions. Furthermore, coexpression of the viral reticulon-like transmembrane protein A17 and the capsid-like scaffold protein D13 was sufficient to form similar ER-associated viral structures in the absence of other major virion proteins. Determination of the mechanism of ER disruption during a normal VACV infection and the likely participation of both viral and cell proteins in this process may provide important insights into membrane dynamics.


Subject(s)
Endoplasmic Reticulum/metabolism , Imaging, Three-Dimensional , Vaccinia virus/physiology , Viral Matrix Proteins/metabolism , Virus Assembly , Capsid/metabolism , Capsid/ultrastructure , Electron Microscope Tomography , Endoplasmic Reticulum/ultrastructure , Mutation , Sequence Deletion , Vaccinia virus/ultrastructure , Viral Matrix Proteins/genetics , Virion
3.
Mol Ther ; 23(9): 1532-40, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26073886

ABSTRACT

Fifteen patients with treatment-refractory colorectal cancer were enrolled on a phase 1b study of Pexa-Vec (pexastimogene devacirepvec; JX-594), an oncolytic and immunotherapeutic vaccinia designed to selectively replicate in cancer cells. Pexa-Vec was administered intravenously every 14 days, at dose levels of 1 × 10(6), 1 × 10(7), or 3 × 10(7) plaque-forming units (pfu)/kg. The primary endpoint was to determine the maximum tolerated dose. Secondary endpoints were pharmacokinetics and pharmacodynamics as well as antitumor activity. Patients were heavily pretreated (mean 4.5 lines of therapy). All patients received at least two Pexa-Vec doses (median = 4; range = 2-4). No dose-limiting toxicities were reported, and the maximum tolerated dose was not reached. The most common adverse events were grade 1/2 flu-like symptoms, generally lasting <24 hours. During the first and last cycles, genome pharmacokinetics were unchanged. Infectious pfu could be detected in plasma up to 2 hours after cycle 1 and up to 30 minutes after cycle 4 (when antivaccinia antibody titers are known to have peaked). Ten patients (67%) had radiographically stable disease. Given the acceptable safety profile of multiple intravenous Pexa-Vec infusions in patients with treatment-refractory colorectal cancer, further trials evaluating efficacy of intravenous Pexa-Vec, as monotherapy or in combination with chemotherapeutic agents, is warranted in this patient population.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunotherapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Administration, Intravenous , Adult , Aged , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Combined Modality Therapy , Cytokines/blood , Drug Administration Schedule , Female , Genetic Vectors/administration & dosage , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Leukocyte Count , Male , Middle Aged , Neoplasm Metastasis , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Skin Diseases/etiology , Skin Diseases/pathology , Treatment Outcome
4.
Traffic ; 16(7): 787-95, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25761760

ABSTRACT

The split green fluorescent protein (GFP) system was adapted for investigation of the topology of ER-associated proteins. A 215-amino acid fragment of GFP (S1-10) was expressed in the cytoplasm as a free protein or fused to the N-terminus of calnexin and in the ER as an intraluminal protein or fused to the C-terminus of calnexin. A 16-amino acid fragment of GFP (S11) was fused to the N- or C-terminus of the target protein. Fluorescence occurred when both GFP fragments were in the same intracellular compartment. After validation with the cellular proteins PDI and tapasin, we investigated two vaccinia virus proteins (L2 and A30.5) of unknown topology that localize to the ER and are required for assembly of the viral membrane. Our results indicated that the N- and C-termini of L2 faced the cytoplasmic and luminal sides of the ER, respectively. In contrast both the N- and C-termini of A30.5 faced the cytoplasm. The system offers advantages for quickly determining the topology of intracellular proteins: the S11 tag is similar in length to commonly used epitope tags; multiple options are available for detecting fluorescence in live or fixed cells; transfection protocols are adaptable to numerous expression systems and can enable high throughput applications.


Subject(s)
Calnexin/metabolism , Endoplasmic Reticulum/metabolism , Viral Proteins/metabolism , Animals , Calnexin/genetics , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Protein Transport , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vaccinia virus/genetics , Viral Proteins/genetics
5.
Virology ; 452-453: 59-66, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606683

ABSTRACT

Some orthopoxviruses including cowpox virus embed virus particles in dense bodies, comprised of the A-type inclusion (ATI) protein, which may provide long-term environmental protection. This strategy could be beneficial if the host population is sparse or spread is inefficient or indirect. However, the formation of ATI may be neutral or disadvantageous for orthopoxviruses that rely on direct respiratory spread. Disrupted ATI open reading frames in orthopoxviruses such as variola virus, the agent of smallpox, and monkeypox virus suggests that loss of this feature provided positive selection. To test this hypothesis, we constructed cowpox virus mutants with deletion of the ATI gene or another gene required for embedding virions. The ATI deletion mutant caused greater weight loss and higher replication in the respiratory tract than control viruses, supporting our hypothesis. Deletion of the gene for embedding virions had a lesser effect, possibly due to known additional functions of the encoded protein.


Subject(s)
Biological Evolution , Cowpox virus/physiology , Cowpox/virology , Gene Deletion , Orthopoxvirus/genetics , Viral Proteins/genetics , Virus Replication , Animals , Cowpox virus/genetics , Humans , Inclusion Bodies, Viral/virology , Mice , Mice, Inbred BALB C , Orthopoxvirus/physiology , Viral Proteins/metabolism
6.
Virology ; 447(1-2): 213-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24210117

ABSTRACT

Vaccinia virus contains ~200 genes classified temporally as early, intermediate or late. We analyzed 53 intermediate promoters to determine whether any have dual late promoter activity. Our strategy involved (i) construction of a cell line that stably expressed the three late transcription factors, (ii) infection with a vaccinia virus mutant that expresses RNA polymerase but neither intermediate nor late transcription factors, and (iii) transfection with plasmids containing a luciferase reporter regulated by an intermediate promoter. After confirming the specificity of the system for late promoters, we found that many intermediate promoters had late promoter activity, the strength of which correlated with a TAAAT at the initiator site and T-content from positions -12 to -8 of the coding strand. In contrast, intermediate promoter activity correlated with the A-content from positions -22 to -14. The sequence correlations were confirmed by altering the specificities of strict intermediate and late promoters.


Subject(s)
Gene Expression Regulation, Viral , Promoter Regions, Genetic , Vaccinia virus/genetics , Animals , Cell Line , DNA Mutational Analysis , Rabbits
7.
J Virol ; 87(22): 12313-26, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24027302

ABSTRACT

Crescents consisting of a single lipoprotein membrane with an external protein scaffold comprise the initial structural elements of poxvirus morphogenesis. Crescents enlarge to form spherical immature virions, which enclose viroplasm consisting of proteins destined to form the cores of mature virions. Previous studies suggest that the L2 protein participates in the recruitment of endoplasmic reticulum (ER)-derived membranes to form immature virions within assembly sites of cytoplasmic factories. Here we show that L2 interacts with the previously uncharacterized 42-amino-acid A30.5 protein. An open reading frame similar in size to the one encoding A30.5 is at the same genome location in representatives of all chordopoxvirus genera. A30.5 has a putative transmembrane domain and colocalized with markers of the endoplasmic reticulum and with L2. By constructing a complementing cell line expressing A30.5, we isolated a deletion mutant virus that exhibits a defect in morphogenesis in normal cells. Large electron-dense cytoplasmic inclusions and clusters of scaffold protein-coated membranes that resemble crescents and immature virions devoid of viroplasm were seen in place of normal structures. Crescent-shaped membranes were continuous with the endoplasmic reticulum membrane and oriented with the convex scaffold protein-coated side facing the lumen, while clusters of completed spherical immature-virion-like forms were trapped within the expanded lumen. Immature-virion-like structures were more abundant in infected RK-13 cells than in BS-C-1 or HeLa cells, in which cytoplasmic inclusions were decorated with scaffold protein-coated membrane arcs. We suggest that the outer surface of the poxvirus virion is derived from the luminal side of the ER membrane.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Vaccinia virus/physiology , Vaccinia/metabolism , Viral Proteins/metabolism , Virion/physiology , Virus Assembly , Virus Replication , Amino Acid Sequence , Animals , Blotting, Western , Cells, Cultured , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasm/virology , HeLa Cells , Humans , Immunoprecipitation , Kidney/cytology , Kidney/metabolism , Kidney/virology , Mass Spectrometry , Molecular Sequence Data , Rabbits , Sequence Homology, Amino Acid , Vaccinia/virology
8.
J Virol ; 87(18): 10195-206, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23864611

ABSTRACT

The apparent de novo formation of viral membranes within cytoplasmic factories is a mysterious, poorly understood first step in poxvirus morphogenesis. Genetic studies identified several viral proteins essential for membrane formation and the assembly of immature virus particles. Their repression results in abortive replication with the accumulation of dense masses of viroplasm. In the present study, we further characterized one of these proteins, A11, and investigated its association with cellular and viral membranes under normal and abortive replication conditions. We discovered that A11 colocalized in cytoplasmic factories with the endoplasmic reticulum (ER) and L2, another viral protein required for morphogenesis. Confocal microscopy and subcellular fractionation indicated that A11 was not membrane associated in uninfected cells, whereas L2 still colocalized with the ER. Cell-free transcription and translation experiments indicated that both A11 and L2 are tail-anchored proteins that associate posttranslationally with membranes and likely require specific cytoplasmic targeting chaperones. Transmission electron microscopy indicated that A11, like L2, associated with crescent membranes and immature virions during normal infection and with vesicles and tubules near masses of dense viroplasm during abortive infection in the absence of the A17 or A14 protein component of viral membranes. When the synthesis of A11 was repressed, "empty" immature-virion-like structures formed in addition to masses of viroplasm. The immature-virion-like structures were labeled with antibodies to A17 and to the D13 scaffold protein and were closely associated with calnexin-labeled ER. These studies revealed similarities and differences between A11 and L2, both of which may be involved in the recruitment of the ER for virus assembly.


Subject(s)
Endoplasmic Reticulum/virology , Vaccinia virus/physiology , Viral Proteins/metabolism , Virus Assembly , Virus Replication , Cell Line , Humans , Microscopy, Electron, Transmission , Protein Binding , Virion/ultrastructure
9.
J Virol ; 87(3): 1861-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192873

ABSTRACT

Assembly of the poxvirus immature virion (IV) membrane is a poorly understood event that occurs within the cytoplasm. At least eight viral proteins participate in formation of the viral membrane. Of these, A14, A17, and D13 are structural components whereas A6, A11, F10, H7, and L2 participate in membrane biogenesis. L2, the object of this study, is conserved in all chordopoxviruses, expressed early in infection, and associated with the endoplasmic reticulum (ER) throughout the cell and at the edges of crescent-shaped IV precursors. Previous studies with an inducible L2 mutant revealed abortive formation of the crescent membrane. However, possible low-level L2 synthesis under nonpermissive conditions led to ambiguity in interpretation. Here, we constructed a cell line that expresses L2, which allowed the creation of an L2-deletion mutant. In noncomplementing cells, replication was aborted prior to formation of mature virions and two types of aberrant structures were recognized. One consisted of short crescents, at the surface of dense masses of viroplasm, which were labeled with antibodies to the A11, A14, A17, and D13 proteins. The other structure consisted of "empty" IV-like membranes, also labeled with antibodies to the viral proteins, which appeared to be derived from adjacent calnexin-containing ER. A subset of 25 proteins examined, exemplified by components of the entry-fusion complex, were greatly diminished in amount. The primary role of L2 may be to recruit ER and modulate its transformation to viral membranes in juxtaposition with the viroplasm, simultaneously preventing the degradation of viral proteins dependent on viral membranes for stability.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Vaccinia virus/physiology , Virus Assembly , Cell Line , Humans , Sequence Deletion , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
10.
J Virol ; 85(23): 12594-604, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937647

ABSTRACT

The unfolded protein response (UPR) is a cellular mechanism that is triggered in order to cope with the stress caused by the accumulation of misfolded proteins in the endoplasmic reticulum (ER). This response is initiated by the endoribonuclease inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), and PKR-like ER kinase, which increase the expression of the genes involved in the folding and degradation processes and decrease the protein input into the ER by inhibiting translation. It has been shown that viruses both induce and manipulate the UPR in order to protect the host cells from an ER stress-mediated death, thus permitting the translation of viral proteins and the efficient replication of the virus. To understand the cellular events that occur during the rotavirus replication cycle, we examined the activation of the three UPR arms following infection, using luciferase reporters driven by promoters of the ER stress-responsive genes and real-time reverse transcription-PCR to determine the levels of the stress-induced mRNAs. Our findings indicated that during rotavirus infection two of the three arms of the UPR (IRE1 and ATF6) become activated; however, these pathways are interrupted at the translational level by the general inhibition of protein synthesis caused by NSP3. This response seems to be triggered by more than one viral protein synthesized during the replication of the virus, but not by the viral double-stranded RNA (dsRNA), since cells transfected with psoralen-inactivated virions, or with naked viral dsRNA, did not induce UPR.


Subject(s)
Endoplasmic Reticulum/metabolism , Rotavirus Infections/metabolism , Rotavirus Infections/virology , Rotavirus/pathogenicity , Unfolded Protein Response/physiology , Viral Nonstructural Proteins/metabolism , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/genetics , Endoribonucleases/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Heat-Shock Proteins/genetics , Humans , Macaca mulatta , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmids , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Double-Stranded/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Rotavirus Infections/pathology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Nonstructural Proteins/genetics
11.
J Virol ; 85(23): 12431-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21917978

ABSTRACT

The initial step in poxvirus morphogenesis, the formation of crescent membranes, occurs within cytoplasmic factories. L2 is one of several vaccinia virus proteins known to be necessary for formation of crescents and the only one synthesized early in infection. Virus replication was unaffected when the L2R open reading frame was replaced by L2R containing an N-terminal epitope tag while retaining the original promoter. L2 colocalized with the endoplasmic reticulum (ER) protein calnexin throughout the cytoplasm of infected and transfected cells. Topological studies indicated that the N terminus of L2 is exposed to the cytoplasm with the hydrophobic C terminus anchored in the ER. Using immunogold labeling and electron microscopy, L2 was detected in tubular membranes outside factories and inside factories near crescents and close to the edge or rim of crescents; a similar labeling pattern was found for the ER luminal protein disulfide isomerase (PDI). The phenotype of L2 conditional lethal mutants and the localization of L2 suggest that it participates in elongation of crescents by the addition of ER membrane to the growing edge. Small amounts of L2 and PDI were detected within immature and mature virions, perhaps trapped during assembly. The repression of L2, as well as A11 and A17, two other proteins that are required for viral crescent formation, profoundly decreased the stability of a subset of viral membrane proteins including those comprising the entry-fusion complex. To avoid degradation, these unstable membrane proteins may need to directly insert into the viral membrane or be rapidly shunted there from the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Vaccinia virus/pathogenicity , Vaccinia/virology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Virion/growth & development , Virus Replication , Blotting, Western , Cytoplasm/metabolism , Cytoplasm/virology , HeLa Cells , Humans , Protein Stability , Vaccinia/metabolism , Virion/metabolism , Virion/pathogenicity , Virus Assembly
12.
J Virol ; 85(6): 2504-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21228235

ABSTRACT

Morphogenesis of vaccinia virus begins with the appearance of crescent-shaped membrane precursors of immature virions in cytoplasmic factories. During the initial characterization of the product of the L2R reading frame, we discovered that it plays an important role in crescent formation. The L2 protein was expressed early in infection and was associated with the detergent-soluble membrane fraction of mature virions, consistent with two potential membrane-spanning domains. All chordopoxviruses have L2 homologs, suggesting an important function. Indeed, we were unable to isolate an infectious L2R deletion mutant. Consequently, we constructed an inducible mutant with a conditional lethal phenotype. When L2 expression was repressed, proteolytic processing of the major core proteins and the A17 protein, which is an essential component of the immature virion membrane, failed to occur, suggesting an early block in viral morphogenesis. At 8 h after infection in the presence of inducer, immature and mature virions were abundantly seen by electron microscopy. In contrast, those structures were rare in the absence of inducer and were replaced by large, dense aggregates of viroplasm. A minority of these aggregates had short spicule-coated membranes, which resembled the beginnings of crescent formation, at their periphery. These short membrane segments at the edge of the dense viroplasm increased in number at later times, and some immature virions were seen. Although the L2 protein was not detected under nonpermissive conditions, minute amounts could account for stunted and delayed viral membrane formation. These findings suggested that L2 is required for the formation or elongation of crescent membranes.


Subject(s)
Vaccinia virus/chemistry , Vaccinia virus/physiology , Viral Proteins/metabolism , Virion/chemistry , Virion/metabolism , Virus Assembly , Amino Acid Sequence , Cell Line , Gene Deletion , Genes, Essential , Genes, Viral , Humans , Microscopy, Electron, Transmission , Molecular Sequence Data , Sequence Alignment , Vaccinia virus/genetics , Viral Plaque Assay , Viral Proteins/genetics
13.
J Virol ; 82(11): 5368-80, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385250

ABSTRACT

The final assembly of rotavirus particles takes place in the endoplasmic reticulum (ER). In this work, we evaluated by RNA interference the relevance to rotavirus assembly and infectivity of grp78, protein disulfide isomerase (PDI), grp94, calnexin, calreticulin, and ERp57, members of the two ER folding systems described herein. Silencing the expression of grp94 and Erp57 had no effect on rotavirus infectivity, while knocking down the expression of any of the other four chaperons caused a reduction in the yield of infectious virus of about 50%. In grp78-silenced cells, the maturation of the oligosaccharide chains of NSP4 was retarded. In cells with reduced levels of calnexin, the oxidative folding of VP7 was impaired and the trimming of NSP4 was accelerated, and in calreticulin-silenced cells, the formation of disulfide bonds of VP7 was also accelerated. The knockdown of PDI impaired the formation and/or rearrangement of the VP7 disulfide bonds. All these conditions also affected the correct assembly of virus particles, since compared with virions from control cells, they showed an altered susceptibility to EGTA and heat treatments, a decreased specific infectivity, and a diminished reactivity to VP7 with monoclonal antibody M60, which recognizes only this protein when its disulfide bonds have been correctly formed. In the case of grp78-silenced cells, the virus produced bound less efficiently to MA104 cells than virus obtained from control cells. All these results suggest that these chaperones are involved in the quality control of rotavirus morphogenesis. The complexity of the steps of rotavirus assembly that occur in the ER provide a useful model for studying the organization and operation of the complex network of chaperones involved in maintaining the quality control of this organelle.


Subject(s)
Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Rotavirus/growth & development , Rotavirus/metabolism , Virion/metabolism , Animals , Calnexin/genetics , Calnexin/metabolism , Calreticulin/genetics , Calreticulin/metabolism , Cell Line , Disulfides/metabolism , Gene Expression Regulation , Macaca mulatta , Molecular Chaperones/genetics , Protein Disulfide-Isomerases/metabolism , Protein Folding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Rotavirus/genetics , Sensitivity and Specificity , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/growth & development , Virus Internalization
14.
Water Res ; 42(10-11): 2618-28, 2008 May.
Article in English | MEDLINE | ID: mdl-18291437

ABSTRACT

In this work, we have characterized the survival of Rhesus rotavirus (RRV) and human astrovirus Yuc8 in clean groundwater and contaminated surface water, as well as in phosphate-buffered solutions maintained in the same conditions as the environmental waters, and have compared the dynamics of virus inactivation with the persistence of the viral genomes, as determined by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). In addition, we also studied the tolerance of these viruses to chlorine disinfection. The reduction of infectivity of astrovirus was higher than for rotavirus, and also higher for both viruses in surface water as compared to groundwater. The enterobacterial content of the water as well as extrinsic factors, such as temperature and light, correlated with the stability of virus infectivity, and with the persistence of the virus genetic material, suggesting that molecular techniques to detect and quantify viral genomes would be suitable for the detection of viruses in water. The virus infectivity persisted in both types of water as well as in chlorine for times longer than previously reported. No decrease of infectivity was observed after 15 days of incubation in either type of water and the viruses remained infectious for months in groundwater. After 120 min in groundwater containing 2 mg/L of free chlorine, the infectivity of rotavirus and astrovirus was reduced by 0.78 and 1.3 logs, respectively. The longer persistence of viruses in this study could result from a combination of factors, including aggregation of the virus.


Subject(s)
Mamastrovirus/genetics , Mamastrovirus/pathogenicity , Rotavirus/genetics , Rotavirus/pathogenicity , Soil , Water Supply , Cell Line , Chlorine , Genome, Viral , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...