1
|
Varsani A, Custer JM, Cobb IN, Harding C, Collins CL, Suazo C, Schreck J, Fontenele RS, Stainton D, Dayaram A, Goldstein S, Kazlauskas D, Kraberger S, Krupovic M. Bacilladnaviridae: refined taxonomy and new insights into the biology and evolution of diatom-infecting DNA viruses. J Gen Virol 2025; 106. [PMID: 40072902 PMCID: PMC11903649 DOI: 10.1099/jgv.0.002084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Bacilladnaviruses are single-stranded DNA viruses that infect diatoms that, so far, have been primarily identified in marine organisms and environments. Using a viral metagenomics approach, we discovered 13 novel bacilladnaviruses originating from samples of mud-flat snail (Amphibola crenata; n=3 genomes) and benthic sediments (n=10 genomes) collected from the Avon-Heathcote Estuary in New Zealand. Comparative genomics and phylogenetic analysis of the new bacilladnavirus sequences in the context of the previously classified members of the family helped refine and further expand the Bacilladnaviridae taxonomy. Here, based on the replication-associated protein phylogeny and pairwise identities, we established 4 new genera - Aberdnavirus, Keisodnavirus, Puahadnavirus and Seawadnavirus - and 13 new species within the family. Comparison of the bacilladnavirus capsid protein sequences suggests that the positively charged N-terminal region (R-arm) is required for encapsidation of the larger genomes, whereas the smaller bacilladnavirus genomes can be packaged in the absence of the R-arm subdomain. Furthermore, analysis of the bacilladnavirus genomes revealed that members of three genera encode a highly derived variant of a phospholipase A1, which is predicted to be involved in the lysis of the infected diatoms and/or facilitates the entry of the virions into the host cells. Collectively, our results allow refining of the taxonomy of bacilladnaviruses and provide new insights into the biology and evolution of this understudied group of diatom viruses.
Collapse
Affiliation(s)
- Arvind Varsani
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Structural Biology Research Unit, Department of Integrative Biomedical Sciences, University of Cape Town, 7925, Cape Town, South Africa
| | - Joy M Custer
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Ilaria N Cobb
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Ciara Harding
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Courtney L Collins
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Crystal Suazo
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Joshua Schreck
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rafaela S Fontenele
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Daisy Stainton
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Anisha Dayaram
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Sharyn Goldstein
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Darius Kazlauskas
- Institute of Biotechnology, Vilnius University, Saulėtekio av. 7, Vilnius 10257, Lithuania
| | - Simona Kraberger
- The Biodesign Center for Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Mart Krupovic
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Archaeal Virology Unit, Paris, France
| |
Collapse
|
2
|
Diya DVD, Muneer A, Linu E, Sajeevan TP, Jayesh P, Joseph V, Philip R, Singh ISB. Replication kinetics, morphogenesis and interaction of shrimp parvovirus Penaeus stylirostris penstyldensovirus (PstDV1) in PmLyO-Sf9 cells. Microb Pathog 2025; 199:107179. [PMID: 39615708 DOI: 10.1016/j.micpath.2024.107179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Penaeus stylirostris penstyldensovirus (PstDV1) is one of the significant shrimp parvovirus which causes runt deformity syndrome in shrimps. In the current study, we attempted to elucidate the replication cycle of the virus in PmLyO-Sf9 cells. PstDV1 needs 4-5 h to complete replication in the cell line and release. Viral capsid gene expression leads to three peaks during the time course of the study at 4hpi, 12hpi and 72hpi. Immediate release of virus from the cell and reinfection in the cells observed. Calveloe mediated entry of virus to cells was noted through endosomal inhibition assay. Electron micrographs supported the findings which also depicted the receptor mediated entry, endosomal transport, viroplasm showing active virus replication and assembly of virus inside the nucleus to be released through cell lysis and occlusion bodies. Also, changes in cell cycle to make the cells to enter S phase was observed in cell cycle related genes and flow cytometric analysis. This reveal the potency of the cell line to study host-pathogen interaction, viral morphogenesis which could promotes successful development of antiviral formulations against the virus while preserving the environment for future generations.
Collapse
Affiliation(s)
- D V Dominic Diya
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| | - A Muneer
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| | - Eldho Linu
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| | - T P Sajeevan
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Kochi, India
| | - Puthumana Jayesh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| | - Valsamma Joseph
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| | - Rosamma Philip
- Department of Marine Biology, Microbiology and Biochemistry, School of Marine Sciences, Cochin University of Science and Technology, Kochi, India
| | - I S Bright Singh
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India.
| |
Collapse
|
3
|
Bieri J, Suter C, Caliaro O, Bartetzko S, Bircher C, Ros C. Globoside Is an Essential Intracellular Factor Required for Parvovirus B19 Endosomal Escape. Cells 2024; 13:1254. [PMID: 39120285 PMCID: PMC11311400 DOI: 10.3390/cells13151254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Human parvovirus B19 (B19V), like most parvoviruses, possesses phospholipase A2 (PLA2) activity, which is thought to mediate endosomal escape by membrane disruption. Here, we challenge this model and find evidence for a mechanism of B19V entry mediated by the glycosphingolipid globoside without endosome disruption and retrograde transport to the Golgi. We show that B19V PLA2 activity requires specific calcium levels and pH conditions that are not optimal in endosomes. Accordingly, endosomal membrane integrity was maintained during B19V entry. Furthermore, endosomes remained intact when loaded with MS2 bacteriophage particles pseudotyped with multiple B19V PLA2 subunits, providing superior enzymatic potential compared to native B19V. In globoside knockout cells, incoming viruses are arrested in the endosomal compartment and the infection is blocked. Infection can be rescued by promoting endosomal leakage with polyethyleneimine (PEI), demonstrating the essential role of globoside in facilitating endosomal escape. Incoming virus colocalizes with Golgi markers and interfering with Golgi function blocks infection, suggesting that globoside-mediated entry involves the Golgi compartment, which provides conditions favorable for the lipolytic PLA2. Our study challenges the current model of B19V entry and identifies globoside as an essential intracellular receptor required for endosomal escape.
Collapse
Affiliation(s)
- Jan Bieri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Corinne Suter
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Oliver Caliaro
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, 3012 Bern, Switzerland
| | - Seraina Bartetzko
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Cornelia Bircher
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| | - Carlos Ros
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland
| |
Collapse
|
4
|
Khanduja S, Bloom SM, Raman V, Deshpande CP, Hall CL, Forbes NS. Intracellular delivery of oncolytic viruses with engineered Salmonella causes viral replication and cell death. iScience 2024; 27:109813. [PMID: 38799578 PMCID: PMC11126981 DOI: 10.1016/j.isci.2024.109813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
As therapies, oncolytic viruses regress tumors and have the potential to induce antitumor immune responses that clear hard-to-treat and late-stage cancers. Despite this promise, clearance from the blood prevents treatment of internal solid tumors. To address this issue, we developed virus-delivering Salmonella (VDS) to carry oncolytic viruses into cancer cells. The VDS strain contains the PsseJ-lysE delivery circuit and has deletions in four homologous recombination genes (ΔrecB, ΔsbcB, ΔsbcCD, and ΔrecF) to preserve essential hairpins in the viral genome required for replication and infectivity. VDS delivered the genome for minute virus of mice (MVMp) to multiple cancers, including breast, pancreatic, and osteosarcoma. Viral delivery produced functional viral particles that are cytotoxic and infective to neighboring cells. The release of mature virions initiated new rounds of infection and amplified the infection. Using Salmonella for delivery will circumvent the limitations of oncolytic viruses and will provide a new therapy for many cancers.
Collapse
Affiliation(s)
- Shradha Khanduja
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Shoshana M.K. Bloom
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Chinmay P. Deshpande
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Christopher L. Hall
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
- Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA, USA
- Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA, USA
| |
Collapse
|
5
|
Reddy R, Yan SC, Hasanpour Segherlou Z, Hosseini-Siyanaki MR, Poe J, Perez-Vega C, Chiocca EA, Lucke-Wold B. Oncolytic viral therapy: a review and promising future directions. J Neurosurg 2024; 140:319-327. [PMID: 37877961 DOI: 10.3171/2023.6.jns23243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/06/2023] [Indexed: 10/26/2023]
Abstract
Oncolytic viral therapy is quickly emerging as a promising subset of immunotherapy, which theoretically can target tumor cells while sparing surrounding healthy cells by harnessing the replication machinery of viruses with tropism for tumor cells, resulting in direct oncolysis, and by transforming immunologically "cold" tumor into areas that elicit the host's immune response. This review provides an overview of oncolytic viral therapy until the present day, starting with the original concept in 1912. The general mechanism of oncolytic viruses (OVs) depends on selectively integrating them into tumor cells based on genetic engineering of viral genomic material, inducing oncolysis and eliciting the host's innate immune response. Moreover, a major component of oncolytic viral therapy has been herpes simplex virus, with talimogene laherparepvec being the only FDA-approved oncolytic viral therapy for the treatment of melanomas. This review explores the characteristics, advantages, disadvantages, and therapeutic uses of several DNA and RNA viral families. A snapshot of the oncolytic viral treatments used in the most recent and advanced clinical trials is also provided. Lastly, the challenges of implementing oncolytic viral therapy are explored, both at a molecular and clinical level, with a highlight of promising future directions. In particular, the lack of an optimal delivery method based on tumor type for oncolytic viral therapy poses a significant obstacle, even in clinical studies. Intrathecal continuous delivery of OVs is a promising prospect, potentially by adapting the novel continuous irrigation and drainage IRRAflow catheter. Further exploration and testing of the IRRAflow catheter should be undertaken.
Collapse
Affiliation(s)
- Ramya Reddy
- 1Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Sandra C Yan
- 1Department of Neurosurgery, University of Florida, Gainesville, Florida
| | | | | | - Jordan Poe
- 1Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Carlos Perez-Vega
- 3Department of Neurosurgery, Mayo Clinic, Jackonsville, Florida; and
| | - E Antonio Chiocca
- 4Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brandon Lucke-Wold
- 1Department of Neurosurgery, University of Florida, Gainesville, Florida
| |
Collapse
|
6
|
Glinšek K, Bozovičar K, Bratkovič T. CRISPR Technologies in Chinese Hamster Ovary Cell Line Engineering. Int J Mol Sci 2023; 24:ijms24098144. [PMID: 37175850 PMCID: PMC10179654 DOI: 10.3390/ijms24098144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
The Chinese hamster ovary (CHO) cell line is a well-established platform for the production of biopharmaceuticals due to its ability to express complex therapeutic proteins with human-like glycopatterns in high amounts. The advent of CRISPR technology has opened up new avenues for the engineering of CHO cell lines for improved protein production and enhanced product quality. This review summarizes recent advances in the application of CRISPR technology for CHO cell line engineering with a particular focus on glycosylation modulation, productivity enhancement, tackling adventitious agents, elimination of problematic host cell proteins, development of antibiotic-free selection systems, site-specific transgene integration, and CRISPR-mediated gene activation and repression. The review highlights the potential of CRISPR technology in CHO cell line genome editing and epigenetic engineering for the more efficient and cost-effective development of biopharmaceuticals while ensuring the safety and quality of the final product.
Collapse
Affiliation(s)
- Katja Glinšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Krištof Bozovičar
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tomaž Bratkovič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Hashemzadeh MS, Gharari N. Biosynthesis of a VLP-type nanocarrier specific to cancer cells using the BEVS expression system for targeted drug delivery. J Genet Eng Biotechnol 2023; 21:20. [PMID: 36795253 PMCID: PMC9932404 DOI: 10.1186/s43141-023-00479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVE Canine parvovirus (CPV) is a small virus without an envelope that consists of three viral proteins including VP1, VP2, and VP3. Exclusively, the VP2 can form a typically CPV-sized virus-like particle (CPV-VLP) that can be used as a biological nanocarrier for diagnostic and therapeutic purposes since these VLPs can target cancer cells specially through the transferrin surface receptors (TFRs). Consequently, we aimed to produce these nanocarriers to be used for specific targeting of cancer cells. METHODS Sf9 insect cells were transfected with constructed recombinant bacmid shuttle vector encoding an enhanced green fluorescent protein (EGFP) and CPV-VP2 by the cationic lipids of Cellfectin II. Subsequently, two recombinant baculoviruses expressing EGFP and VP2 were produced and expression of VP2 was increased under the optimal condition. In consequence, the CPV-VLP nanoparticles composed of recombinant VP2 subunits were extracted. The purity of VLPs was then evaluated by SDS-PAGE, and the structural integrity and quality of the final product were evaluated by TEM and HA methods. Eventually, the size distribution of the produced biological nanoparticles and their uniformity were determined by the DLS method. RESULTS The expression of EGFP protein was confirmed by fluorescent microscopy, and the expression of VP2 protein was evaluated by SDS-PAGE and western blotting. Infected Sf9 insect cells also showed cytopathic effects (CPEs), and the maximum expression of VP2 occurred at MOI of 10 (pfu/cell) at the harvest time of 72 h post-infection (hpi). After performing various stages of purification, buffer exchange, and concentration, the quality and structural integrity of the VLP product were confirmed. The results of the DLS technique showed the presence of uniform particles (PdI below 0.5) with an approximate size of 25 nm. CONCLUSION The results indicate BEVS as an appropriate and efficient system for generating CPV-VLPs, and the used method based on two-stage ultracentrifugation was appropriate for purifying these nanoparticles. Produced nanoparticles can be used as the biologic nano-carriers in future studies.
Collapse
Affiliation(s)
| | - Nariman Gharari
- grid.7605.40000 0001 2336 6580Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
8
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
9
|
Pedroza-Roldán C, Hernández-Almaraz MA, Elizondo-Quiroga D, Gutierrez-Ortega A, Acosta-Monroy CM, Charles-Niño C, Realpe-Quintero M, Robles-Gil SDC. Exclusive circulation of canine parvovirus type 2c in the Guadalajara metropolitan area in western Mexico: a five-year study. Arch Virol 2022; 167:2109-2121. [PMID: 35794491 DOI: 10.1007/s00705-022-05522-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/21/2022] [Indexed: 11/28/2022]
Abstract
Canine parvovirus type 2 (CPV-2) infection in dogs is associated with severe gastroenteritis, bloody diarrhea, and vomiting, resulting in high rates of death, especially in unvaccinated puppies within the first months of age. There are three variants, called CPV-2a, CPV-2b, and CPV-2c, co-circulating worldwide. Our group previously reported that the only circulating CPV-2 variant in the Guadalajara metropolitan area in western Mexico was type 2c. Now, a five-year study was performed in order to investigate the possible dominance of CPV-2c in our region. Rectal swabs were collected from 146 dogs with clinical gastroenteritis from May 2014 to August 2019 at the Veterinary Hospital of the University of Guadalajara. Of these, 90 dogs tested positive for canine parvovirus by PCR. Most of the infected dogs with CPV-2 had a partial or incomplete vaccination status (n = 88, 97.8%). Approximately 65% (n = 59) of them were mixed-breed dogs, 77.8% (n = 70) were under 6 months of age, and 37.8% (n = 34) of them died from clinical complications. RFLP analysis of amplicons derived from the vp2 gene showed that all 90 DNA samples corresponded to CPV-2c, with no evidence of the presence of CPV-2a or CPV-2b variants. Twenty-nine of the 90 DNA samples were selected for amplification of a portion of the vp2 gene, and sequencing of these amplicons showed that all of them had the sequence GAA at codon 426, encoding the amino acid glutamic acid, which is characteristic of CPV-2c. Phylogenetic analysis showed that the CPV-2c sequences were related to those of viruses from Europe and South America. The present study indicates that CPV-2c is still the only variant circulating in the dog population of the Guadalajara metropolitan area.
Collapse
Affiliation(s)
- César Pedroza-Roldán
- Departmento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Las Agujas, Jalisco, 44600, Zapopan, Jalisco, Mexico.
| | - Martín Alejandro Hernández-Almaraz
- Departmento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Las Agujas, Jalisco, 44600, Zapopan, Jalisco, Mexico
| | - Darwin Elizondo-Quiroga
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. Av. Normalistas No. 800, Col. Colinas de la Normal, CP 44270, Guadalajara, Jalisco, Mexico
| | - Abel Gutierrez-Ortega
- Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. Av. Normalistas No. 800, Col. Colinas de la Normal, CP 44270, Guadalajara, Jalisco, Mexico
| | - Carlos Maximiliano Acosta-Monroy
- Departmento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Las Agujas, Jalisco, 44600, Zapopan, Jalisco, Mexico
| | - Claudia Charles-Niño
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio P, CP 44340, Guadalajara, Jalisco, Mexico
| | - Mauricio Realpe-Quintero
- Departmento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Las Agujas, Jalisco, 44600, Zapopan, Jalisco, Mexico
| | - Sandra Del Carmen Robles-Gil
- Departmento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Camino Ramón Padilla Sánchez 2100, Las Agujas, Jalisco, 44600, Zapopan, Jalisco, Mexico
| |
Collapse
|
10
|
Orozco L, López-Pérez AM, Zarza H, Suzán G, List R. Dog demography and husbandry practices facilitate dog-wildlife conflict in a suburban-forest interface. Urban Ecosyst 2022. [DOI: 10.1007/s11252-022-01251-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
11
|
Vereecke N, Kvisgaard LK, Baele G, Boone C, Kunze M, Larsen LE, Theuns S, Nauwynck H. Molecular Epidemiology of Porcine Parvovirus Type 1 (PPV1) and the Reactivity of Vaccine-Induced Antisera Against Historical and Current PPV1 Strains. Virus Evol 2022; 8:veac053. [PMID: 35815310 PMCID: PMC9252332 DOI: 10.1093/ve/veac053] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/13/2022] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
Porcine Parvovirus Type 1 (PPV1) contributes to important losses in the swine industry worldwide. During a PPV1 infection, embryos and fetuses are targeted, resulting in stillbirth, mummification, embryonic death, and infertility (SMEDI syndrome). Even though vaccination is common in gilts and sows, strains mainly belonging to the 27a-like group have been spreading in Europe since early 2000s, resulting in SMEDI problems and requiring in-depth studies into the molecular epidemiology and vaccination efficacy of commercial vaccines. Here, we show that PPV1 has evolved since 1855 [1737, 1933] at a rate of 4.71 × 10−5 nucleotide substitutions per site per year. Extensive sequencing allowed evaluating and reassessing the current PPV1 VP1-based classifications, providing evidence for the existence of four relevant phylogenetic groups. While most European strains belong to the PPV1a (G1) or PPV1b (G2 or 27a-like) group, most Asian and American G2 strains and some European strains were divided into virulent PPV1c (e.g. NADL-8) and attenuated PPV1d (e.g. NADL-2) groups. The increase in the swine population, vaccination degree, and health management (vaccination and biosafety) influenced the spread of PPV1. The reactivity of anti-PPV1 antibodies from sows vaccinated with Porcilis© Parvo, Eryseng© Parvo, or ReproCyc© ParvoFLEX against different PPV1 field strains was the highest upon vaccination with ReproCyc© ParvoFLEX, followed by Eryseng© Parvo, and Porcilis© Parvo. Our findings contribute to the evaluation of the immunogenicity of existing vaccines and support the development of new vaccine candidates. Finally, the potential roles of cluster-specific hallmark amino acids in elevated pathogenicity and viral entry are discussed.
Collapse
Affiliation(s)
- Nick Vereecke
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University , Merelbeke, Belgium
- PathoSense BV , Lier, Belgium
| | - Lise Kirstine Kvisgaard
- Veterinary Clinical Microbiology, Department of Veterinary and Animal Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Guy Baele
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Carine Boone
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University , Merelbeke, Belgium
| | - Marius Kunze
- Boehringer Ingelheim Vetmedica GmbH , Binger Str. 173, 55216 Ingelheim am Rhein, Germany
| | - Lars Erik Larsen
- Veterinary Clinical Microbiology, Department of Veterinary and Animal Sciences, University of Copenhagen , Copenhagen, Denmark
| | | | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University , Merelbeke, Belgium
| |
Collapse
|
12
|
Ferreira T, Kulkarni A, Bretscher C, Nazarov PV, Hossain JA, Ystaas LAR, Miletic H, Röth R, Niesler B, Marchini A. Oncolytic H-1 Parvovirus Hijacks Galectin-1 to Enter Cancer Cells. Viruses 2022; 14:1018. [PMID: 35632759 PMCID: PMC9146882 DOI: 10.3390/v14051018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/16/2022] Open
Abstract
Clinical studies in glioblastoma and pancreatic carcinoma patients strongly support the further development of H-1 protoparvovirus (H-1PV)-based anticancer therapies. The identification of cellular factors involved in the H-1PV life cycle may provide the knowledge to improve H-1PV anticancer potential. Recently, we showed that sialylated laminins mediate H-1PV attachment at the cell membrane. In this study, we revealed that H-1PV also interacts at the cell surface with galectin-1 and uses this glycoprotein to enter cancer cells. Indeed, knockdown/out of LGALS1, the gene encoding galectin-1, strongly decreases the ability of H-1PV to infect and kill cancer cells. This ability is rescued by the re-introduction of LGALS1 into cancer cells. Pre-treatment with lactose, which is able to bind to galectins and modulate their cellular functions, decreased H-1PV infectivity in a dose dependent manner. In silico analysis reveals that LGALS1 is overexpressed in various tumours including glioblastoma and pancreatic carcinoma. We show by immunohistochemistry analysis of 122 glioblastoma biopsies that galectin-1 protein levels vary between tumours, with levels in recurrent glioblastoma higher than those in primary tumours or normal tissues. We also find a direct correlation between LGALS1 transcript levels and H-1PV oncolytic activity in 53 cancer cell lines from different tumour origins. Strikingly, the addition of purified galectin-1 sensitises poorly susceptible GBM cell lines to H-1PV killing activity by rescuing cell entry. Together, these findings demonstrate that galectin-1 is a crucial determinant of the H-1PV life cycle.
Collapse
Affiliation(s)
- Tiago Ferreira
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
| | - Amit Kulkarni
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg;
| | - Clemens Bretscher
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
| | - Petr V. Nazarov
- Bioinformatics Platform and Multiomics Data Science Research Group, Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| | - Jubayer A. Hossain
- Department of Biomedicine, University of Bergen, 5007 Bergen, Norway; (J.A.H.); (L.A.R.Y.); (H.M.)
| | - Lars A. R. Ystaas
- Department of Biomedicine, University of Bergen, 5007 Bergen, Norway; (J.A.H.); (L.A.R.Y.); (H.M.)
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, 5007 Bergen, Norway; (J.A.H.); (L.A.R.Y.); (H.M.)
- Department of Pathology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Ralph Röth
- nCounter Core Facility, Institute of Human Genetics, University of Heidelberg, 69120 Heidelberg, Germany; (R.R.); (B.N.)
- Department of Human Molecular Genetics, University of Heidelberg, 69120 Heidelberg, Germany
| | - Beate Niesler
- nCounter Core Facility, Institute of Human Genetics, University of Heidelberg, 69120 Heidelberg, Germany; (R.R.); (B.N.)
- Department of Human Molecular Genetics, University of Heidelberg, 69120 Heidelberg, Germany
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg;
| |
Collapse
|
13
|
Ferreira T, Kulkarni A, Bretscher C, Richter K, Ehrlich M, Marchini A. Oncolytic H-1 Parvovirus Enters Cancer Cells through Clathrin-Mediated Endocytosis. Viruses 2020; 12:v12101199. [PMID: 33096814 PMCID: PMC7594094 DOI: 10.3390/v12101199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
H-1 protoparvovirus (H-1PV) is a self-propagating virus that is non-pathogenic in humans and has oncolytic and oncosuppressive activities. H-1PV is the first member of the Parvoviridae family to undergo clinical testing as an anticancer agent. Results from clinical trials in patients with glioblastoma or pancreatic carcinoma show that virus treatment is safe, well-tolerated and associated with first signs of efficacy. Characterisation of the H-1PV life cycle may help to improve its efficacy and clinical outcome. In this study, we investigated the entry route of H-1PV in cervical carcinoma HeLa and glioma NCH125 cell lines. Using electron and confocal microscopy, we detected H-1PV particles within clathrin-coated pits and vesicles, providing evidence that the virus uses clathrin-mediated endocytosis for cell entry. In agreement with these results, we found that blocking clathrin-mediated endocytosis using specific inhibitors or small interfering RNA-mediated knockdown of its key regulator, AP2M1, markedly reduced H-1PV entry. By contrast, we found no evidence of viral entry through caveolae-mediated endocytosis. We also show that H-1PV entry is dependent on dynamin, while viral trafficking occurs from early to late endosomes, with acidic pH necessary for a productive infection. This is the first study that characterises the cell entry pathways of oncolytic H-1PV.
Collapse
Affiliation(s)
- Tiago Ferreira
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
| | - Amit Kulkarni
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg;
| | - Clemens Bretscher
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
| | - Karsten Richter
- Core Facility Electron Microscopy, German Cancer Research Centre, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - Marcelo Ehrlich
- Laboratory of Signal Transduction and Membrane Biology, The Shumins School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 69978 Tel Aviv, Israel;
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Centre, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany; (T.F.); (C.B.)
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg;
- Correspondence: or ; Tel.: +49-6221-424969 or +352-26-970-856
| |
Collapse
|
14
|
Rius-Rocabert S, García-Romero N, García A, Ayuso-Sacido A, Nistal-Villan E. Oncolytic Virotherapy in Glioma Tumors. Int J Mol Sci 2020; 21:ijms21207604. [PMID: 33066689 PMCID: PMC7589679 DOI: 10.3390/ijms21207604] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Glioma tumors are one of the most devastating cancer types. Glioblastoma is the most advanced stage with the worst prognosis. Current therapies are still unable to provide an effective cure. Recent advances in oncolytic immunotherapy have generated great expectations in the cancer therapy field. The use of oncolytic viruses (OVs) in cancer treatment is one such immune-related therapeutic alternative. OVs have a double oncolytic action by both directly destroying the cancer cells and stimulating a tumor specific immune response to return the ability of tumors to escape the control of the immune system. OVs are one promising alternative to conventional therapies in glioma tumor treatment. Several clinical trials have proven the feasibility of using some viruses to specifically infect tumors, eluding undesired toxic effects in the patient. Here, we revisited the literature to describe the main OVs proposed up to the present moment as therapeutic alternatives in order to destroy glioma cells in vitro and trigger tumor destruction in vivo. Oncolytic viruses were divided with respect to the genome in DNA and RNA viruses. Here, we highlight the results obtained in various clinical trials, which are exploring the use of these agents as an alternative where other approaches provide limited hope.
Collapse
Affiliation(s)
- Sergio Rius-Rocabert
- Microbiology Section, Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, 28668 Madrid, Spain;
- Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, 28668 Madrid, Spain
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, 28668 Madrid, Spain;
| | - Noemí García-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain;
| | - Antonia García
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, 28668 Madrid, Spain;
| | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain;
- Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
- Correspondence: (A.A.-S.); (E.N.-V.); Tel.: +34-913-724-714 (E.N.-V.)
| | - Estanislao Nistal-Villan
- Microbiology Section, Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, 28668 Madrid, Spain;
- Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo-CEU, 28668 Madrid, Spain
- Correspondence: (A.A.-S.); (E.N.-V.); Tel.: +34-913-724-714 (E.N.-V.)
| |
Collapse
|
15
|
Hartley A, Kavishwar G, Salvato I, Marchini A. A Roadmap for the Success of Oncolytic Parvovirus-Based Anticancer Therapies. Annu Rev Virol 2020; 7:537-557. [PMID: 32600158 DOI: 10.1146/annurev-virology-012220-023606] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autonomous rodent protoparvoviruses (PVs) are promising anticancer agents due to their excellent safety profile, natural oncotropism, and oncosuppressive activities. Viral infection can trigger immunogenic cell death, activating the immune system against the tumor. However, the efficacy of this treatment in recent clinical trials is moderate compared with results seen in preclinical work. Various strategies have been employed to improve the anticancer activities of oncolytic PVs, including development of second-generation parvoviruses with enhanced oncolytic and immunostimulatory activities and rational combination of PVs with other therapies. Understanding the cellular factors involved in the PV life cycle is another important area of investigation. Indeed, these studies may lead to the identification of biomarkers that would allow a more personalized use of PV-based therapies. This review focuses on this work and the challenges that still need to be overcome to move PVs forward into clinical practice as an effective therapeutic option for cancer patients.
Collapse
Affiliation(s)
- Anna Hartley
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Gayatri Kavishwar
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Ilaria Salvato
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany; .,Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| |
Collapse
|
16
|
Kemmerer M, Bonning BC. Transcytosis of Junonia coenia densovirus VP4 across the gut epithelium of Spodoptera frugiperda (Lepidoptera: Noctuidae). INSECT SCIENCE 2020; 27:22-32. [PMID: 29704325 DOI: 10.1111/1744-7917.12600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/10/2018] [Accepted: 04/19/2018] [Indexed: 06/08/2023]
Abstract
The Junonia coenia densovirus rapidly traverses the gut epithelium of the host lepidopteran without replicating in the gut cells. The ability of this virus to transcytose across the gut epithelium is of interest for the potential use of virus structural proteins as delivery vehicles for insecticidal peptides that act within the insect hemocoel, rather than in the gut. In this study, we used fall armyworm, Spodoptera frugiperda to examine the binding of the virus to brush border membrane vesicle proteins by two-dimensional ligand blot analysis. We also assessed the rate of flux of the primary viral structural protein, VP4 fused to eGFP with a proline-rich linker (VP4-P-eGFP) through the gut epithelium ex vivo in an Ussing chamber. The mechanisms involved with transcytosis of VP4-P-eGFP were assessed by use of inhibitors. Bovine serum albumin (BSA) and eGFP were used as positive and negative control proteins, respectively. In contrast to BSA, which binds to multiple proteins on the brush border membrane, VP4-P-eGFP binding was specific to a protein of high molecular mass. Protein flux was significantly higher for VP4-P-eGFP after 2 h than for albumin or eGFP, with rapid transcytosis of VP4-P-eGFP within the first 30 min. In contrast to BSA which transcytosed following clathrin-mediated endocytosis, the movement of VP4-P-eGFP was vesicle-mediated but clathrin-independent. The specificity of binding combined with the efficiency of transport across the gut epithelium suggest that VP4 will provide a useful carrier for insecticidal peptides active within the hemocoel of key lepidopteran pests including S. frugiperda.
Collapse
Affiliation(s)
- Mariah Kemmerer
- Department of Entomology, Iowa State University, Ames, Iowa, USA
| | - Bryony C Bonning
- Department of Entomology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
17
|
Diversity and Evolution of Novel Invertebrate DNA Viruses Revealed by Meta-Transcriptomics. Viruses 2019; 11:v11121092. [PMID: 31775324 PMCID: PMC6950620 DOI: 10.3390/v11121092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/22/2019] [Accepted: 11/23/2019] [Indexed: 12/16/2022] Open
Abstract
DNA viruses comprise a wide array of genome structures and infect diverse host species. To date, most studies of DNA viruses have focused on those with the strongest disease associations. Accordingly, there has been a marked lack of sampling of DNA viruses from invertebrates. Bulk RNA sequencing has resulted in the discovery of a myriad of novel RNA viruses, and herein we used this methodology to identify actively transcribing DNA viruses in meta-transcriptomic libraries of diverse invertebrate species. Our analysis revealed high levels of phylogenetic diversity in DNA viruses, including 13 species from the Parvoviridae, Circoviridae, and Genomoviridae families of single-stranded DNA virus families, and six double-stranded DNA virus species from the Nudiviridae, Polyomaviridae, and Herpesviridae, for which few invertebrate viruses have been identified to date. By incorporating the sequence of a "blank" experimental control we also highlight the importance of reagent contamination in metagenomic studies. In sum, this work expands our knowledge of the diversity and evolution of DNA viruses and illustrates the utility of meta-transcriptomic data in identifying organisms with DNA genomes.
Collapse
|
18
|
Liu X, Wang H, Liu X, Li Y, Chen J, Zhang J, Wang X, Shen S, Wang H, Deng F, Wang M, Guan W, Hu Z. Genomic and transcriptional analyses of novel parvoviruses identified from dead peafowl. Virology 2019; 539:80-91. [PMID: 31706163 DOI: 10.1016/j.virol.2019.10.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/30/2019] [Accepted: 10/26/2019] [Indexed: 01/20/2023]
Abstract
To identify potential pathogens responsible for a disease outbreak of cultured peafowls in China in 2013, metagenomic sequencing was conducted. The genomes of two closely related parvoviruses, namely peafowl parvovirus 1 (PePV1) and PePV2, were identified with size of 4428 bp and 4348 bp, respectively. Phylogenetic analysis revealed that both viruses are novel parvoviruses, belonging to the proposed genus Chapparvovirus of Parvoviridae. The transcriptional profile of PePV1 was analyzed by transfecting a nearly complete PePV1 genome into HEK-293T cells. Results revealed that PePV1 employs one promoter and two polyadenylation sites to start and terminate its transcriptions, with one donor site and two acceptor sites for pre-mRNA splicing. PePV1 DNA and structural protein were detected in several tissues of a dead peafowl, which appeared to have suffered enteritis, pneumonia and viremia. These results provide novel information of chapparvoviruses, and call for attention to the potential pathogens.
Collapse
Affiliation(s)
- Xiaoping Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hanzhong Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaoqian Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Li
- Hubei Wildlife Rescue Center, China
| | | | | | - Xi Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shu Shen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Hualin Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fei Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Manli Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Wuxiang Guan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Zhihong Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
19
|
López-Pérez AM, Moreno K, Chaves A, Ibarra-Cerdeña CN, Rubio A, Foley J, List R, Suzán G, Sarmiento RE. Carnivore Protoparvovirus 1 at the Wild-Domestic Carnivore Interface in Northwestern Mexico. ECOHEALTH 2019; 16:502-511. [PMID: 31375949 DOI: 10.1007/s10393-019-01436-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Eighty-three wild and domestic carnivores of nine species from Janos Biosphere Reserve (JBR), Mexico, were tested by serologic and molecular assays to determine exposure and infection rates of carnivore protoparvovirus 1. Overall, 50.8% (33/65) of the wild carnivores and 100% (18/18) of the domestic dogs tested were seropositive for Canine protoparvovirus 1 (CPV), while 23% (15/65) of the wild carnivores and 22.2% (4/18) of the domestic dogs were PCR positive for CPV. Phylogenetic analysis confirmed circulation of CVP-2 with residues 426 Asn (CPV2a = 1/19) and 426 Glu (CPV-2c = 18/19) among carnivores in JBR. The prevalence of both PCR positivity and antibodies to CPV varied significantly among wild host species. Of the six identified haplotypes, three were unique to kit foxes (Vulpes macrotis) (the species with higher haplotype richness) and two to striped skunks (Mephitis mephitis). The remaining haplotype was shared among all carnivore species including dogs suggesting non-host specificity and bidirectional and continuous viral transmission cycle in the JBR. The phylogenetic similarity of CPV strains from dogs and wild carnivores and the higher prevalence of CPV in wild carnivores captured near towns relative to those captured far from towns suggest that dogs might be an important source of CPV infection for wild carnivores in the JBR. We provide evidence that cross-species transmission occurs at the domestic-wildlife interface in JBR.
Collapse
Affiliation(s)
- Andres M López-Pérez
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Av. Universidad #3000, 04510, Mexico City, D.F., Mexico
- Fundación para el Manejo y la Conservación de la Vida Silvestre FMCOVIS A.C., Ciudad de México, Mexico
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, CA, USA
| | - Karen Moreno
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Av. Universidad #3000, 04510, Mexico City, D.F., Mexico
| | - Andrea Chaves
- Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, CA, USA
| | - Carlos N Ibarra-Cerdeña
- Departamento de Ecología Humana, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Unidad Mérida, Mérida, Yucatán, Mexico
| | - Andre Rubio
- Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - Janet Foley
- School of Veterinary Medicine, Department of Medicine and Epidemiology, University of California, Davis, CA, USA
| | - Rurik List
- Área de Investigación en Biología de la Conservación, Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma de Villada, Estado de México, Mexico
| | - Gerardo Suzán
- Laboratorio de Ecología de Enfermedades y Una Salud, Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Av. Universidad #3000, 04510, Mexico City, D.F., Mexico.
| | - Rosa Elena Sarmiento
- Laboratorio de Virología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico, D.F., Mexico
| |
Collapse
|
20
|
Canine parvovirus (CPV) phylogeny is associated with disease severity. Sci Rep 2019; 9:11266. [PMID: 31375758 PMCID: PMC6677720 DOI: 10.1038/s41598-019-47773-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/22/2019] [Indexed: 12/02/2022] Open
Abstract
After its first identification in 1978, canine parvovirus (CPV) has been recognized all around the world as a major threat for canine population health. This ssDNA virus is characterized by a high substitution rate and several genetic and phenotypic variants emerged over time. Overall, the definition of 3 main antigenic variants was established based on specific amino acid markers located in a precise capsid position. However, the detection of several minor variants and incongruence observed between the antigenic classification and phylogeny have posed doubts on the reliability of this scheme. At the same time, CPV heterogeneity has favored the hypothesis of a differential virulence among variants, although no robust and consistent demonstration has been provided yet. The present study rejects the antigenic variant concept and attempts to evaluate the association between CPV strain phylogeny, reconstructed using the whole information contained in the VP2 coding gene, and several clinical and hemato-biochemical parameters, assessed from 34 CPV infected dogs at admission. By using different statistical approaches, the results of the present study show an association between viral phylogeny and host parameters ascribable to immune system, coagulation profile, acute phase response and, more generally, to the overall picture of the animal response. Particularly, a strong and significant phylogenetic signal was proven for neutrophil count and WBC. Therefore, despite the limited sample size, a relation between viral phylogeny and disease severity has been observed for the first time, suggesting that CPV virulence is an inherited trait. The likely existence of clades with different virulence highlights once more the relevance of intensive epidemiological monitoring and research on CPV evolution to better understand the virulence determinants, their epidemiology and develop adequate countermeasures.
Collapse
|
21
|
Wang K, Du S, Wang Y, Wang S, Luo X, Zhang Y, Liu C, Wang H, Pei Z, Hu G. Isolation and identification of tiger parvovirus in captive siberian tigers and phylogenetic analysis of VP2 gene. INFECTION GENETICS AND EVOLUTION 2019; 75:103957. [PMID: 31299323 DOI: 10.1016/j.meegid.2019.103957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 01/03/2023]
Abstract
To better understand the prevalence and molecular epidemiology of parvovirus, this study reports the isolation and characterization of a tiger parvovirus (TPV) named CHJL-Siberian Tiger-01/2017 from a captive Siberian tiger in Jilin Province, China. A phylogenetic tree based on the full-length VP2 nucleotide sequence was constructed using the isolated strain in this study and 56 reference strains. The results showed that all the parvoviruses can be grouped into two large branches: the canine parvovirus (CPV) branch and the feline parvovirus (FPV) branch. FPV strains comprised TPVs, FPVs, blue fox parvoviruses (BFPVs), mink enteritis viruses (MEVs), and raccoon feline parvoviruses (RFPVs), and CPV strains comprised CPVs and raccoon dog parvoviruses (RDPVs). RFPVs are also often very closely related to those sampled from other carnivorous species, and raccoons may represent conduits for parvovirus transmission to other hosts. The results of amino acid changes in the VP2 protein of the isolated strain showed that amino acid Ile 101 was mutated to Thr (I 101T). Taken together, a field TPV strain CHJL-Siberian Tiger-01/2017 was isolated, which may be suitable for future studies on FPV infection, replication and vaccine development. This study provided new important findings about the evolution of parvovirus infection in tigers.
Collapse
Affiliation(s)
- Kai Wang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China.
| | - Shuaishuai Du
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Yiqi Wang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Shaoying Wang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Xiaoqing Luo
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Yuanyuan Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Cunfa Liu
- Wildlife Ambulance Breeding Center of Jilin Province, Jingyue Street No.10500, Changchun, PR China
| | - Haijun Wang
- Wildlife Ambulance Breeding Center of Jilin Province, Jingyue Street No.10500, Changchun, PR China
| | - Zhihua Pei
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| | - Guixue Hu
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No. 2888, Changchun, PR China
| |
Collapse
|
22
|
Bretscher C, Marchini A. H-1 Parvovirus as a Cancer-Killing Agent: Past, Present, and Future. Viruses 2019; 11:v11060562. [PMID: 31216641 PMCID: PMC6630270 DOI: 10.3390/v11060562] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
The rat protoparvovirus H-1PV is nonpathogenic in humans, replicates preferentially in cancer cells, and has natural oncolytic and oncosuppressive activities. The virus is able to kill cancer cells by activating several cell death pathways. H-1PV-mediated cancer cell death is often immunogenic and triggers anticancer immune responses. The safety and tolerability of H-1PV treatment has been demonstrated in early clinical studies in glioma and pancreatic carcinoma patients. Virus treatment was associated with surrogate signs of efficacy including immune conversion of tumor microenvironment, effective virus distribution into the tumor bed even after systemic administration, and improved patient overall survival compared with historical control. However, monotherapeutic use of the virus was unable to eradicate tumors. Thus, further studies are needed to improve H-1PV's anticancer profile. In this review, we describe H-1PV's anticancer properties and discuss recent efforts to improve the efficacy of H-1PV and, thereby, the clinical outcome of H-1PV-based therapies.
Collapse
Affiliation(s)
- Clemens Bretscher
- Laboratory of Oncolytic Virus Immuno-Therapeutics, F011, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany.
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, F011, German Cancer Research Center, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany.
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, 84 Val Fleuri, L-1526 Luxembourg, Luxembourg.
| |
Collapse
|
23
|
Parvovirus B19 Uncoating Occurs in the Cytoplasm without Capsid Disassembly and It Is Facilitated by Depletion of Capsid-Associated Divalent Cations. Viruses 2019; 11:v11050430. [PMID: 31083301 PMCID: PMC6563316 DOI: 10.3390/v11050430] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/19/2022] Open
Abstract
Human parvovirus B19 (B19V) traffics to the cell nucleus where it delivers the genome for replication. The intracellular compartment where uncoating takes place, the required capsid structural rearrangements and the cellular factors involved remain unknown. We explored conditions that trigger uncoating in vitro and found that prolonged exposure of capsids to chelating agents or to buffers with chelating properties induced a structural rearrangement at 4 °C resulting in capsids with lower density. These lighter particles remained intact but were unstable and short exposure to 37 °C or to a freeze-thaw cycle was sufficient to trigger DNA externalization without capsid disassembly. The rearrangement was not observed in the absence of chelating activity or in the presence of MgCl2 or CaCl2, suggesting that depletion of capsid-associated divalent cations facilitates uncoating. The presence of assembled capsids with externalized DNA was also detected during B19V entry in UT7/Epo cells. Following endosomal escape and prior to nuclear entry, a significant proportion of the incoming capsids rearranged and externalized the viral genome without capsid disassembly. The incoming capsids with accessible genomes accumulated in the nuclear fraction, a process that was prevented when endosomal escape or dynein function was disrupted. In their uncoated conformation, capsids immunoprecipitated from cytoplasmic or from nuclear fractions supported in vitro complementary-strand synthesis at 37 °C. This study reveals an uncoating strategy of B19V based on a limited capsid rearrangement prior to nuclear entry, a process that can be mimicked in vitro by depletion of divalent cations.
Collapse
|
24
|
Characterization of the RNA Transcription Profile of Bombyx mori Bidensovirus. Viruses 2019; 11:v11040325. [PMID: 30987230 PMCID: PMC6521256 DOI: 10.3390/v11040325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 01/15/2023] Open
Abstract
Bombyx mori bidensovirus (BmBDV) is a single-stranded DNA (ssDNA) virus from the genus Bidensovirus of the Bidnaviridae family, which, thus far, solely infects insects. It has a unique genome that contains bipartite DNA molecules (VD1 and VD2). In this study, we explored the detailed transcription mapping of the complete BmBDV genome (VD1 and VD2) by rapid amplification of cDNA ends (RACE), reverse transcription quantitative real-time PCR (RT-qPCR), and luciferase assays. For the first time, we report the transcription map of VD2. Our mapping of the transcriptional start sites reveals that the NS genes in VD1 have separate transcripts that are derived from overlapping promoters, P5 and P5.5. Thus, our study provides a strategy for alternative promoter usage in the expression of BmBDV genes.
Collapse
|
25
|
Yan Z, Zou W, Feng Z, Shen W, Park SY, Deng X, Qiu J, Engelhardt JF. Establishment of a High-Yield Recombinant Adeno-Associated Virus/Human Bocavirus Vector Production System Independent of Bocavirus Nonstructural Proteins. Hum Gene Ther 2019; 30:556-570. [PMID: 30398383 DOI: 10.1089/hum.2018.173] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The genome of recombinant adeno-associated virus 2 (rAAV2) remains a promising candidate for gene therapy for cystic fibrosis (CF) lung disease, but due to limitations in the packaging capacity and the tropism of this virus with respect to the airways, strategies have evolved for packaging an rAAV2 genome (up to 5.8 kb) into the capsid of human bocavirus 1 (HBoV1) to produce a chimeric rAAV2/HBoV1 vector. Although a replication-incompetent HBoV1 genome has been established as a trans helper for capsid complementation, this system remains suboptimal with respect to virion yield. Here, a streamlined production system is described based on knowledge of the involvement of HBoV1 nonstructural (NS) proteins NS1, NS2, NS3, NS4, and NP1 in the process of virion production. The analyses reveal that NS1 and NS2 negatively impact virion production, NP1 is required to prevent premature termination of transcription of the cap mRNA from the native genome, and silent mutations within the polyadenylation sites of the cap coding sequence can eliminate this requirement for NP1. It is further shown that preventing the expression of all NS proteins significantly increases virion yield. Whereas the expression of capsid proteins VP1, VP2, and VP3 from a codon-optimized cap mRNA was highly efficient, optimal virion assembly, and thus potency, required enhanced VP1 expression, entailing a separate VP1 expression cassette. The final NS protein-free production system uses three-plasmid co-transfection of HEK293 cells, with one trans helper plasmid encoding VP1 and the AAV2 Rep proteins, and another encoding VP2-3 and components from adenovirus. This system yielded >16-fold more virions than the prototypic system, without reducing transduction potency. This increase in virion production is expected to facilitate greatly both research on the biology of rAAV2/HBoV1 and preclinical studies testing the effectiveness of this vector for gene therapy of CF lung disease in large animal models.
Collapse
Affiliation(s)
- Ziying Yan
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa, Iowa City, Iowa
| | - Wei Zou
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Zehua Feng
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| | - Weiran Shen
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Soo Yeun Park
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa
| | - Xuefeng Deng
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jianming Qiu
- 3 Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas
| | - John F Engelhardt
- 1 Department of Anatomy and Cell Biology, University of Iowa, Iowa City, Iowa.,2 Center for Gene Therapy, University of Iowa, Iowa City, Iowa
| |
Collapse
|
26
|
Recessive Host Range Mutants and Unsusceptible Cells That Inactivate Virions without Genome Penetration: Ecological and Technical Implications. J Virol 2019; 93:JVI.01767-18. [PMID: 30429341 DOI: 10.1128/jvi.01767-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/05/2018] [Indexed: 12/20/2022] Open
Abstract
Although microviruses do not possess a visible tail structure, one vertex rearranges after interacting with host lipopolysaccharides. Most examinations of host range, eclipse, and penetration were conducted before this "host-induced" unique vertex was discovered and before DNA sequencing became routine. Consequently, structure-function relationships dictating host range remain undefined. Biochemical and genetic analyses were conducted with two closely related microviruses, α3 and ST-1. Despite ∼90% amino acid identity, the natural host of α3 is Escherichia coli C, whereas ST-1 is a K-12-specific phage. Virions attached and eclipsed to both native and unsusceptible hosts; however, they breached only the native host's cell wall. This suggests that unsusceptible host-phage interactions promote off-pathway reactions that can inactivate viruses without penetration. This phenomenon may have broader ecological implications. To determine which structural proteins conferred host range specificity, chimeric virions were generated by individually interchanging the coat, spike, or DNA pilot proteins. Interchanging the coat protein switched host range. However, host range expansion could be conferred by single point mutations in the coat protein. The expansion phenotype was recessive: genetically mutant progeny from coinfected cells did not display the phenotype. Thus, mutant isolation required populations generated in environments with low multiplicities of infection (MOI), a phenomenon that may have impacted past host range studies in both prokaryotic and eukaryotic systems. The resulting genetic and structural data were consistent enough that host range expansion could be predicted, broadening the classical definition of antireceptors to include interfaces between protein complexes within the capsid.IMPORTANCE To expand host range, viruses must interact with unsusceptible host cell surfaces, which could be detrimental. As observed in this study, virions were inactivated without genome penetration. This may be advantageous to potential new hosts, culling the viral population from which an expanded host range mutant could emerge. When identified, altered host range mutations were recessive. Accordingly, isolation required populations generated in low-MOI environments. However, in laboratory settings, viral propagation includes high-MOI conditions. Typically, infected cultures incubate until all cells produce progeny. Thus, coinfections dominate later replication cycles, masking recessive host range expansion phenotypes. This may have impacted similar studies with other viruses. Last, structural and genetic data could be used to predict site-directed mutant phenotypes, which may broaden the classic antireceptor definition to include interfaces between capsid complexes.
Collapse
|
27
|
Development and evaluation of a gold nanoparticle-based immunochromatographic strip test for the detection of canine parvovirus. Arch Virol 2018; 163:2359-2368. [DOI: 10.1007/s00705-018-3846-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/12/2018] [Indexed: 11/26/2022]
|
28
|
Imaging, Tracking and Computational Analyses of Virus Entry and Egress with the Cytoskeleton. Viruses 2018; 10:v10040166. [PMID: 29614729 PMCID: PMC5923460 DOI: 10.3390/v10040166] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Viruses have a dual nature: particles are “passive substances” lacking chemical energy transformation, whereas infected cells are “active substances” turning-over energy. How passive viral substances convert to active substances, comprising viral replication and assembly compartments has been of intense interest to virologists, cell and molecular biologists and immunologists. Infection starts with virus entry into a susceptible cell and delivers the viral genome to the replication site. This is a multi-step process, and involves the cytoskeleton and associated motor proteins. Likewise, the egress of progeny virus particles from the replication site to the extracellular space is enhanced by the cytoskeleton and associated motor proteins. This overcomes the limitation of thermal diffusion, and transports virions and virion components, often in association with cellular organelles. This review explores how the analysis of viral trajectories informs about mechanisms of infection. We discuss the methodology enabling researchers to visualize single virions in cells by fluorescence imaging and tracking. Virus visualization and tracking are increasingly enhanced by computational analyses of virus trajectories as well as in silico modeling. Combined approaches reveal previously unrecognized features of virus-infected cells. Using select examples of complementary methodology, we highlight the role of actin filaments and microtubules, and their associated motors in virus infections. In-depth studies of single virion dynamics at high temporal and spatial resolutions thereby provide deep insight into virus infection processes, and are a basis for uncovering underlying mechanisms of how cells function.
Collapse
|
29
|
Mutations in the Non-Structural Protein-Coding Sequence of Protoparvovirus H-1PV Enhance the Fitness of the Virus and Show Key Benefits Regarding the Transduction Efficiency of Derived Vectors. Viruses 2018; 10:v10040150. [PMID: 29584637 PMCID: PMC5923444 DOI: 10.3390/v10040150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 11/27/2022] Open
Abstract
Single nucleotide changes were introduced into the non-structural (NS) coding sequence of the H-1 parvovirus (PV) infectious molecular clone and the corresponding virus stocks produced, thereby generating H1-PM-I, H1-PM-II, H1-PM-III, and H1-DM. The effects of the mutations on viral fitness were analyzed. Because of the overlapping sequences of NS1 and NS2, the mutations affected either NS2 (H1-PM-II, -III) or both NS1 and NS2 proteins (H1-PM-I, H1-DM). Our results show key benefits of PM-I, PM-II, and DM mutations with regard to the fitness of the virus stocks produced. Indeed, these mutants displayed a higher production of infectious virus in different cell cultures and better spreading capacity than the wild-type virus. This correlated with a decreased particle-to-infectivity (P/I) ratio and stimulation of an early step(s) of the viral cycle prior to viral DNA replication, namely, cell binding and internalization. These mutations also enhance the transduction efficiency of H-1PV-based vectors. In contrast, the PM-III mutation, which affects NS2 at a position downstream of the sequence deleted in Del H-1PV, impaired virus replication and spreading. We hypothesize that the NS2 protein—modified in H1-PM-I, H1-PM-II, and H1-DM—may result in the stimulation of some maturation step(s) of the capsid and facilitate virus entry into subsequently infected cells.
Collapse
|
30
|
Gil-Ranedo J, Hernando E, Valle N, Riolobos L, Maroto B, Almendral JM. Differential phosphorylation and n-terminal configuration of capsid subunits in parvovirus assembly and viral trafficking. Virology 2018. [PMID: 29524834 DOI: 10.1016/j.virol.2018.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The T1 parvovirus Minute Virus of Mice (MVM) was used to study the roles that phosphorylation and N-terminal domains (Nt) configuration of capsid subunits may play in icosahedral nuclear viruses assembly. In synchronous MVM infection, capsid subunits newly assembled as two types of cytoplasmic trimeric intermediates (3VP2, and 1VP1:2VP2) harbored a VP1 phosphorylation level fivefold higher than that of VP2, and hidden Nt. Upon nuclear translocation at S phase, VP1-Nt became exposed in the heterotrimer and subsequent subviral assembly intermediates. Empty capsid subunits showed a phosphorylation level restored to VP1:VP2 stoichiometry, and the Nt concealed in their interior. However ssDNA-filled virus maturing at S/G2 lacked VP1 phosphorylation and one major VP2 phosphopeptide, and exposed VP2-Nt. Endosomal VP2-Nt cleavage resulted in VP3 subunits devoid of any phospholabel, implying that incoming viral particles specifically harbor a low phosphorylation status. Phosphorylation provides a mechanistic coupling of parvovirus nuclear assembly to the cell cycle.
Collapse
Affiliation(s)
- Jon Gil-Ranedo
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain
| | - Eva Hernando
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain
| | - Noelia Valle
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain
| | - Laura Riolobos
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain
| | - Beatriz Maroto
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain
| | - José M Almendral
- Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
31
|
Ros C, Bayat N, Wolfisberg R, Almendral JM. Protoparvovirus Cell Entry. Viruses 2017; 9:v9110313. [PMID: 29072600 PMCID: PMC5707520 DOI: 10.3390/v9110313] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 01/25/2023] Open
Abstract
The Protoparvovirus (PtPV) genus of the Parvoviridae family of viruses includes important animal pathogens and reference molecular models for the entire family. Some virus members of the PtPV genus have arisen as promising tools to treat tumoral processes, as they exhibit marked oncotropism and oncolytic activities while being nonpathogenic for humans. The PtPVs invade and replicate within the nucleus making extensive use of the transport, transcription and replication machineries of the host cells. In order to reach the nucleus, PtPVs need to cross over several intracellular barriers and traffic through different cell compartments, which limit their infection efficiency. In this review we summarize molecular interactions, capsid structural transitions and hijacking of cellular processes, by which the PtPVs enter and deliver their single-stranded DNA genome into the host cell nucleus. Understanding mechanisms that govern the complex PtPV entry will be instrumental in developing approaches to boost their anticancer therapeutic potential and improving their safety profile.
Collapse
Affiliation(s)
- Carlos Ros
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Nooshin Bayat
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain.
| | - Raphael Wolfisberg
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark.
| | - José M Almendral
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
32
|
Carrillo PJP, Medrano M, Valbuena A, Rodríguez-Huete A, Castellanos M, Pérez R, Mateu MG. Amino Acid Side Chains Buried along Intersubunit Interfaces in a Viral Capsid Preserve Low Mechanical Stiffness Associated with Virus Infectivity. ACS NANO 2017; 11:2194-2208. [PMID: 28117975 DOI: 10.1021/acsnano.6b08549] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Single-molecule experimental techniques and theoretical approaches reveal that important aspects of virus biology can be understood in biomechanical terms at the nanoscale. A detailed knowledge of the relationship in virus capsids between small structural changes caused by single-point mutations and changes in mechanical properties may provide further physics-based insights into virus function; it may also facilitate the engineering of viral nanoparticles with improved mechanical behavior. Here, we used the minute virus of mice to undertake a systematic experimental study on the contribution to capsid stiffness of amino acid side chains at interprotein interfaces and the specific noncovalent interactions they establish. Selected side chains were individually truncated by introducing point mutations to alanine, and the effects on local and global capsid stiffness were determined using atomic force microscopy. The results revealed that, in the natural virus capsid, multiple, mostly hydrophobic, side chains buried along the interfaces between subunits preserve a comparatively low stiffness of most (S2 and S3) regions. Virtually no point mutation tested substantially reduced stiffness, whereas most mutations increased stiffness of the S2/S3 regions. This stiffening was invariably associated with reduced virus yields during cell infection. The experimental evidence suggests that a comparatively low stiffness at S3/S2 capsid regions may have been biologically selected because it facilitates capsid assembly, increasing infectious virus yields. This study demonstrated also that knowledge of individual amino acid side chains and biological pressures that determine the physical behavior of a protein nanoparticle may be used for engineering its mechanical properties.
Collapse
Affiliation(s)
- Pablo José P Carrillo
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - María Medrano
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Alejandro Valbuena
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Alicia Rodríguez-Huete
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Milagros Castellanos
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Rebeca Pérez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
33
|
Zhao H, Zhao G, Wang W. Susceptibility of porcine preimplantation embryos to viruses associated with reproductive failure. Theriogenology 2016; 86:1631-6. [PMID: 27423729 DOI: 10.1016/j.theriogenology.2016.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/13/2016] [Accepted: 06/03/2016] [Indexed: 11/16/2022]
Abstract
In the modern biological area, the applications of pig as a laboratory model have extensive prospects, such as gene transfer, IVF, SCNT, and xenotransplantation. However, the risk of pathogen transmission by porcine embryos is always a topic to be investigated, especially the viruses related to reproductive failure, for instance, pseudorabies virus, porcine reproductive and respiratory syndrome virus, porcine parvovirus, and porcine circovirus type 2. It should be mentioned that the zona pellucida (ZP) of porcine embryos can be a barrier against the viruses, but certain pathogens may stick to or even pass through the ZP. With intact, free, and damaged ZP, porcine preimplantation embryos are susceptible to these viruses in varying degrees, which may be associated with the virus-specific receptor on embryonic cell membrane. These topics are discussed in the present review.
Collapse
Affiliation(s)
- Haijing Zhao
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Guangyuan Zhao
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Wenjun Wang
- Center for Reproductive Medicine, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|
34
|
Mascarenhas JX, Korokhov N, Burger L, Kassim A, Tuter J, Miller D, Borgschulte T, George HJ, Chang A, Pintel DJ, Onions D, Kayser KJ. Genetic engineering of CHO cells for viral resistance to minute virus of mice. Biotechnol Bioeng 2016; 114:576-588. [PMID: 27642072 DOI: 10.1002/bit.26186] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
Contamination by the parvovirus minute virus of mice (MVM) remains a challenge in Chinese hamster ovary (CHO) biopharmaceutical production processes. Although infrequent, infection of a bioreactor can be catastrophic for a manufacturer, can impact patient drug supply and safety, and can have regulatory implications. We evaluated engineering a CHO parental cell line (CHOZN® GS-/- ) to create a new host cell line that is resistant to MVM infection by modifying the major receptors used by the virus to enter cells. Attachment to a cell surface receptor is a key first step in the infection cycle for many viruses. While the exact functional receptor for MVM binding to CHO cell surface is unknown, sialic acid on the cell surface has been implicated. In this work, we used the zinc finger nuclease gene editing technology to validate the role of sialic acid on the cell surface in the binding and internalization of the MVM virus. Our approach was to systematically mutate genes involved in cell surface sialylation and then challenge each cell line for their ability to resist viral entry and propagation. To test the importance of sialylation, the following genes were knocked out: the CMP-sialic acid transporter, solute carrier family 35A1 (Slc35a1), the core 1-β-1,3-galactosyltransferase-1 specific chaperone (Cosmc), and mannosyl (α-1,3-)-glycoprotein β-1,2-N-acetylglucosaminyltransferase (Mgat1) as well as members of the sialyltransferase family. Slc35a1 is responsible for transporting sialic acid into the Golgi. Knocking out function of this gene in a cell results in asialylated glycan structures, thus eliminating the ability of MVM to bind to and enter the cell. The complete absence of sialic acid on the Slc35a1 knockout cell line led to complete resistance to MVM infection. The Cosmc and Mgat1 knockouts also show significant inhibition of infection likely due to their effect on decreasing cell surface sialic acid. Previously in vitro glycan analysis has been used to elucidate the precise sialic acid structures required for MVM binding and internalization. In this work, we performed the sequential knockout of various sialyltransferases that add terminal sialic acid to glycans with different linkage specificities. Cell lines with modifications of the various genes included in this study resulted in varying effects on MVM infection expanding on the knowledge of MVM receptors. MVM resistant host cell lines were also tested for the production of model recombinant proteins. Our data demonstrate that resistance against the MVM virus can be incorporated into CHO production cell lines, adding another level of defense against the devastating financial consequences of MVM infection without compromising recombinant protein yield or quality. Biotechnol. Bioeng. 2017;114: 576-588. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Lisa Burger
- University of Missouri School of Medicine, Columbia, Missouri
| | - Ademola Kassim
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Jason Tuter
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Daniel Miller
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Trissa Borgschulte
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Henry J George
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| | - Audrey Chang
- Bioreliance, MilliporeSigma, Rockville, Maryland
| | - David J Pintel
- University of Missouri School of Medicine, Columbia, Missouri
| | - David Onions
- Bioreliance, MilliporeSigma, Rockville, Maryland
| | - Kevin J Kayser
- Cell Sciences and Development, SAFC MilliporeSigma, Saint Louis, Missouri, 63103
| |
Collapse
|
35
|
Zamarin D, Pesonen S. Replication-Competent Viruses as Cancer Immunotherapeutics: Emerging Clinical Data. Hum Gene Ther 2016; 26:538-49. [PMID: 26176173 PMCID: PMC4968310 DOI: 10.1089/hum.2015.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Replication-competent (oncolytic) viruses (OV) as cancer immunotherapeutics have gained an increasing level of attention over the last few years while the clinical evidence of virus-mediated antitumor immune responses is still anecdotal. Multiple clinical studies are currently ongoing and more immunomonitoring results are expected within the next five years. All viruses can be recognized by the immune system and are therefore potential candidates for immune therapeutics. However, each virus activates innate immune system by using different combination of recognition receptors/pathways which leads to qualitatively different adaptive immune responses. This review summarizes immunological findings in cancer patients following treatment with replication-competent viruses.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- 1 Memorial Sloan Kettering Cancer Center , New York, New York
| | | |
Collapse
|
36
|
Moving oncolytic viruses into the clinic: clinical-grade production, purification, and characterization of diverse oncolytic viruses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16018. [PMID: 27088104 PMCID: PMC4822647 DOI: 10.1038/mtm.2016.18] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 12/13/2022]
Abstract
Oncolytic viruses (OVs) are unique anticancer agents based on their pleotropic modes of action, which include, besides viral tumor cell lysis, activation of antitumor immunity. A panel of diverse viruses, often genetically engineered, has advanced to clinical investigation, including phase 3 studies. This diversity of virotherapeutics not only offers interesting opportunities for the implementation of different therapeutic regimens but also poses challenges for clinical translation. Thus, manufacturing processes and regulatory approval paths need to be established for each OV individually. This review provides an overview of clinical-grade manufacturing procedures for OVs using six virus families as examples, and key challenges are discussed individually. For example, different virus features with respect to particle size, presence/absence of an envelope, and host species imply specific requirements for measures to ensure sterility, for handling, and for determination of appropriate animal models for toxicity testing, respectively. On the other hand, optimization of serum-free culture conditions, increasing virus yields, development of scalable purification strategies, and formulations guaranteeing long-term stability are challenges common to several if not all OVs. In light of the recent marketing approval of the first OV in the Western world, strategies for further upscaling OV manufacturing and optimizing product characterization will receive increasing attention.
Collapse
|
37
|
Palma M, de la Roja N, Montón M, Sastre P, Ramírez S, Barreiro B, Venteo A, Rueda P. Development of a duplex rapid assay for immunoglobulins M and G to evaluate the parvoviral immune status of clinically healthy dogs. J Vet Diagn Invest 2016; 28:299-303. [PMID: 26951330 DOI: 10.1177/1040638716634400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
A duplex rapid assay for detection of serum antibodies to canine parvovirus (CPV) was developed. Canine immunoglobulin (Ig)M or IgG were captured in immunotubes with anti-canine IgM or IgG and detected with parvovirus VP2 recombinant protein followed by an anti-VP2 monoclonal antibody. The assay was tested using a collection of sera from dogs that were vaccinated against CPV on arrival at an animal shelter in Madrid, Spain. Results were compared with those of 2 commercial enzyme-linked immunosorbent assays (ELISAs) considered as reference techniques. A high correlation was found between the duplex rapid assay and the ELISAs, presenting an accuracy of 98% and 100% for IgG and IgM, respectively. According to the IgG and IgM levels at days 0-3 postvaccination, the samples were divided into 2 groups. One group of dogs showed high IgG and low IgM values at the first sampling post-vaccination and during the following 14 days, indicating that they had previously been in contact with the virus, either by vaccination or infection before arrival at the animal shelter. A second group of dogs appeared to be unvaccinated or uninfected before arrival at the animal shelter because they had negative IgM and IgG values soon after vaccination. These animals responded to vaccination, as demonstrated by seroconversion of both isotypes of immunoglobulins. The developed assay appears to be useful in determining the unknown immune status of dogs to CPV, especially in kennels and shelters where the rate of infection by CPV is relatively high.
Collapse
Affiliation(s)
- Marco Palma
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Nuria de la Roja
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Mercedes Montón
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Patricia Sastre
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Susana Ramírez
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Belén Barreiro
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Angel Venteo
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| | - Paloma Rueda
- Inmunología y Genética Aplicada SA, Madrid, Spain (Palma, de la Roja, Montón, Sastre, Barreiro, Venteo, Rueda)Centro de Protección Animal, Madrid, Spain (Ramírez)
| |
Collapse
|
38
|
Adventitious agent contamination risk mitigation: engineering MMV virus resistance into CHO cells. BMC Proc 2015. [PMCID: PMC4685385 DOI: 10.1186/1753-6561-9-s9-o2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
39
|
Yutin N, Shevchenko S, Kapitonov V, Krupovic M, Koonin EV. A novel group of diverse Polinton-like viruses discovered by metagenome analysis. BMC Biol 2015; 13:95. [PMID: 26560305 PMCID: PMC4642659 DOI: 10.1186/s12915-015-0207-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/28/2015] [Indexed: 01/08/2023] Open
Abstract
Background The rapidly growing metagenomic databases provide increasing opportunities for computational discovery of new groups of organisms. Identification of new viruses is particularly straightforward given the comparatively small size of viral genomes, although fast evolution of viruses complicates the analysis of novel sequences. Here we report the metagenomic discovery of a distinct group of diverse viruses that are distantly related to the eukaryotic virus-like transposons of the Polinton superfamily. Results The sequence of the putative major capsid protein (MCP) of the unusual linear virophage associated with Phaeocystis globosa virus (PgVV) was used as a bait to identify potential related viruses in metagenomic databases. Assembly of the contigs encoding the PgVV MCP homologs followed by comprehensive sequence analysis of the proteins encoded in these contigs resulted in the identification of a large group of Polinton-like viruses (PLV) that resemble Polintons (polintoviruses) and virophages in genome size, and share with them a conserved minimal morphogenetic module that consists of major and minor capsid proteins and the packaging ATPase. With a single exception, the PLV lack the retrovirus-type integrase that is encoded in the genomes of all Polintons and the Mavirus group of virophages. However, some PLV encode a newly identified tyrosine recombinase-integrase that is common in bacteria and bacteriophages and is also found in the Organic Lake virophage group. Although several PLV genomes and individual genes are integrated into algal genomes, it appears likely that most of the PLV are viruses. Given the absence of protease and retrovirus-type integrase, the PLV could resemble the ancestral polintoviruses that evolved from bacterial tectiviruses. Apart from the conserved minimal morphogenetic module, the PLV widely differ in their genome complements but share a gene network with Polintons and virophages, suggestive of multiple gene exchanges within a shared gene pool. Conclusions The discovery of PLV substantially expands the emerging class of eukaryotic viruses and transposons that also includes Polintons and virophages. This class of selfish elements is extremely widespread and might have been a hotbed of eukaryotic virus, transposon and plasmid evolution. New families of these elements are expected to be discovered. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0207-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Natalya Yutin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Sofiya Shevchenko
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Vladimir Kapitonov
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA
| | - Mart Krupovic
- Unité Biologie Moléculaire du Gène chez les Extrêmophiles, Department of Microbiology, Institut Pasteur, Paris, France
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, 20894, USA.
| |
Collapse
|
40
|
Chen H, Dou Y, Tang Y, Zhang Z, Zheng X, Niu X, Yang J, Yu X, Diao Y. Isolation and Genomic Characterization of a Duck-Origin GPV-Related Parvovirus from Cherry Valley Ducklings in China. PLoS One 2015; 10:e0140284. [PMID: 26465143 PMCID: PMC4605506 DOI: 10.1371/journal.pone.0140284] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/23/2015] [Indexed: 11/25/2022] Open
Abstract
A newly emerged duck parvovirus, which causes beak atrophy and dwarfism syndrome (BADS) in Cherry Valley ducks, has appeared in Northern China since March 2015. To explore the genetic diversity among waterfowl parvovirus isolates, the complete genome of an identified isolate designated SDLC01 was sequenced and analyzed in the present study. Genomic sequence analysis showed that SDLC01 shared 90.8%–94.6% of nucleotide identity with goose parvovirus (GPV) isolates and 78.6%–81.6% of nucleotide identity with classical Muscovy duck parvovirus (MDPV) isolates. Phylogenetic analysis of 443 nucleotides (nt) of the fragment A showed that SDLC01 was highly similar to a mule duck isolate (strain D146/02) and close to European GPV isolates but separate from Asian GPV isolates. Analysis of the left inverted terminal repeat regions revealed that SDLC01 had two major segments deleted between positions 160–176 and 306–322 nt compared with field GPV and MDPV isolates. Phylogenetic analysis of Rep and VP1 encoded by two major open reading frames of parvoviruses revealed that SDLC01 was distinct from all GPV and MDPV isolates. The viral pathogenicity and genome characterization of SDLC01 suggest that the novel GPV (N-GPV) is the causative agent of BADS and belongs to a distinct GPV-related subgroup. Furthermore, N-GPV sequences were detected in diseased ducks by polymerase chain reaction and viral proliferation was demonstrated in duck embryos and duck embryo fibroblast cells.
Collapse
Affiliation(s)
- Hao Chen
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Yanguo Dou
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Yi Tang
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Zhenjie Zhang
- College of Basic Medicine, Taishan Medical University, Tai’an, Shandong, 271000, China
| | - Xiaoqiang Zheng
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Xiaoyu Niu
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Jing Yang
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Xianglong Yu
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
| | - Youxiang Diao
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an, Shandong, 271018, China
- * E-mail:
| |
Collapse
|
41
|
Abstract
DNA viruses undertake their replication within the cell nucleus, and therefore they must first deliver their genome into the nucleus of their host cells. Thus, trafficking across the nuclear envelope is at the basis of DNA virus infections. Nuclear transport of molecules with diameters up to 39 nm is a tightly regulated process that occurs through the nuclear pore complex (NPC). Due to the enormous diversity of virus size and structure, each virus has developed its own strategy for entering the nucleus of their host cells, with no two strategies alike. For example, baculoviruses target their DNA-containing capsid to the NPC and subsequently enter the nucleus intact, while the hepatitis B virus capsid crosses the NPC but disassembles at the nuclear side of the NPC. For other viruses such as herpes simplex virus and adenovirus, although both dock at the NPC, they have each developed a distinct mechanism for the subsequent delivery of their genome into the nucleus. Remarkably, other DNA viruses, such as parvoviruses and human papillomaviruses, access the nucleus through an NPC-independent mechanism. This review discusses our current understanding of the mechanisms used by DNA viruses to deliver their genome into the nucleus, and further presents the experimental evidence for such mechanisms.
Collapse
Affiliation(s)
- Nikta Fay
- Department of Zoology, University of British Columbia Vancouver, BC, Canada
| | - Nelly Panté
- Department of Zoology, University of British Columbia Vancouver, BC, Canada
| |
Collapse
|
42
|
Lezhnin YN, Kravchenko YE, Frolova EI, Chumakov PM, Chumakov SP. Oncotoxic proteins in cancer therapy: Mechanisms of action. Mol Biol 2015. [DOI: 10.1134/s0026893315020077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Castellanos M, Carrillo PJP, Mateu MG. Quantitatively probing propensity for structural transitions in engineered virus nanoparticles by single-molecule mechanical analysis. NANOSCALE 2015; 7:5654-5664. [PMID: 25744136 DOI: 10.1039/c4nr07046a] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Viruses are increasingly being studied from the perspective of fundamental physics at the nanoscale as biologically evolved nanodevices with many technological applications. In viral particles of the minute virus of mice (MVM), folded segments of the single-stranded DNA genome are bound to the capsid inner wall and act as molecular buttresses that increase locally the mechanical stiffness of the particle. We have explored whether a quantitative linkage exists in MVM particles between their DNA-mediated stiffening and impairment of a heat-induced, virus-inactivating structural change. A series of structurally modified virus particles with disrupted capsid-DNA interactions and/or distorted capsid cavities close to the DNA-binding sites were engineered and characterized, both in classic kinetics assays and by single-molecule mechanical analysis using atomic force microscopy. The rate constant of the virus inactivation reaction was found to decrease exponentially with the increase in elastic constant (stiffness) of the regions closer to DNA-binding sites. The application of transition state theory suggests that the height of the free energy barrier of the virus-inactivating structural transition increases linearly with local mechanical stiffness. From a virological perspective, the results indicate that infectious MVM particles may have acquired the biological advantage of increased survival under thermal stress by evolving architectural elements that rigidify the particle and impair non-productive structural changes. From a nanotechnological perspective, this study provides proof of principle that determination of mechanical stiffness and its manipulation by protein engineering may be applied for quantitatively probing and tuning the conformational dynamics of virus-based and other protein-based nanoassemblies.
Collapse
Affiliation(s)
- Milagros Castellanos
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain.
| | | | | |
Collapse
|
44
|
Gupta SK, Gandham RK, Sahoo AP, Tiwari AK. Viral genes as oncolytic agents for cancer therapy. Cell Mol Life Sci 2015; 72:1073-94. [PMID: 25408521 PMCID: PMC11113997 DOI: 10.1007/s00018-014-1782-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 10/29/2014] [Accepted: 11/13/2014] [Indexed: 12/20/2022]
Abstract
Many viruses have the ability to modulate the apoptosis, and to accomplish it; viruses encode proteins which specifically interact with the cellular signaling pathways. While some viruses encode proteins, which inhibit the apoptosis or death of the infected cells, there are viruses whose encoded proteins can kill the infected cells by multiple mechanisms, including apoptosis. A particular class of these viruses has specific gene(s) in their genomes which, upon ectopic expression, can kill the tumor cells selectively without affecting the normal cells. These genes and their encoded products have demonstrated great potential to be developed as novel anticancer therapeutic agents which can specifically target and kill the cancer cells leaving the normal cells unharmed. In this review, we will discuss about the viral genes having specific cancer cell killing properties, what is known about their functioning, signaling pathways and their therapeutic applications as anticancer agents.
Collapse
Affiliation(s)
- Shishir Kumar Gupta
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - Ravi Kumar Gandham
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. P. Sahoo
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| | - A. K. Tiwari
- Molecular Biology Lab, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122 UP India
| |
Collapse
|
45
|
Marchini A, Bonifati S, Scott EM, Angelova AL, Rommelaere J. Oncolytic parvoviruses: from basic virology to clinical applications. Virol J 2015; 12:6. [PMID: 25630937 PMCID: PMC4323056 DOI: 10.1186/s12985-014-0223-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/03/2014] [Indexed: 12/28/2022] Open
Abstract
Accumulated evidence gathered over recent decades demonstrated that some members of the Parvoviridae family, in particular the rodent protoparvoviruses H-1PV, the minute virus of mice and LuIII have natural anticancer activity while being nonpathogenic to humans. These studies have laid the foundations for the launch of a first phase I/IIa clinical trial, in which the rat H-1 parvovirus is presently undergoing evaluation for its safety and first signs of efficacy in patients with glioblastoma multiforme. After a brief overview of the biology of parvoviruses, this review focuses on the studies which unraveled the antineoplastic properties of these agents and supported their clinical use as anticancer therapeutics. Furthermore, the development of novel parvovirus-based anticancer strategies with enhanced specificity and efficacy is discussed, in particular the development of second and third generation vectors and the combination of parvoviruses with other anticancer agents. Lastly, we address the key challenges that remain towards a more rational and efficient use of oncolytic parvoviruses in clinical settings, and discuss how a better understanding of the virus life-cycle and of the cellular factors involved in virus infection, replication and cytotoxicity may promote the further development of parvovirus-based anticancer therapies, open new prospects for treatment and hopefully improve clinical outcome.
Collapse
Affiliation(s)
- Antonio Marchini
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Serena Bonifati
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Eleanor M Scott
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Assia L Angelova
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| | - Jean Rommelaere
- Infection and Cancer Program, Tumor Virology Division (F010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 242, 69120, Heidelberg, Germany.
| |
Collapse
|
46
|
Bodewes R, Hapsari R, Rubio García A, Sánchez Contreras GJ, van de Bildt MWG, de Graaf M, Kuiken T, Osterhaus ADME. Molecular epidemiology of seal parvovirus, 1988-2014. PLoS One 2014; 9:e112129. [PMID: 25390639 PMCID: PMC4229121 DOI: 10.1371/journal.pone.0112129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/13/2014] [Indexed: 01/30/2023] Open
Abstract
A novel parvovirus was discovered recently in the brain of a harbor seal (Phoca vitulina) with chronic meningo-encephalitis. Phylogenetic analysis of this virus indicated that it belongs to the genus Erythroparvovirus, to which also human parvovirus B19 belongs. In the present study, the prevalence, genetic diversity and clinical relevance of seal parvovirus (SePV) infections was evaluated in both harbor and grey seals (Halichoerus grypus) that lived in Northwestern European coastal waters from 1988 to 2014. To this end, serum and tissue samples collected from seals were tested for the presence of seal parvovirus DNA by real-time PCR and the sequences of the partial NS gene and the complete VP2 gene of positive samples were determined. Seal parvovirus DNA was detected in nine (8%) of the spleen tissues tested and in one (0.5%) of the serum samples tested, including samples collected from seals that died in 1988. Sequence analysis of the partial NS and complete VP2 genes of nine SePV revealed multiple sites with nucleotide substitutions but only one amino acid change in the VP2 gene. Estimated nucleotide substitution rates per year were 2.00 × 10(-4) for the partial NS gene and 1.15 × 10(-4) for the complete VP2 gene. Most samples containing SePV DNA were co-infected with phocine herpesvirus 1 or PDV, so no conclusions could be drawn about the clinical impact of SePV infection alone. The present study is one of the few in which the mutation rates of parvoviruses were evaluated over a period of more than 20 years, especially in a wildlife population, providing additional insights into the genetic diversity of parvoviruses.
Collapse
Affiliation(s)
- Rogier Bodewes
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Ana Rubio García
- Seal Rehabilitation and Research Centre, Pieterburen, the Netherlands
| | | | | | - Miranda de Graaf
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Thijs Kuiken
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Albert D. M. E. Osterhaus
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
- Viroclinics Biosciences BV, Rotterdam, the Netherlands
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine, Hannover, Germany
- Artemis One Health, Utrecht, the Netherlands
| |
Collapse
|
47
|
Venkataram Prasad BV, Shanker S, Hu L, Choi JM, Crawford SE, Ramani S, Czako R, Atmar RL, Estes MK. Structural basis of glycan interaction in gastroenteric viral pathogens. Curr Opin Virol 2014; 7:119-27. [PMID: 25073118 PMCID: PMC4251800 DOI: 10.1016/j.coviro.2014.05.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 01/12/2023]
Abstract
A critical event in the life cycle of a virus is its initial attachment to host cells. This involves recognition by the viruses of specific receptors on the cell surface, including glycans. Viruses typically exhibit strain-dependent variations in recognizing specific glycan receptors, a feature that contributes significantly to cell tropism, host specificity, host adaptation and interspecies transmission. Examples include influenza viruses, noroviruses, rotaviruses, and parvoviruses. Both rotaviruses and noroviruses are well known gastroenteric pathogens that are of significant global health concern. While rotaviruses, in the family Reoviridae, are the major causative agents of life-threatening diarrhea in children, noroviruses, which belong to the Caliciviridae family, cause epidemic and sporadic cases of acute gastroenteritis across all age groups. Both exhibit enormous genotypic and serotypic diversity. Consistent with this diversity each exhibits strain-dependent variations in the types of glycans they recognize for cell attachment. This chapter reviews the current status of the structural biology of such strain-dependent glycan specificities in these two families of viruses.
Collapse
Affiliation(s)
- B V Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, United States; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States.
| | - Sreejesh Shanker
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Jae-Mun Choi
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Rita Czako
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Robert L Atmar
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
48
|
Abstract
Parvoviruses are small, rugged, nonenveloped protein particles containing a linear, nonpermuted, single-stranded DNA genome of ∼5 kb. Their limited coding potential requires optimal adaptation to the environment of particular host cells, where entry is mediated by a variable program of capsid dynamics, ultimately leading to genome ejection from intact particles within the host nucleus. Genomes are amplified by a continuous unidirectional strand-displacement mechanism, a linear adaptation of rolling circle replication that relies on the repeated folding and unfolding of small hairpin telomeres to reorient the advancing fork. Progeny genomes are propelled by the viral helicase into the preformed capsid via a pore at one of its icosahedral fivefold axes. Here we explore how the fine-tuning of this unique replication system and the mechanics that regulate opening and closing of the capsid fivefold portals have evolved in different viral lineages to create a remarkably complex spectrum of phenotypes.
Collapse
Affiliation(s)
| | - Peter Tattersall
- Departments of 1Laboratory Medicine and.,Genetics, Yale University Medical School, New Haven, Connecticut 06510;
| |
Collapse
|
49
|
Krupovic M, Koonin EV. Evolution of eukaryotic single-stranded DNA viruses of the Bidnaviridae family from genes of four other groups of widely different viruses. Sci Rep 2014; 4:5347. [PMID: 24939392 PMCID: PMC4061559 DOI: 10.1038/srep05347] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022] Open
Abstract
Single-stranded (ss)DNA viruses are extremely widespread, infect diverse hosts from all three domains of life and include important pathogens. Most ssDNA viruses possess small genomes that replicate by the rolling-circle-like mechanism initiated by a distinct virus-encoded endonuclease. However, viruses of the family Bidnaviridae, instead of the endonuclease, encode a protein-primed type B DNA polymerase (PolB) and hence break this pattern. We investigated the provenance of all bidnavirus genes and uncover an unexpected turbulent evolutionary history of these unique viruses. Our analysis strongly suggests that bidnaviruses evolved from a parvovirus ancestor from which they inherit a jelly-roll capsid protein and a superfamily 3 helicase. The radiation of bidnaviruses from parvoviruses was probably triggered by integration of the ancestral parvovirus genome into a large virus-derived DNA transposon of the Polinton (polintovirus) family resulting in the acquisition of the polintovirus PolB gene along with terminal inverted repeats. Bidnavirus genes for a receptor-binding protein and a potential novel antiviral defense modulator are derived from dsRNA viruses (Reoviridae) and dsDNA viruses (Baculoviridae), respectively. The unusual evolutionary history of bidnaviruses emphasizes the key role of horizontal gene transfer, sometimes between viruses with completely different genomes but occupying the same niche, in the emergence of new viral types.
Collapse
Affiliation(s)
- Mart Krupovic
- Institut Pasteur, Unité Biologie Moléculaire du Gène chez les Extrêmophiles, Department of Microbiology, Paris 75015, France
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
50
|
Complementary induction of immunogenic cell death by oncolytic parvovirus H-1PV and gemcitabine in pancreatic cancer. J Virol 2014; 88:5263-76. [PMID: 24574398 DOI: 10.1128/jvi.03688-13] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Novel therapies employing oncolytic viruses have emerged as promising anticancer modalities. The cure of particularly aggressive malignancies requires induction of immunogenic cell death (ICD), coupling oncolysis with immune responses via calreticulin, ATP, and high-mobility group box protein B1 (HMGB1) release from dying tumor cells. The present study shows that in human pancreatic cancer cells (pancreatic ductal adenocarcinoma [PDAC] cells n=4), oncolytic parvovirus H-1 (H-1PV) activated multiple interconnected death pathways but failed to induce calreticulin exposure or ATP release. In contrast, H-1PV elevated extracellular HMGB1 levels by 4.0±0.5 times (58%±9% of total content; up to 100 ng/ml) in all infected cultures, whether nondying, necrotic, or apoptotic. An alternative secretory route allowed H-1PV to overcome the failure of gemcitabine to trigger HMGB1 release, without impeding cytotoxicity or other ICD activities of the standard PDAC medication. Such broad resistance of H-1PV-induced HMGB1 release to apoptotic blockage coincided with but was uncoupled from an autocrine interleukin-1β (IL-1β) loop. That and the pattern of viral determinants maintained in gemcitabine-treated cells suggested the activation of an inflammasome/caspase 1 (CASP1) platform alongside DNA detachment and/or nuclear exclusion of HMGB1 during early stages of the viral life cycle. We concluded that H-1PV infection of PDAC cells is signaled through secretion of the alarmin HMGB1 and, besides its own oncolytic effect, might convert drug-induced apoptosis into an ICD process. A transient arrest of cells in the cyclin A1-rich S phase would suffice to support compatibility of proliferation-dependent H-1PV with cytotoxic regimens. These properties warrant incorporation of the oncolytic virus H-1PV, which is not pathogenic in humans, into multimodal anticancer treatments. IMPORTANCE The current therapeutic concepts targeting aggressive malignancies require an induction of immunogenic cell death characterized by exposure of calreticulin (CRT) as well as release of ATP and HMGB1 from dying cells. In pancreatic tumor cells (PDAC cells) infected with the oncolytic parvovirus H-1PV, only HMGB1 was released by all infected cells, whether nondying, necrotic, or succumbing to one of the programmed death pathways, including contraproductive apoptosis. Our data suggest that active secretion of HMGB1 from PDAC cells is a sentinel reaction emerging during early stages of the viral life cycle, irrespective of cell death, that is compatible with and complements cytotoxic regimens. Consistent induction of HMGB1 secretion raised the possibility that this reaction might be a general "alarming" phenomenon characteristic of H-1PV's interaction with the host cell; release of IL-1β points to the possible involvement of a danger-sensing inflammasome platform. Both provide a basis for further virus-oriented studies.
Collapse
|