Next Issue
Volume 2, February
Previous Issue
Volume 1, December
 
 

Viruses, Volume 2, Issue 1 (January 2010) – 18 articles , Pages 1-333

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
1056 KiB  
Review
Herpesviruses and Autophagy: Catch Me If You Can!
by Yolaine Cavignac and Audrey Esclatine
Viruses 2010, 2(1), 314-333; https://doi.org/10.3390/v2010314 - 26 Jan 2010
Cited by 51 | Viewed by 15203
Abstract
Autophagy is an evolutionarily conserved cellular degradation pathway involving the digestion of intracellular components via the lysosomal pathway. The autophagic pathway constitutively maintains cellular homeostasis by recycling cytoplasmic organelles and proteins, but it is also stimulated by environmental stress conditions, such as starvation, [...] Read more.
Autophagy is an evolutionarily conserved cellular degradation pathway involving the digestion of intracellular components via the lysosomal pathway. The autophagic pathway constitutively maintains cellular homeostasis by recycling cytoplasmic organelles and proteins, but it is also stimulated by environmental stress conditions, such as starvation, oxidative stress, and the accumulation of misfolded proteins. It also acts as a cellular defense mechanism against microorganisms by contributing to both the innate and adaptive immunity, and by eliminating intracellular pathogens (xenophagy). There is growing evidence that host cells try to control Herpesvirus infections by activating the autophagic machinery. However, it is well-known that Herpesviruses are smart pathogens and several, such as HSV-1, HCMV and HHV-8, are known to have developed numerous defense strategies for evading the host’s immune response. Inhibition of the antiviral autophagic mechanism has also been reported. Autophagy has also been shown to enhance the major histocompatibility complex presentation of at least two viral proteins, the EBVencoded EBNA-1 and the HSV-1 encoded gB. In this review, we present an overview of recent advances in our understanding of the complex interplay between autophagy and Herpesviruses. Full article
(This article belongs to the Special Issue Antiviral Responses to Herpes Viruses)
Show Figures

Figure 1

148 KiB  
Review
Dual Role of p53 in Innate Antiviral Immunity
by Carmen Rivas, Stuart A. Aaronson and Cesar Munoz-Fontela
Viruses 2010, 2(1), 298-313; https://doi.org/10.3390/v2010298 - 22 Jan 2010
Cited by 91 | Viewed by 16740
Abstract
Tumor suppressor p53 is widely known as ‘the guardian of the genome’ due to its ability to prevent the emergence of transformed cells by the induction of cell cycle arrest and apoptosis. However, recent studies indicate that p53 is also a direct transcriptional [...] Read more.
Tumor suppressor p53 is widely known as ‘the guardian of the genome’ due to its ability to prevent the emergence of transformed cells by the induction of cell cycle arrest and apoptosis. However, recent studies indicate that p53 is also a direct transcriptional target of type I interferons (IFNs) and thus, it is activated by these cytokines upon viral infection. p53 has been shown to contribute to virus-induced apoptosis, therefore dampening the ability of a wide range of viruses to replicate and spread. Interestingly, recent studies also indicate that several IFN-inducible genes such as interferon regulatory factor 9 (IRF9), IRF5, IFN-stimulated gene 15 (ISG15) and toll-like receptor 3 (TLR3) are in fact, p53 direct transcriptional targets. These findings indicate that p53 may play a key role in antiviral innate immunity by both inducing apoptosis in response to viral infection, and enforcing the type I IFN response, and provide a new insight into the evolutionary reasons why many viruses encode p53 antagonistic proteins. Full article
(This article belongs to the Special Issue Interferon Antiviral Response and Viral Evasion)
Show Figures

Graphical abstract

107 KiB  
Review
The Development of an AIDS Mucosal Vaccine
by Xian Tang and Zhiwei Chen
Viruses 2010, 2(1), 283-297; https://doi.org/10.3390/v2010283 - 22 Jan 2010
Cited by 5 | Viewed by 11807
Abstract
It is well known that mucosal tissues contain the largest surface area of the human body and are the front line of natural host defense against various pathogens. In fact, more than 80% of infectious disease pathogens probably gain entry into the susceptible [...] Read more.
It is well known that mucosal tissues contain the largest surface area of the human body and are the front line of natural host defense against various pathogens. In fact, more than 80% of infectious disease pathogens probably gain entry into the susceptible human hosts through open mucosal surfaces. Human immunodeficiency virus type one (HIV-1), a mainly sexually transmitted virus, also primarily targets the vaginal and gastrointestinal mucosa as entry sites for viral transmission, seeding, replication and amplification. Since HIV-1 establishes its early replication in vaginal or rectal mucosal tissues, the induction of sufficient mucosal immunity at the initial site of HIV-1 transmission becomes essential for a protective vaccine. However, despite the fact that current conventional vaccine strategies have remained unsuccessful in preventing HIV-1 infection, sufficient financial support and resources have yet to be given to develop a vaccine able to elicit protective mucosal immunity against sexual transmissions. Interestingly, Chinese ancestors invented variolation through intranasal administration about one thousand years ago, which led to the discovery of a successful smallpox vaccine and the final eradication of the disease. It is the hope for all mankind that the development of a mucosal AIDS vaccine will ultimately help control the AIDS pandemic. In order to discover an effective mucosal AIDS vaccine, it is necessary to have a deep understanding of mucosal immunology and to test various mucosal vaccination strategies. Full article
(This article belongs to the Special Issue AIDS Vaccine)
352 KiB  
Review
Evasion of the Interferon-Mediated Antiviral Response by Filoviruses
by Washington B. Cárdenas
Viruses 2010, 2(1), 262-282; https://doi.org/10.3390/v2010262 - 21 Jan 2010
Cited by 9 | Viewed by 14764
Abstract
The members of the filoviruses are recognized as some of the most lethal viruses affecting human and non-human primates. The only two genera of the Filoviridae family, Marburg virus (MARV) and Ebola virus (EBOV), comprise the main etiologic agents of severe hemorrhagic fever [...] Read more.
The members of the filoviruses are recognized as some of the most lethal viruses affecting human and non-human primates. The only two genera of the Filoviridae family, Marburg virus (MARV) and Ebola virus (EBOV), comprise the main etiologic agents of severe hemorrhagic fever outbreaks in central Africa, with case fatality rates ranging from 25 to 90%. Fatal outcomes have been associated with a late and dysregulated immune response to infection, very likely due to the virus targeting key host immune cells, such as macrophages and dendritic cells (DCs) that are necessary to mediate effective innate and adaptive immune responses. Despite major progress in the development of vaccine candidates for filovirus infections, a licensed vaccine or therapy for human use is still not available. During the last ten years, important progress has been made in understanding the molecular mechanisms of filovirus pathogenesis. Several lines of evidence implicate the impairment of the host interferon (IFN) antiviral innate immune response by MARV or EBOV as an important determinant of virulence. In vitro and in vivo experimental infections with recombinant Zaire Ebola virus (ZEBOV), the best characterized filovirus, demonstrated that the viral protein VP35 plays a key role in inhibiting the production of IFN-α/β. Further, the action of VP35 is synergized by the inhibition of cellular responses to IFN-α/β by the minor matrix viral protein VP24. The dual action of these viral proteins may contribute to an efficient initial virus replication and dissemination in the host. Noticeably, the analogous function of these viral proteins in MARV has not been reported. Because the IFN response is a major component of the innate immune response to virus infection, this chapter reviews recent findings on the molecular mechanisms of IFN-mediated antiviral evasion by filovirus infection. Full article
(This article belongs to the Special Issue Interferon Antiviral Response and Viral Evasion)
Show Figures

Graphical abstract

130 KiB  
Review
Virus Infection Recognition and Early Innate Responses to Non-Enveloped Viral Vectors
by Dmitry M. Shayakhmetov
Viruses 2010, 2(1), 244-261; https://doi.org/10.3390/v2010244 - 19 Jan 2010
Cited by 8 | Viewed by 11281
Abstract
Numerous human genetic and acquired diseases could be corrected or ameliorated if viruses are harnessed to safely and effectively deliver therapeutic genes to diseased cells and tissues in vivo. Innate immune and inflammatory response represents one of the key stumbling blocks during [...] Read more.
Numerous human genetic and acquired diseases could be corrected or ameliorated if viruses are harnessed to safely and effectively deliver therapeutic genes to diseased cells and tissues in vivo. Innate immune and inflammatory response represents one of the key stumbling blocks during the development of viral-based therapies. In this review, current data on the early innate immune responses to viruses and to the most commonly used gene therapy vectors (using adenovirus and adeno-associated virus) will be discussed. Recent findings in the field may help develop new approaches to moderate these innate immune anti-viral responses and thus improve the safety of viral vectors for human gene therapy applications. Full article
(This article belongs to the Special Issue Novel Viral Vector Systems for Gene Therapy)
Show Figures

Figure 1

623 KiB  
Review
Initiation of HIV Reverse Transcription
by Catherine Isel, Chantal Ehresmann and Roland Marquet
Viruses 2010, 2(1), 213-243; https://doi.org/10.3390/v2010213 - 18 Jan 2010
Cited by 37 | Viewed by 14639
Abstract
Reverse transcription of retroviral genomes into double stranded DNA is a key event for viral replication. The very first stage of HIV reverse transcription, the initiation step, involves viral and cellular partners that are selectively packaged into the viral particle, leading to an [...] Read more.
Reverse transcription of retroviral genomes into double stranded DNA is a key event for viral replication. The very first stage of HIV reverse transcription, the initiation step, involves viral and cellular partners that are selectively packaged into the viral particle, leading to an RNA/protein complex with very specific structural and functional features, some of which being, in the case of HIV-1, linked to particular isolates. Recent understanding of the tight spatio-temporal regulation of reverse transcription and its importance for viral infectivity further points toward reverse transcription and potentially its initiation step as an important drug target. Full article
(This article belongs to the Special Issue Retroviral Enzymes)
Show Figures

Figure 1

224 KiB  
Review
Interaction of Host Cellular Proteins with Components of the Hepatitis Delta Virus
by Valerie Greco-Stewart and Martin Pelchat
Viruses 2010, 2(1), 189-212; https://doi.org/10.3390/v2010189 - 18 Jan 2010
Cited by 40 | Viewed by 14033
Abstract
The hepatitis delta virus (HDV) is the smallest known RNA pathogen capable of propagation in the human host and causes substantial global morbidity and mortality. Due to its small size and limited protein coding capacity, HDV is exquisitely reliant upon host cellular proteins [...] Read more.
The hepatitis delta virus (HDV) is the smallest known RNA pathogen capable of propagation in the human host and causes substantial global morbidity and mortality. Due to its small size and limited protein coding capacity, HDV is exquisitely reliant upon host cellular proteins to facilitate its transcription and replication. Remarkably, HDV does not encode an RNA-dependent RNA polymerase which is traditionally required to catalyze RNA-templated RNA synthesis. Furthermore, HDV lacks enzymes responsible for post-transcriptional and -translational modification, processes which are integral to the HDV life cycle. This review summarizes the known HDV-interacting proteins and discusses their significance in HDV biology. Full article
(This article belongs to the Special Issue Subviral RNAs)
Show Figures

Figure 1

179 KiB  
Review
Targets for the Induction of Protective Immunity Against Influenza A Viruses
by Rogier Bodewes, Albert D.M.E. Osterhaus and Guus F. Rimmelzwaan
Viruses 2010, 2(1), 166-188; https://doi.org/10.3390/v2010166 - 14 Jan 2010
Cited by 11 | Viewed by 12292
Abstract
The current pandemic caused by the new influenza A(H1N1) virus of swine origin and the current pandemic threat caused by the highly pathogenic avian influenza A viruses of the H5N1 subtype have renewed the interest in the development of vaccines that can induce [...] Read more.
The current pandemic caused by the new influenza A(H1N1) virus of swine origin and the current pandemic threat caused by the highly pathogenic avian influenza A viruses of the H5N1 subtype have renewed the interest in the development of vaccines that can induce broad protective immunity. Preferably, vaccines not only provide protection against the homologous strains, but also against heterologous strains, even of another subtype. Here we describe viral targets and the arms of the immune response involved in protection against influenza virus infections such as antibodies directed against the hemagglutinin, neuraminidase and the M2 protein and cellular immune responses directed against the internal viral proteins. Full article
(This article belongs to the Special Issue Influenza: Pandemics and Vaccinations)
Show Figures

Graphical abstract

1020 KiB  
Review
Comparative Studies on Retroviral Proteases: Substrate Specificity
by József Tözsér
Viruses 2010, 2(1), 147-165; https://doi.org/10.3390/v2010147 - 14 Jan 2010
Cited by 35 | Viewed by 14139
Abstract
Exogenous retroviruses are subclassified into seven genera and include viruses that cause diseases in humans. The viral Gag and Gag-Pro-Pol polyproteins are processed by the retroviral protease in the last stage of replication and inhibitors of the HIV-1 protease are widely used in [...] Read more.
Exogenous retroviruses are subclassified into seven genera and include viruses that cause diseases in humans. The viral Gag and Gag-Pro-Pol polyproteins are processed by the retroviral protease in the last stage of replication and inhibitors of the HIV-1 protease are widely used in AIDS therapy. Resistant mutations occur in response to the drug therapy introducing residues that are frequently found in the equivalent position of other retroviral proteases. Therefore, besides helping to understand the general and specific features of these enzymes, comparative studies of retroviral proteases may help to understand the mutational capacity of the HIV-1 protease. Full article
(This article belongs to the Special Issue Retroviral Enzymes)
Show Figures

Graphical abstract

530 KiB  
Review
RNA Editing and its Control in Hepatitis Delta Virus Replication
by Renxiang Chen, Sarah D. Linnstaedt and John L. Casey
Viruses 2010, 2(1), 131-146; https://doi.org/10.3390/v2010131 - 12 Jan 2010
Cited by 9 | Viewed by 12737
Abstract
The hepatitis delta virus genome is a small circular RNA, similar to viroids. Although HDV contains a gene, the protein produced (HDAg) is encoded by less than half the genome and possesses no RNA polymerase activity. Because of this limited coding capacity, HDV [...] Read more.
The hepatitis delta virus genome is a small circular RNA, similar to viroids. Although HDV contains a gene, the protein produced (HDAg) is encoded by less than half the genome and possesses no RNA polymerase activity. Because of this limited coding capacity, HDV relies heavily on host functions and on structural features of the viral RNA—very much like viroids. The virus’ use of host RNA editing activity to produce two functionally distinct forms of HDAg is a particularly good example of this reliance. This review covers the mechanisms and control of RNA editing in the HDV replication cycle. Full article
(This article belongs to the Special Issue Subviral RNAs)
Show Figures

Figure 1

194 KiB  
Review
Retroviral Integration Site Selection
by Sébastien Desfarges and Angela Ciuffi
Viruses 2010, 2(1), 111-130; https://doi.org/10.3390/v2010111 - 12 Jan 2010
Cited by 52 | Viewed by 17021
Abstract
The stable insertion of a copy of their genome into the host cell genome is an essential step of the life cycle of retroviruses. The site of viral DNA integration, mediated by the viral-encoded integrase enzyme, has important consequences for both the virus [...] Read more.
The stable insertion of a copy of their genome into the host cell genome is an essential step of the life cycle of retroviruses. The site of viral DNA integration, mediated by the viral-encoded integrase enzyme, has important consequences for both the virus and the host cell. The analysis of retroviral integration site distribution was facilitated by the availability of the human genome sequence, revealing the non-random feature of integration site selection and identifying different favored and disfavored genomic locations for individual retroviruses. This review will summarize the current knowledge about retroviral differences in their integration site preferences as well as the mechanisms involved in this process. Full article
(This article belongs to the Special Issue Retroviral Enzymes)
Show Figures

Graphical abstract

248 KiB  
Commentary
Correlative Structural Biology: How to Investigate the Fine Details of Viral Structure
by Elizabeth R. Wright
Viruses 2010, 2(1), 107-110; https://doi.org/10.3390/v2010107 - 11 Jan 2010
Cited by 1 | Viewed by 8736
Abstract
Commentary on Byeon, I.J.; Meng, X.; Jung, J.; Zhao, G.; Yang, R.; Ahn, J.; Shi, J.; Concel, J.; Aiken, C.; Zhang, P.; Gronenborn, A.M. Structural convergence between Cryo-EM and NMR reveals intersubunit interactions critical for HIV-1 capsid function. Cell 2009, 139, 780-790. Full article
Show Figures

Figure 1

351 KiB  
Review
Oncolytic Viruses for Cancer Therapy: Overcoming the Obstacles
by Han Hsi Wong, Nicholas R. Lemoine and Yaohe Wang
Viruses 2010, 2(1), 78-106; https://doi.org/10.3390/v2010078 - 11 Jan 2010
Cited by 134 | Viewed by 27525
Abstract
Targeted therapy of cancer using oncolytic viruses has generated much interest over the past few years in the light of the limited efficacy and side effects of standard cancer therapeutics for advanced disease. In 2006, the world witnessed the first government-approved oncolytic virus [...] Read more.
Targeted therapy of cancer using oncolytic viruses has generated much interest over the past few years in the light of the limited efficacy and side effects of standard cancer therapeutics for advanced disease. In 2006, the world witnessed the first government-approved oncolytic virus for the treatment of head and neck cancer. It has been known for many years that viruses have the ability to replicate in and lyse cancer cells. Although encouraging results have been demonstrated in vitro and in animal models, most oncolytic viruses have failed to impress in the clinical setting. The explanation is multifactorial, determined by the complex interactions between the tumor and its microenvironment, the virus, and the host immune response. This review focuses on discussion of the obstacles that oncolytic virotherapy faces and recent advances made to overcome them, with particular reference to adenoviruses. Full article
(This article belongs to the Special Issue Novel Viral Vector Systems for Gene Therapy)
Show Figures

Figure 1

35 KiB  
Communication
Preliminary Report on HIV-1 Vaccine Preparedness in Nigeria: Advantages of Recruiting University Students
by Abigail Edubio, Simon Agwale, Marc Bulterys, Dadik Jelpe, John Idoko, Chris Isichei, Ruth Guyit and Alash’le Abimiku
Viruses 2010, 2(1), 73-77; https://doi.org/10.3390/v2010073 - 11 Jan 2010
Cited by 2 | Viewed by 10828
Abstract
The national HIV seroprevalence in Nigeria has risen steeply from about 3% in 1993 to 5-8% in 2001 and now stands at 4.4%. HIV epidemic continues to be a serious threat to the most populous country in Africa with a population of 140 [...] Read more.
The national HIV seroprevalence in Nigeria has risen steeply from about 3% in 1993 to 5-8% in 2001 and now stands at 4.4%. HIV epidemic continues to be a serious threat to the most populous country in Africa with a population of 140 million, with limited use of antiviral drugs that is taken for life since it only suppresses the virus without completely eliminating the virus or leading to cure. Only a change in social behavior and an affordable vaccine can halt the epidemic in Africa. We report here results of a pilot study on the recruitment strategies, sociodemographic aspects and HIV risk behavior of a cohort of normal volunteers recruited at the University of Jos, Nigeria. Our study recorded a high degree of interest and zeal to participate in HIV vaccine studies by volunteers, and demonstrated the superiority of snowballing over invitation by mail, as a recruitment strategy. A cohort of university students may be particularly suitable for conducting HIV vaccine trials because of the assurance of prospective follow-up for up to four years (time to graduation), and a good understanding of the risks and benefits of participation as outlined in the informed consent. We had 100% retention during a follow-up period of two years. Most importantly, the cohort reflected a relatively low HIV seroprevalence, which gives preventive programs the potential to blunt or halt the epidemic. Full article
(This article belongs to the Special Issue AIDS Vaccine)
3182 KiB  
Review
The IKK Kinases: Operators of Antiviral Signaling
by Alissa M. Pham and Benjamin R. TenOever
Viruses 2010, 2(1), 55-72; https://doi.org/10.3390/v2010055 - 08 Jan 2010
Cited by 21 | Viewed by 12930
Abstract
The ability of a cell to combat an intracellular pathogen requires a mechanism to recognize the threat and elicit a transcriptional response against it. In the context of virus infection, the cell must take measures to inhibit viral replication, meanwhile, convey warning signals [...] Read more.
The ability of a cell to combat an intracellular pathogen requires a mechanism to recognize the threat and elicit a transcriptional response against it. In the context of virus infection, the cell must take measures to inhibit viral replication, meanwhile, convey warning signals to neighboring cells of the imminent threat. This immune response is predominantly mediated by the production of cytokines, notably, interferon beta (IFNβ). IFNβ signaling results in the transcriptional induction of over one hundred antiviral gene products whose timely expression renders infected cells more capable of inhibiting virus replication, while providing the uninfected cells with the reinforcements to generate a less permissive cellular environment. Induction of IFNβ and many aspects of the antiviral response pivot on the function of the IKK and IKK-related kinases. Despite sharing high levels of homology and some degree of functional redundancy, the classic IKK kinases: IKKα and IKKβ, and the IKK-related kinases: TBK1 and IKKε, perform distinct roles in regulating the host antiviral defense. These kinases serve as molecular operators in their cooperative ability to integrate incoming cellular cues and act on a range of essential antiviral transcription factors to reshape the cellular transcriptome during infection. Full article
(This article belongs to the Special Issue Interferon Antiviral Response and Viral Evasion)
Show Figures

Figure 1

294 KiB  
Article
HIV-1 Subtype C Phylodynamics in the Global Epidemic
by Vlad Novitsky, Rui Wang, Stephen Lagakos and Max Essex
Viruses 2010, 2(1), 33-54; https://doi.org/10.3390/v2010033 - 07 Jan 2010
Cited by 23 | Viewed by 14554
Abstract
The diversity of HIV-1 and its propensity to generate escape mutants present fundamental challenges to control efforts, including HIV vaccine design. Intra-host diversification of HIV is determined by immune responses elicited by an HIV-infected individual over the course of the infection. Complex and [...] Read more.
The diversity of HIV-1 and its propensity to generate escape mutants present fundamental challenges to control efforts, including HIV vaccine design. Intra-host diversification of HIV is determined by immune responses elicited by an HIV-infected individual over the course of the infection. Complex and dynamic patterns of transmission of HIV lead to an even more complex population viral diversity over time, thus presenting enormous challenges to vaccine development. To address inter-patient viral evolution over time, a set of 653 unique HIV-1 subtype C gag sequences were retrieved from the LANL HIV Database, grouped by sampling year as <2000, 2000, 2001–2002, 2003, and 2004–2006, and analyzed for the site-specific frequency of translated amino acid residues. Phylogenetic analysis revealed that a total of 289 out of 653 (44.3%) analyzed sequences were found within 16 clusters defined by aLRT of more than 0.90. Median (IQR) inter-sample diversity of analyzed gag sequences was 8.7% (7.7%; 9.8%). Despite the heterogeneous origins of analyzed sequences, the gamut and frequency of amino acid residues in wild-type Gag were remarkably stable over the last decade of the HIV-1 subtype C epidemic. The vast majority of amino acid residues demonstrated minor frequency fluctuation over time, consistent with the conservative nature of the HIV-1 Gag protein. Only 4.0% (20 out of 500; HXB2 numbering) amino acid residues across Gag displayed both statistically significant (p<0.05 by both a trend test and heterogeneity test) changes in amino acid frequency over time as well as a range of at least 10% in the frequency of the major amino acid. A total of 59.2% of amino acid residues with changing frequency of 10%+ were found within previously identified CTL epitopes. The time of the most recent common ancestor of the HIV-1 subtype C was dated to around 1950 (95% HPD from 1928 to 1962). This study provides evidence for the overall stability of HIV-1 subtype C Gag among viruses circulating in the epidemic over the last decade. However selected sites across HIV-1C Gag with changing amino acid frequency are likely to be under selection pressure at the population level. Full article
(This article belongs to the Special Issue AIDS Vaccine)
Show Figures

Figure 1

201 KiB  
Commentary
Nucleoproteins of Negative Strand RNA Viruses; RNA Binding, Oligomerisation and Binding to Polymerase Co-Factor
by Rob W. H. Ruigrok and Thibaut Crépin
Viruses 2010, 2(1), 27-32; https://doi.org/10.3390/v2010027 - 07 Jan 2010
Cited by 19 | Viewed by 10033
Abstract
Commentary on Tawar, R.G.; Duquerroy, S.; Vonrhein, C.; Varela, P.F.; Damier-Piolle, L.; Castagné, N.; MacLellan, K.; Bedouelle, H.; Bricogne, G.; Bhella, D.; Eléouët, J.-F.; Rey, F.A. Crystal structure of a nucleocapsid-like nucleoprotein-RNA complex of respiratory syncytial virus. Science 2009, 326, 1279-1283. Full article
Show Figures

Figure 1

485 KiB  
Review
Applying Genomic and Bioinformatic Resources to Human Adenovirus Genomes for Use in Vaccine Development and for Applications in Vector Development for Gene Delivery
by Jason Seto, Michael P. Walsh, Padmanabhan Mahadevan, Qiwei Zhang and Donald Seto
Viruses 2010, 2(1), 1-26; https://doi.org/10.3390/v2010001 - 06 Jan 2010
Cited by 15 | Viewed by 13185
Abstract
Technological advances and increasingly cost-effect methodologies in DNA sequencing and computational analysis are providing genome and proteome data for human adenovirus research. Applying these tools, data and derived knowledge to the development of vaccines against these pathogens will provide effective prophylactics. The same [...] Read more.
Technological advances and increasingly cost-effect methodologies in DNA sequencing and computational analysis are providing genome and proteome data for human adenovirus research. Applying these tools, data and derived knowledge to the development of vaccines against these pathogens will provide effective prophylactics. The same data and approaches can be applied to vector development for gene delivery in gene therapy and vaccine delivery protocols. Examination of several field strain genomes and their analyses provide examples of data that are available using these approaches. An example of the development of HAdV-B3 both as a vaccine and also as a vector is presented. Full article
(This article belongs to the Special Issue Novel Viral Vector Systems for Gene Therapy)
Show Figures

Figure 1

Previous Issue
Next Issue
Back to TopTop