51
|
Coupling viruses to dynein and kinesin-1. EMBO J 2011; 30:3527-39. [PMID: 21878994 DOI: 10.1038/emboj.2011.283] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 07/21/2011] [Indexed: 12/13/2022] Open
Abstract
It is now clear that transport on microtubules by dynein and kinesin family motors has an important if not critical role in the replication and spread of many different viruses. Understanding how viruses hijack dynein and kinesin motors using a limited repertoire of proteins offers a great opportunity to determine the molecular basis of motor recruitment. In this review, we discuss the interactions of dynein and kinesin-1 with adenovirus, the α herpes viruses: herpes simplex virus (HSV1) and pseudorabies virus (PrV), human immunodeficiency virus type 1 (HIV-1) and vaccinia virus. We highlight where the molecular links to these opposite polarity motors have been defined and discuss the difficulties associated with identifying viral binding partners where the basis of motor recruitment remains to be established. Ultimately, studying microtubule-based motility of viruses promises to answer fundamental questions as to how the activity and recruitment of the dynein and kinesin-1 motors are coordinated and regulated during bi-directional transport.
Collapse
|
52
|
Adenovirus recruits dynein by an evolutionary novel mechanism involving direct binding to pH-primed hexon. Viruses 2011; 3:1417-31. [PMID: 21994788 PMCID: PMC3185799 DOI: 10.3390/v3081417] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 08/03/2011] [Accepted: 08/06/2011] [Indexed: 12/19/2022] Open
Abstract
Following receptor-mediated uptake into endocytic vesicles and escape from the endosome, adenovirus is transported by cytoplasmic dynein along microtubules to the perinuclear region of the cell. How motor proteins are recruited to viruses for their own use has begun to be investigated only recently. We review here the evidence for a role for dynein and other motor proteins in adenovirus infectivity. We also discuss the implications of recent studies on the mechanism of dynein recruitment to adenovirus for understanding the relationship between pathogenic and physiological cargo recruitment and for the evolutionary origins of dynein-mediated adenovirus transport.
Collapse
|
53
|
Merino-Gracia J, García-Mayoral MF, Rodríguez-Crespo I. The association of viral proteins with host cell dynein components during virus infection. FEBS J 2011; 278:2997-3011. [PMID: 21777384 PMCID: PMC7164101 DOI: 10.1111/j.1742-4658.2011.08252.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
After fusion with the cellular plasma membrane or endosomal membranes, viral particles are generally too large to diffuse freely within the crowded cytoplasm environment. Thus, they will never reach the cell nucleus or the perinuclear areas where replication or reverse transcription usually takes place. It has been proposed that many unrelated viruses are transported along microtubules in a retrograde manner using the cellular dynein machinery or, at least, some dynein components. A putative employment of the dynein motor in a dynein‐mediated transport has been suggested from experiments in which viral capsid proteins were used as bait in yeast two‐hybrid screens using libraries composed of cellular proteins and dynein‐associated chains were retrieved as virus‐interacting proteins. In most cases DYNLL1, DYNLT1 or DYNLRB1 were identified as the dynein chains that interact with viral proteins. The importance of these dynein–virus interactions has been supported, in principle, by the observation that in some cases the dynein‐interacting motifs of viral proteins altered by site‐directed mutagenesis result in non‐infective virions. Furthermore, overexpression of p50 dynamitin, which blocks the dynein–dynactin interaction, or incubation of infected cells with peptides that compete with viral polypeptides for dynein binding have been shown to alter the viral retrograde transport. Still, it remains to be proved that dynein light chains can bind simultaneously to incoming virions and to the dynein motor for retrograde transport to take place. In this review, we will analyse the association of viral proteins with dynein polypeptides and its implications for viral infection.
Collapse
Affiliation(s)
- Javier Merino-Gracia
- Departamento de Bioquímica y Biología Molecular I, Universidad Complutense, Madrid, Spain
| | | | | |
Collapse
|
54
|
Henaff D, Salinas S, Kremer EJ. An adenovirus traffic update: from receptor engagement to the nuclear pore. Future Microbiol 2011; 6:179-92. [PMID: 21366418 DOI: 10.2217/fmb.10.162] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Adenoviruses have a bipolar nature: they are ubiquitous pathogens that occasionally cause life-threatening diseases or they can be engineered into powerful gene transfer vectors. The goal of this article is to summarize the most recent advances in adenovirus receptor engagement, internalization, endosomal maturation, endosomal escape and trafficking to the nuclear pore. A better understanding of this initial part of the adenovirus lifecycle may identify new mechanistic-based treatments for adenovirus-induced diseases and help in the engineering of more efficient vectors.
Collapse
Affiliation(s)
- Daniel Henaff
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, 34293 Montpellier, France
| | | | | |
Collapse
|
55
|
Corjon S, Gonzalez G, Henning P, Grichine A, Lindholm L, Boulanger P, Fender P, Hong SS. Cell entry and trafficking of human adenovirus bound to blood factor X is determined by the fiber serotype and not hexon:heparan sulfate interaction. PLoS One 2011; 6:e18205. [PMID: 21637339 PMCID: PMC3102659 DOI: 10.1371/journal.pone.0018205] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 02/28/2011] [Indexed: 01/29/2023] Open
Abstract
Human adenovirus serotype 5 (HAdV5)-based vectors administered intravenously accumulate in the liver as the result of their direct binding to blood coagulation factor X (FX) and subsequent interaction of the FX-HAdV5 complex with heparan sulfate proteoglycan (HSPG) at the surface of liver cells. Intriguingly, the serotype 35 fiber-pseudotyped vector HAdV5F35 has liver transduction efficiencies 4-logs lower than HAdV5, even though both vectors carry the same hexon capsomeres. In order to reconcile this apparent paradox, we investigated the possible role of other viral capsid proteins on the FX/HSPG-mediated cellular uptake of HAdV5-based vectors. Using CAR- and CD46-negative CHO cells varying in HSPG expression, we confirmed that FX bound to serotype 5 hexon protein and to HAdV5 and HAdV5F35 virions via its Gla-domain, and enhanced the binding of both vectors to surface-immobilized hypersulfated heparin and cellular HSPG. Using penton mutants, we found that the positive effect of FX on HAdV5 binding to HSPG and cell transduction did not depend on the penton base RGD and fiber shaft KKTK motifs. However, we found that FX had no enhancing effect on the HAdV5F35-mediated cell transduction, but a negative effect which did not involve the cell attachment or endocytic step, but the intracellular trafficking and nuclear import of the FX-HAdV5F35 complex. By cellular imaging, HAdV5F35 particles were observed to accumulate in the late endosomal compartment, and were released in significant amounts into the extracellular medium via exocytosis. We showed that the stability of serotype 5 hexon:FX interaction was higher at low pH compared to neutral pH, which could account for the retention of FX-HAdV5F35 complexes in the late endosomes. Our results suggested that, despite the high affinity interaction of hexon capsomeres to FX and cell surface HSPG, the adenoviral fiber acted as the dominant determinant of the internalization and trafficking pathway of HAdV5-based vectors.
Collapse
Affiliation(s)
- Stéphanie Corjon
- University Lyon 1, INRA UMR 754, Retrovirus
and Comparative Pathology, Lyon, France
| | - Gaëlle Gonzalez
- University Lyon 1, INRA UMR 754, Retrovirus
and Comparative Pathology, Lyon, France
| | - Petra Henning
- Department of Microbiology and Immunology,
University of Göteborg, Institute for Biomedicine, Göteborg,
Sweden
| | - Alexei Grichine
- Institut Albert Bonniot, CRI INSERM-UJF U-823,
La Tronche, France
| | | | - Pierre Boulanger
- University Lyon 1, INRA UMR 754, Retrovirus
and Comparative Pathology, Lyon, France
| | - Pascal Fender
- Unit for Virus-Host Interaction, UMI-3265,
CNRS-EMBL-UJF, Grenoble, France
| | - Saw-See Hong
- University Lyon 1, INRA UMR 754, Retrovirus
and Comparative Pathology, Lyon, France
| |
Collapse
|
56
|
Xu CH, Sui MH, Tang JB, Shen YQ. What can we learn from virus in designing nonviral gene vectors. CHINESE JOURNAL OF POLYMER SCIENCE 2011. [DOI: 10.1007/s10118-011-1047-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
57
|
The dynactin complex enhances the speed of microtubule-dependent motions of adenovirus both towards and away from the nucleus. Viruses 2011; 3:233-253. [PMID: 21994728 PMCID: PMC3185697 DOI: 10.3390/v3030233] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/28/2011] [Indexed: 12/29/2022] Open
Abstract
Unlike transport vesicles or organelles, human adenovirus (HAdV) directly binds to the microtubule minus end-directed motor dynein for transport to the nucleus. The dynein cofactor dynactin enhances nuclear transport of HAdV and boosts infection. To determine if dynactin has a specific role in cytoplasmic trafficking of incoming HAdV on microtubules, we used live cell spinning disc confocal microscopy at 25 Hz acquisition frequency and automated tracking of single virus particles at 20–50 nm spatial resolution. Computational dissection by machine-learning algorithms extracted specific motion patterns of viral trajectories. We found that unperturbed cells supported two kinds of microtubule-dependent motions, directed motions (DM) and fast drifts (FD). DM had speeds of 0.2 to 2 μm/s and run lengths of 0.4 up to 7 μm, while FD were slower and less extensive at 0.02 to 0.4 μm/s and 0.05 to 2.5 μm. Dynactin interference by overexpression of p50/dynamitin or a coiled-coil domain of p150/Glued reduced the speeds and amounts of both center- and periphery-directed DM but not FD, and inhibited infection. These results indicate that dynactin enhances adenovirus infection by increasing the speed and efficiency of dynein-mediated virus motion to the nucleus, and, surprisingly, also supports a hereto unknown motor activity for virus transport to the cell periphery.
Collapse
|
58
|
Nyarko A, Barbar E. Light chain-dependent self-association of dynein intermediate chain. J Biol Chem 2011; 286:1556-66. [PMID: 20974845 PMCID: PMC3020764 DOI: 10.1074/jbc.m110.171686] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 10/15/2010] [Indexed: 11/06/2022] Open
Abstract
Dynein light chains are bivalent dimers that bind two copies of dynein intermediate chain IC to form a cargo attachment subcomplex. The interaction of light chain LC8 with the natively disordered N-terminal domain of IC induces helix formation at distant IC sites in or near a region predicted to form a coiled-coil. This fostered the hypothesis that LC8 binding promotes IC self-association to form a coiled-coil or other interchain helical structure. However, recent studies show that the predicted coiled-coil sequence partially overlaps the light chain LC7 recognition sequence on IC, raising questions about the apparently contradictory effects of LC8 and LC7. Here, we use NMR and fluorescence quenching to localize IC self-association to residues within the predicted coiled-coil that also correspond to helix 1 of the LC7 recognition sequence. LC8 binding promotes IC self-association of helix 1 from each of two IC chains, whereas LC7 binding reverses self-association by incorporating the same residues into two symmetrical, but distant, helices of the LC7-IC complex. Isothermal titration experiments confirm the distinction of LC8 enhancement of IC self-association and LC7 binding effects. When all three light chains are bound, IC self-association is shifted to another region. Such flexibility in association modes may function in maintaining a stable and versatile light chain-intermediate chain assembly under changing cellular conditions.
Collapse
Affiliation(s)
- Afua Nyarko
- From the Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331
| | - Elisar Barbar
- From the Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon 97331
| |
Collapse
|
59
|
Cohen S, Au S, Panté N. How viruses access the nucleus. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:1634-45. [PMID: 21167871 DOI: 10.1016/j.bbamcr.2010.12.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 11/24/2010] [Accepted: 12/08/2010] [Indexed: 10/25/2022]
Abstract
Many viruses depend on nuclear proteins for replication. Therefore, their viral genome must enter the nucleus of the host cell. In this review we briefly summarize the principles of nucleocytoplasmic transport, and then describe the diverse strategies used by viruses to deliver their genomes into the host nucleus. Some of the emerging mechanisms include: (1) nuclear entry during mitosis, when the nuclear envelope is disassembled, (2) viral genome release in the cytoplasm followed by entry of the genome through the nuclear pore complex (NPC), (3) capsid docking at the cytoplasmic side of the NPC, followed by genome release, (4) nuclear entry of intact capsids through the NPC, followed by genome release, and (5) nuclear entry via virus-induced disruption of the nuclear envelope. Which mechanism a particular virus uses depends on the size and structure of the virus, as well as the cellular cues used by the virus to trigger capsid disassembly and genome release. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.
Collapse
Affiliation(s)
- Sarah Cohen
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
60
|
Tropism-modification strategies for targeted gene delivery using adenoviral vectors. Viruses 2010; 2:2290-2355. [PMID: 21994621 PMCID: PMC3185574 DOI: 10.3390/v2102290] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/07/2010] [Indexed: 02/08/2023] Open
Abstract
Achieving high efficiency, targeted gene delivery with adenoviral vectors is a long-standing goal in the field of clinical gene therapy. To achieve this, platform vectors must combine efficient retargeting strategies with detargeting modifications to ablate native receptor binding (i.e. CAR/integrins/heparan sulfate proteoglycans) and “bridging” interactions. “Bridging” interactions refer to coagulation factor binding, namely coagulation factor X (FX), which bridges hepatocyte transduction in vivo through engagement with surface expressed heparan sulfate proteoglycans (HSPGs). These interactions can contribute to the off-target sequestration of Ad5 in the liver and its characteristic dose-limiting hepatotoxicity, thereby significantly limiting the in vivo targeting efficiency and clinical potential of Ad5-based therapeutics. To date, various approaches to retargeting adenoviruses (Ad) have been described. These include genetic modification strategies to incorporate peptide ligands (within fiber knob domain, fiber shaft, penton base, pIX or hexon), pseudotyping of capsid proteins to include whole fiber substitutions or fiber knob chimeras, pseudotyping with non-human Ad species or with capsid proteins derived from other viral families, hexon hypervariable region (HVR) substitutions and adapter-based conjugation/crosslinking of scFv, growth factors or monoclonal antibodies directed against surface-expressed target antigens. In order to maximize retargeting, strategies which permit detargeting from undesirable interactions between the Ad capsid and components of the circulatory system (e.g. coagulation factors, erythrocytes, pre-existing neutralizing antibodies), can be employed simultaneously. Detargeting can be achieved by genetic ablation of native receptor-binding determinants, ablation of “bridging interactions” such as those which occur between the hexon of Ad5 and coagulation factor X (FX), or alternatively, through the use of polymer-coated “stealth” vectors which avoid these interactions. Simultaneous retargeting and detargeting can be achieved by combining multiple genetic and/or chemical modifications.
Collapse
|
61
|
Lan D, Tang C, Li M, Yue H. Screening and identification of differentially expressed genes from chickens infected with Newcastle disease virus by suppression subtractive hybridization. Avian Pathol 2010; 39:151-9. [PMID: 20544419 DOI: 10.1080/03079451003716383] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Newcastle disease is an important viral infectious disease caused by Newcastle disease virus (NDV), which leads to severe economic losses in the poultry industry worldwide. The molecular mechanisms involved in the pathogenesis of NDV and the host-directed antiviral responses remain poorly understood. In this study, we screened and identified the differentially expressed transcripts from chicken spleen 36 h post NDV infection using suppression subtractive hybridization (SSH). From the SSH library, we obtained 140 significant differentially expressed sequence tags (ESTs), which could be divided into three categories: high homology genes (58), high homology ESTs (62) and novel ESTs (20). The 58 high homology genes could be grouped into nine clusters based on the best known function of their protein products, which involved signalling transduction (HSPC166, PDE7B, GRIA4, GARNL1), transcriptional regulation (ANP32A, LOC423724, SATB1, QKI, ETV6), cellular molecular dynamics (MYLK, MYO7A, DCTN6), cytoskeleton (LAMA4, LAMC1, COL4A1), stress response (DNAJC15, CIRBP), immune response (TIA1, TOX, CMIP), metabolism (RPS15A, RPL32, GLUT8, CYPR21, DPYD, LOC417295), oxidation-reduction (TXN, MSRB3, GCLC), and others. In addition, we found that the 20 novel ESTs provide a clue for the discovery of some new genes associated with infection. In summary, our findings demonstrate previously unrecognized changes in gene transcription that are associated with NDV infection in vivo, and many differentially expressed genes identified in the study clearly merit further investigation. Our data provide new insights into better understanding the molecular mechanism of host-NDV interaction.
Collapse
Affiliation(s)
- Daoliang Lan
- College of Life Science and Technology, Southwest University for Nationality, Chengdu, People's Republic of China
| | | | | | | |
Collapse
|
62
|
Chailertvanitkul VA, Pouton CW. Adenovirus: a blueprint for non-viral gene delivery. Curr Opin Biotechnol 2010; 21:627-32. [PMID: 20638266 DOI: 10.1016/j.copbio.2010.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 06/22/2010] [Accepted: 06/23/2010] [Indexed: 02/02/2023]
Abstract
Although adenoviral vectors may not find a direct clinical role in gene therapy, an understanding of the mechanisms of DNA delivery that adenoviruses use is of vital importance to the design of next-generation non-viral gene delivery systems. Adenoviruses overcome a series of biological barriers, including endosomal escape, intracellular trafficking, capsid dissociation, and nuclear import of DNA, to deliver their genome to the host cell nucleus. The understanding of these processes at the molecular level is progressing and is set to inform the design of synthetic gene delivery systems.
Collapse
Affiliation(s)
- V Ann Chailertvanitkul
- Medicinal Chemistry and Drug Action, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), Melbourne, Australia
| | | |
Collapse
|
63
|
Particle tracking of intracellular trafficking of octaarginine-modified liposomes: a comparative study with adenovirus. Mol Ther 2010; 18:955-64. [PMID: 20216528 DOI: 10.1038/mt.2010.33] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is previously reported that octaarginine (R8)-modified liposome (R8-Lip) was taken up via macropinocytosis, and subsequently delivered to the nuclear periphery. In the present study, we investigated the mechanism for the cytoplasmic transport of R8-Lips, comparing with that for adenovirus. Treatment with microtubule-disruption reagent (nocodazole) inhibited the transfection activity of plasmid DNA (pDNA)-encapsulating R8-Lip more extensively than that of adenovirus. The directional transport of R8-Lips along green fluorescent protein (GFP)-tagged microtubules was observed; however, the velocity was slower than those for adenovirus or endosomes that were devoid of R8-Lips. These directional motions were abrogated in R8-Lips by nocodazole treatment, whereas adenovirus continued to undergo random motion. This finding suggests that the nuclear access of R8-Lip predominantly involves microtubule-dependent transport, whereas an apparent diffusive motion is also operative in nuclear access of adenovirus. Furthermore, quantum dot-labeled pDNA underwent directional motion concomitantly with rhodamine-labeled lipid envelopes, indicating that the R8-Lips were subject to microtubule-dependent transport in the intact form. Dual particle tracking of carriers and endosomes revealed that R8-Lip was directionally transported, associated with endosomes, whereas this occurs after endosomal escape in adenovirus. Collectively, the findings reported herein indicate that vesicular transport is a key factor in the cytoplasmic transport of R8-Lips.
Collapse
|
64
|
Bremner KH, Scherer J, Yi J, Vershinin M, Gross SP, Vallee RB. Adenovirus transport via direct interaction of cytoplasmic dynein with the viral capsid hexon subunit. Cell Host Microbe 2010; 6:523-35. [PMID: 20006841 DOI: 10.1016/j.chom.2009.11.006] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 09/18/2009] [Accepted: 10/19/2009] [Indexed: 10/20/2022]
Abstract
Early in infection, adenovirus travels to the nucleus as a naked capsid using the microtubule motor cytoplasmic dynein. How the dynein complex is recruited to viral cargo remains unclear. We find that cytoplasmic dynein and its associated proteins dynactin and NudE/NudEL, but not LIS1 or ZW10, colocalized with incoming, postendosomal adenovirus particles. However, in contrast to physiological cargos, dynein binding to adenovirus was independent of these dynein-associated proteins. Dynein itself directly interacted through its intermediate and light intermediate chains with the adenovirus capsid subunit hexon in a pH-dependent manner. Expression of hexon or injection of anti-hexon antibody inhibited virus transport but not physiological dynein function. These results identify hexon as a direct receptor for cytoplasmic dynein and demonstrate that hexon recruits dynein for transport to the nucleus by a mechanism distinct from that for physiological dynein cargo.
Collapse
Affiliation(s)
- K Helen Bremner
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
65
|
Abstract
DNA-tumor viruses comprise enveloped and non-enveloped agents that cause malignancies in a large variety of cell types and tissues by interfering with cell cycle control and immortalization. Those DNA-tumor viruses that replicate in the nucleus use cellular mechanisms to transport their genome and newly synthesized viral proteins into the nucleus. This requires cytoplasmic transport and nuclear import of their genome. Agents that employ this strategy include adenoviruses, hepadnaviruses, herpesviruses, and likely also papillomaviruses, and polyomaviruses, but not poxviruses which replicate in the cytoplasm. Here, we discuss how DNA-tumor viruses enter cells, take advantage of cytoplasmic transport, and import their DNA genome through the nuclear pore complex into the nucleus. Remarkably, nuclear import of incoming genomes does not necessarily follow the same pathways used by the structural proteins of the viruses during the replication and assembly phases of the viral life cycle. Understanding the mechanisms of DNA nuclear import can identify new pathways of cell regulation and anti-viral therapies.
Collapse
Affiliation(s)
- Urs F Greber
- Institute of Zoology, University of Zürich, Switzerland
| | | |
Collapse
|
66
|
Frampton AR, Uchida H, von Einem J, Goins WF, Grandi P, Cohen JB, Osterrieder N, Glorioso JC. Equine herpesvirus type 1 (EHV-1) utilizes microtubules, dynein, and ROCK1 to productively infect cells. Vet Microbiol 2009; 141:12-21. [PMID: 19713056 DOI: 10.1016/j.vetmic.2009.07.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 07/05/2009] [Accepted: 07/31/2009] [Indexed: 11/17/2022]
Abstract
To initiate infection, equine herpesvirus type 1 (EHV-1) attaches to heparan sulfate on cell surfaces and then interacts with a putative glycoprotein D receptor(s). After attachment, virus entry occurs either by direct fusion of the virus envelope with the plasma membrane or via endocytosis followed by fusion between the virus envelope and an endosomal membrane. Upon fusion, de-enveloped virus particles are deposited into the cytoplasm and travel to the nucleus for viral replication. In this report, we examined the mechanism of EHV-1 intracellular trafficking and investigated the ability of EHV-1 to utilize specific cellular components to efficiently travel to the nucleus post-entry. Using a panel of microtubule-depolymerizing drugs and inhibitors of microtubule motor proteins, we show that EHV-1 infection is dependent on both the integrity of the microtubule network and the minus-end microtubule motor protein, dynein. In addition, we show that EHV-1 actively induces the acetylation of tubulin, a marker of microtubule stabilization, as early as 15 min post-infection. Finally, our data support a role for the cellular kinase, ROCK1, in virus trafficking to the nucleus.
Collapse
Affiliation(s)
- Arthur R Frampton
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, United States.
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Darr S, Madisch I, Hofmayer S, Rehren F, Heim A. Phylogeny and primary structure analysis of fiber shafts of all human adenovirus types for rational design of adenoviral gene-therapy vectors. J Gen Virol 2009; 90:2849-2854. [PMID: 19656960 DOI: 10.1099/vir.0.014514-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The fiber shaft of human adenoviruses (HAdVs) is essential for bringing the penton base into proximity to the secondary cellular receptor. Fiber shaft sequences of all 53 HAdV types were studied. Phylogeny of the fiber shaft revealed clustering corresponding to the HAdV species concept. An intraspecies recombination hot spot was found at the shaft/knob boundary, a highly conserved sequence stretch. For example, HAdV-D20 clustered with HAdV-D23 in the fiber shaft, but with HAdV-D47 in the fiber knob. Although all shafts exhibited the typical pseudorepeats, amino acid sequence identity was found to be as high as 92 % (interspecies) and 54 % (intraspecies). In contrast to a previous study, a flexibility motif (KXGGLXFD/N) was found in eight HAdV-D types, whereas the putative heparan sulfate-binding site (KKTK) was only found in species HAdV-C. Our results suggest that pseudotyping of gene-therapy vectors at the shaft/knob boundary is feasible, but that flexibility data of shafts should be considered.
Collapse
Affiliation(s)
- Sebastian Darr
- Institut für Virologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Ijad Madisch
- Institut für Virologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Sören Hofmayer
- Institut für Virologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Fabienne Rehren
- Institut für Virologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Albert Heim
- Institut für Virologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| |
Collapse
|
68
|
Wang Y, Gangeswaran R, Zhao X, Wang P, Tysome J, Bhakta V, Yuan M, Chikkanna-Gowda CP, Jiang G, Gao D, Cao F, Francis J, Yu J, Liu K, Yang H, Zhang Y, Zang W, Chelala C, Dong Z, Lemoine N. CEACAM6 attenuates adenovirus infection by antagonizing viral trafficking in cancer cells. J Clin Invest 2009; 119:1604-15. [PMID: 19411761 DOI: 10.1172/jci37905] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 03/04/2009] [Indexed: 12/26/2022] Open
Abstract
The changes in cancer cell surface molecules and intracellular signaling pathways during tumorigenesis make delivery of adenovirus-based cancer therapies inefficient. Here we have identified carcinoembryonic antigen- related cell adhesion molecule 6 (CEACAM6) as a cellular protein that restricts the ability of adenoviral vectors to infect cancer cells. We have demonstrated that CEACAM6 can antagonize the Src signaling pathway, downregulate cancer cell cytoskeleton proteins, and block adenovirus trafficking to the nucleus of human pancreatic cancer cells. Similar to CEACAM6 overexpression, treatment with a Src-selective inhibitor significantly reduced adenovirus replication in these cancer cells and normal human epithelial cells. In a mouse xenograft tumor model, siRNA-mediated knockdown of CEACAM6 also significantly enhanced the antitumor effect of an oncolytic adenovirus. We propose that CEACAM6-associated signaling pathways could be potential targets for the development of biomarkers to predict the response of patients to adenovirus-based therapies, as well as for the development of more potent adenovirus-based therapeutics.
Collapse
Affiliation(s)
- Yaohe Wang
- Centre for Molecular Oncology and Imaging, Institute of Cancer, Barts and The London School of Medicine and Dentistry, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
|
70
|
Salinas S, Bilsland LG, Henaff D, Weston AE, Keriel A, Schiavo G, Kremer EJ. CAR-associated vesicular transport of an adenovirus in motor neuron axons. PLoS Pathog 2009; 5:e1000442. [PMID: 19461877 PMCID: PMC2677547 DOI: 10.1371/journal.ppat.1000442] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 04/24/2009] [Indexed: 12/17/2022] Open
Abstract
Axonal transport is responsible for the movement of signals and cargo between nerve termini and cell bodies. Pathogens also exploit this pathway to enter and exit the central nervous system. In this study, we characterised the binding, endocytosis and axonal transport of an adenovirus (CAV-2) that preferentially infects neurons. Using biochemical, cell biology, genetic, ultrastructural and live-cell imaging approaches, we show that interaction with the neuronal membrane correlates with coxsackievirus and adenovirus receptor (CAR) surface expression, followed by endocytosis involving clathrin. In axons, long-range CAV-2 motility was bidirectional with a bias for retrograde transport in nonacidic Rab7-positive organelles. Unexpectedly, we found that CAR was associated with CAV-2 vesicles that also transported cargo as functionally distinct as tetanus toxin, neurotrophins, and their receptors. These results suggest that a single axonal transport carrier is capable of transporting functionally distinct cargoes that target different membrane compartments in the soma. We propose that CAV-2 transport is dictated by an innate trafficking of CAR, suggesting an unsuspected function for this adhesion protein during neuronal homeostasis. Adenoviruses commonly cause subclinical morbidity in the ocular, respiratory, and gastrointestinal tracts, and less frequently, adenovirus-induced disease can be fatal for newborns and immunocompromised hosts. In addition, adenoviruses can reach the central nervous system (CNS) and cause associated encephalitis and tumours. On the flip side, during the last two decades, adenovirus vectors have become powerful tools to treat and address diseases of the CNS. Despite the fact that axonal transport of adenoviruses was reported more than 15 years ago, nothing was known concerning how adenoviruses access the CNS. The characterization of their interactions with brain cells was therefore long overdue. In this study, we describe the axonal trafficking of an adenovirus that preferentially infects neurons and reaches the CNS through long-range axonal transport. We show that this adenovirus exploits an endogenous vesicular pathway used by the adhesion molecule CAR (coxsackievirus and adenovirus receptor). Our study characterizes this endogenous route of access, which is likely to be crucial to neuronal survival, neurodegenerative diseases, gene transfer vectors, and adenovirus-induced morbidity.
Collapse
Affiliation(s)
- Sara Salinas
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | - Lynsey G. Bilsland
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | - Daniel Henaff
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Universités de Montpellier I & II, Montpellier, France
| | - Anne E. Weston
- Electron Microscopy Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | - Anne Keriel
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Universités de Montpellier I & II, Montpellier, France
| | - Giampietro Schiavo
- Molecular NeuroPathobiology Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
- * E-mail: (GS); (EJK)
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Universités de Montpellier I & II, Montpellier, France
- * E-mail: (GS); (EJK)
| |
Collapse
|
71
|
The hepatitis E virus open reading frame 3 product interacts with microtubules and interferes with their dynamics. J Virol 2009; 83:6375-82. [PMID: 19369329 DOI: 10.1128/jvi.02571-08] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hepatitis E virus (HEV) is the causative agent of hepatitis E, a major form of viral hepatitis in developing countries. The open reading frame 3 (ORF3) of HEV encodes a phosphoprotein with a molecular mass of approximately 13 kDa (hereinafter called vp13). vp13 is essential for establishing HEV infections in animals, yet its exact functions are still obscure. Our current study found evidence showing interaction between vp13 and microtubules. Live-cell confocal fluorescence microscopy revealed both filamentous and punctate distribution patterns of vp13 in cells transfected with recombinant ORF3 reporter plasmids. The filamentous pattern of vp13 was altered by a microtubule-destabilizing drug. The vp13 expression led to elevation of acetylated alpha-tubulin, indicating increased microtubule stability. Its association with microtubules was further supported by its presence in microtubule-containing pellets in microtubule isolation assays. Exposure of these pellets to a high-salt buffer caused release of the vp13 to the supernatant, suggesting an electrostatic interaction. Inclusion of ATP and GTP in the lysis buffer during microtubule isolation also disrupted the interaction, indicating its sensitivity to the nucleotides. Further assays showed that motor proteins are needed for the vp13 association with the microtubules because disruption of dynein function abolished the vp13 filamentous pattern. Analysis of ORF3 deletion constructs found that both of the N-terminal hydrophobic domains of vp13 are needed for the interaction. Thus, our findings suggest that the vp13 interaction with microtubules might be needed for establishment of an HEV infection.
Collapse
|
72
|
Mudhakir D, Harashima H. Learning from the viral journey: how to enter cells and how to overcome intracellular barriers to reach the nucleus. AAPS JOURNAL 2009; 11:65-77. [PMID: 19194803 DOI: 10.1208/s12248-009-9080-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 12/24/2008] [Indexed: 02/06/2023]
Abstract
Viruses deliver their genome into host cells where they subsequently replicate and multiply. A variety of relevant strategies have evolved by which viruses gain intracellular access and utilize cellular machinery for the synthesis of their genome. Therefore, the viral journey provides insight into the cell's trafficking machinery and how it can be best exploited to improve nonviral gene delivery systems. This review summarizes viral internalization pathways and intracellular trafficking of viruses, with an emphasis on the endosomal escape processes of nonenveloped viruses. Intracellular events from viral entry through nuclear delivery of the viral complementary DNA are also discussed.
Collapse
Affiliation(s)
- Diky Mudhakir
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | | |
Collapse
|
73
|
Nasr P, Sullivan PG, Smith GM. Mitochondrial imaging in dorsal root ganglion neurons following the application of inducible adenoviral vector expressing two fluorescent proteins. J Neurosci Methods 2008; 172:185-94. [PMID: 18541307 PMCID: PMC2657596 DOI: 10.1016/j.jneumeth.2008.04.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 04/06/2008] [Accepted: 04/22/2008] [Indexed: 01/31/2023]
Abstract
Mitochondrial morphology and dynamics are known to vary considerably depending on the cell type and organism studied. The objective of this study was to assess the potential application of adenoviral-fluorescent protein constructs for long-term tracking of mitochondria in neurons. An adenoviral vector containing two fluorescent proteins, the enhanced green fluorescent protein (eGFP) targeted to the cytoplasm to highlight the neuronal processes, and the red fluorescent protein (RFP) directed to mitochondria under the control of an inducible promoter, facilitated an efficient and accurate method to study mitochondrial dynamics in long-term studies. Dorsal root ganglion neurons from rat embryos were cultured and infected. The infected neurons exhibited green fluorescence after 24h, while 16 h following induction with doxycycline, red fluorescence protein began to localize within mitochondria. The red fluorescent protein was transported into mitochondria at the cell body followed by distribution within processes. As the neurons aged, the expression of red fluorescent protein was confined to cytoplasmic vacuoles and not mitochondria. Further analysis suggested that the cytoplasmic vacuoles were likely of lysosomal origin. Taken together, the current study presents novel strategies to study the life history of cellular organelles such as mitochondria in long-term studies.
Collapse
Affiliation(s)
- Payman Nasr
- Department of Biological Sciences, Kent State University, Ashtabula, OH 44004, United States.
| | | | | |
Collapse
|
74
|
Singh R, Al-Jamal KT, Lacerda L, Kostarelos K. Nanoengineering artificial lipid envelopes around adenovirus by self-assembly. ACS NANO 2008; 2:1040-1050. [PMID: 19206502 DOI: 10.1021/nn8000565] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We have developed a novel, reproducible, and facile methodology for the construction of artificial lipid envelopes for adenoviruses (Ad) by self-assembly of lipid molecules around the viral capsid. No alteration of the viral genome or conjugation surface chemistry at the virus capsid was necessary, therefore difficulties in production and purification associated with generating most surface-modified viruses can be eliminated. Different lipid bilayer compositions produced artificially enveloped Ad with physicochemical and biological characteristics determined by the type of lipid used. Physicochemical characteristics such as vector size, degree of aggregation, stability, and surface charge of the artificially enveloped Ad were correlated to their biological (gene transfer) function. In monolayer cell cultures, binding to the coxsackie and adenovirus receptor (CAR) was blocked using a zwitterionic envelope, whereas enhanced binding to the cell membrane was achieved using a cationic envelope. Envelopment of Ad by both zwitterionic and cationic lipid bilayers led to almost complete ablation of gene expression in cell monolayers, due to blockage of virion endosomal escape. Alternatively, artificial Ad envelopes built from lipid bilayers at the fluid phase in physiological conditions led to enhanced penetration of the vectors inside a three-dimensional tumor spheroid cell culture model and delayed gene expression in the tumor spheroid compared to nonenveloped adenovirus. These results indicate that construction of artificial envelopes for nonenveloped viruses by lipid bilayer wrapping of the viral capsids constitutes a general strategy to rationally engineer viruses at the nanoscale with control over their biological properties.
Collapse
Affiliation(s)
- Ravi Singh
- Nanomedicine Laboratory, Centre for Drug Delivery Research, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | | | | | | |
Collapse
|
75
|
Neutralizing antibody blocks adenovirus infection by arresting microtubule-dependent cytoplasmic transport. J Virol 2008; 82:6492-500. [PMID: 18448546 DOI: 10.1128/jvi.00557-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Neutralizing antibodies are commonly elicited by viral infection. Most antibodies that have been characterized block early stages of virus entry that occur before membrane penetration, whereas inhibition of late stages in entry that occurs after membrane penetration has been poorly characterized. Here we provide evidence that the neutralizing antihexon monoclonal antibody 9C12 inhibits adenovirus infection by blocking microtubule-dependent translocation of the virus to the microtubule-organizing center following endosome penetration. These studies identify a previously undescribed mechanism by which neutralizing antibodies block virus infection, a situation that may be relevant for other nonenveloped viruses that use microtubule-dependent transport during cell entry.
Collapse
|
76
|
Diefenbach RJ, Miranda-Saksena M, Douglas MW, Cunningham AL. Transport and egress of herpes simplex virus in neurons. Rev Med Virol 2008; 18:35-51. [PMID: 17992661 DOI: 10.1002/rmv.560] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mechanisms of axonal transport of the alphaherpesviruses, HSV and pseudorabies virus (PrV), in neuronal axons are of fundamental interest, particularly in comparison with other viruses, and offer potential sites for antiviral intervention or development of gene therapy vectors. These herpesviruses are transported rapidly along microtubules (MTs) in the retrograde direction from the axon terminus to the dorsal root ganglion and then anterogradely in the opposite direction. Retrograde transport follows fusion and deenvelopment of the viral capsid at the axonal membrane followed by loss of most of the tegument proteins and then binding of the capsid via one or more viral proteins (VPs) to the retrograde molecular motor dynein. The HSV capsid protein pUL35 has been shown to bind to the dynein light chain Tctex1 but is likely to be accompanied by additional dynein binding of an inner tegument protein. The mechanism of anterograde transport is much more controversial with different processes being claimed for PrV and HSV: separate transport of HSV capsid/tegument and glycoproteins versus PrV transport as an enveloped virion. The controversy has not been resolved despite application, in several laboratories, of confocal microscopy (CFM), real-time fluorescence with viruses dual labelled on capsid and glycoprotein, electron microscopy in situ and immuno-electron microscopy. Different processes for each virus seem counterintuitive although they are the most divergent in the alphaherpesvirus subfamily. Current hypotheses suggest that unenveloped HSV capsids complete assembly in the axonal growth cones and varicosities, whereas with PrV unenveloped capsids are only found travelling in a retrograde direction.
Collapse
Affiliation(s)
- Russell J Diefenbach
- Centre for Virus Research, Westmead Millennium Institute, Westmead Hospital and the University of Sydney, Westmead, NSW 2145, Australia
| | | | | | | |
Collapse
|
77
|
Warren JC, Cassimeris L. The contributions of microtubule stability and dynamic instability to adenovirus nuclear localization efficiency. ACTA ACUST UNITED AC 2007; 64:675-89. [PMID: 17565754 DOI: 10.1002/cm.20215] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Adenoviruses (Ads) utilize host cell microtubules to traverse the intracellular space and reach the nucleus in a highly efficient manner. Previous studies have shown that Ad infection promotes the formation of stable, posttranslationally modified microtubules by a RhoA-dependent mechanism. Ad infection also shifts key parameters of microtubule dynamic instability by a Rac1-dependent mechanism, resulting in microtubules with lower catastrophe frequencies, persistent growth phases, and a bias toward net growth compared to microtubules in uninfected cells. Until now it was unclear whether changes in RhoGTPase activity or microtubule dynamics had a direct impact on the efficiency of Ad microtubule-dependent nuclear localization. Here we have performed synchronous Ad infections and utilized confocal microscopy to analyze the individual contributions of RhoA activation, Rac1 activation, microtubule stability, dynamic behavior, and posttranslational modifications on Ad nuclear localization efficiency (NLE). We found that drug-induced suppression of microtubule dynamics impaired Ad NLE by disrupting the radial organization of the microtubule array. When the microtubule array was maintained, the suppression or enhancement of microtubule turnover did not significantly affect Ad NLE. Furthermore, RhoA activation or the formation of acetylated microtubules did not enhance Ad NLE. In contrast, active Rac1 was required for efficient Ad nuclear localization. Because Rac1 mediates persistent growth of microtubules to the lamellar regions of cells, we propose that Ad-induced activation of Rac1 enhances the ability of microtubules to "search and capture" incoming virus particles.
Collapse
Affiliation(s)
- James C Warren
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | | |
Collapse
|
78
|
Abstract
To overcome barriers to diffusion, many viruses utilize the microtubule-associated molecular motor cytoplasmic dynein 1 to drive transport towards the nucleus of a target cell. Cytoplasmic dynein 1 generates movement towards the minus end of microtubules located at the microtubule organizing centre (MTOC), a structure that is typically in close proximity to the nucleus. Physiological cargoes for cytoplasmic dynein include membranous organelles, protein complexes and aggregates of misfolded protein. In this review, we discuss the study of microtubule-based translocation of viruses and raise questions about the mechanisms for association with and then dissociation from cytoplasmic dynein with a goal of understanding whether viruses are seen by the intracellular trafficking machinery as functional protein complexes or misfolded protein aggregates.
Collapse
Affiliation(s)
- Philip L Leopold
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY, USA.
| | | |
Collapse
|
79
|
Campos SK, Barry MA. Current advances and future challenges in Adenoviral vector biology and targeting. Curr Gene Ther 2007; 7:189-204. [PMID: 17584037 PMCID: PMC2244792 DOI: 10.2174/156652307780859062] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gene delivery vectors based on Adenoviral (Ad) vectors have enormous potential for the treatment of both hereditary and acquired disease. Detailed structural analysis of the Ad virion, combined with functional studies has broadened our knowledge of the structure/function relationships between Ad vectors and host cells/tissues and substantial achievement has been made towards a thorough understanding of the biology of Ad vectors. The widespread use of Ad vectors for clinical gene therapy is compromised by their inherent immunogenicity. The generation of safer and more effective Ad vectors, targeted to the site of disease, has therefore become a great ambition in the field of Ad vector development. This review provides a synopsis of the structure/function relationships between Ad vectors and host systems and summarizes the many innovative approaches towards achieving Ad vector targeting.
Collapse
Affiliation(s)
- Samuel K. Campos
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Michael A. Barry
- Department of Internal Medicine, Department of Immunology, Division of Infectious Diseases, Translational Immunovirology Program, Molecular Medicine Program, Mayo Clinic, Rochester, MN 55902, USA
- *Address correspondence to this author at the Department of Internal Medicine, Department of Immunology, Division of Infectious Diseases, Translational Immunovirology Program, Molecular Medicine Program, Mayo Clinic, Rochester, MN 55902, USA; E-mail:
| |
Collapse
|
80
|
Cabral-Romero C, Padilla-Noriega L. Association of rotavirus viroplasms with microtubules through NSP2 and NSP5. Mem Inst Oswaldo Cruz 2007; 101:603-11. [PMID: 17072471 DOI: 10.1590/s0074-02762006000600006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Accepted: 04/10/2006] [Indexed: 11/22/2022] Open
Abstract
Rotavirus replication and virus assembly take place in electrodense spherical structures known as viroplasms whose main components are the viral proteins NSP2 and NSP5. The viroplasms are produced since early times after infection and seem to grow by stepwise addition of viral proteins and by fusion, however, the mechanism of viropIasms formation is unknown. In this study we found that the viroplasms surface colocalized with microtubules, and seem to be caged by a microtubule network. Moreover inhibition of microtubule assembly with nocodazole interfered with viroplasms growth in rotavirus infected cells. We searched for a physical link between viroplasms and microtubules by co-immunoprecipitation assays, and we found that the proteins NSP2 and NSP5 were co-immunoprecipitated with anti-tubulin in rotavirus infected cells and also when they were transiently co-expressed or individually expressed. These results indicate that a functional microtubule network is needed for viroplasm growth presumably due to the association of viroplasms with microtubules via NSP2 and NSP5.
Collapse
Affiliation(s)
- Claudio Cabral-Romero
- Departamento de Biología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, DF 04510, México
| | | |
Collapse
|
81
|
Bergen JM, Pun SH. Evaluation of an LC8-binding peptide for the attachment of artificial cargo to dynein. Mol Pharm 2007; 4:119-28. [PMID: 17274669 PMCID: PMC2569131 DOI: 10.1021/mp060086o] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The limited cytoplasmic mobility of nonviral gene carriers is likely to contribute to their low transfection efficiency. This limitation could be overcome by mimicking the viral strategy of recruiting the dynein motor complex for efficient transport toward the host cell nucleus. A promising approach for attaching artificial cargo to dynein is through an adaptor peptide that binds the 8 kDa light chain (LC8) found in the cargo-binding region of the dynein complex. Several viral proteins that bind LC8 have in common an LC8-binding motif defined by (K/R)XTQT. Short peptides containing this motif have also been shown to bind recombinant LC8 in vitro. However, since the majority of intracellular LC8 exists outside of the dynein complex, it remains unclear whether peptides displaying this LC8-binding motif can access and bind to dynein-associated LC8. In this study, we employed biochemical analysis to investigate the feasibility of attaching artificial cargo to the dynein motor complex using a peptide displaying the well-characterized LC8-binding motif. We report that free intracellular LC8 bound specifically to an LC8-binding (TQT) peptide and not to a control peptide with a mutated LC8-binding motif. However, a similar binding interaction between the TQT peptide and intracellular dynein was not detected. To determine whether dynein binding of the TQT peptide was prevented by competition with free intracellular LC8 or due to the inability of the peptide to access its LC8 binding site in the dynein complex, the TQT peptide was evaluated for its ability to bind either purified LC8 or purified dynein. Our results demonstrate that, while the TQT peptide readily binds free LC8, it cannot bind to dynein-associated LC8. The results emphasize the need to identify functional dynein-binding peptides and highlight the importance of designing peptides that bind to the intact dynein motor complex.
Collapse
Affiliation(s)
| | - Suzie H. Pun
- Author to whom correspondence should be addressed. Mailing address: Foege Building, 1705 NE Pacific St., Room N530P, Box 355061, Seattle, WA 98195. Tel: (206) 685-3488. Fax: (206) 616-1984. E-mail:
| |
Collapse
|
82
|
|
83
|
Wodrich H, Cassany A, D'Angelo MA, Guan T, Nemerow G, Gerace L. Adenovirus core protein pVII is translocated into the nucleus by multiple import receptor pathways. J Virol 2006; 80:9608-18. [PMID: 16973564 PMCID: PMC1617226 DOI: 10.1128/jvi.00850-06] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Adenoviruses are nonenveloped viruses with an approximately 36-kb double-stranded DNA genome that replicate in the nucleus. Protein VII, an abundant structural component of the adenovirus core that is strongly associated with adenovirus DNA, is imported into the nucleus contemporaneously with the adenovirus genome shortly after virus infection and may promote DNA import. In this study, we evaluated whether protein VII uses specific receptor-mediated mechanisms for import into the nucleus. We found that it contains potent nuclear localization signal (NLS) activity by transfection of cultured cells with protein VII fusion constructs and by microinjection of cells with recombinant protein VII fusions. We identified three NLS-containing regions in protein VII by deletion mapping and determined important NLS residues by site-specific mutagenesis. We found that recombinant protein VII and its NLS-containing domains strongly and specifically bind to importin alpha, importin beta, importin 7, and transportin, which are among the most abundant cellular nuclear import receptors. Moreover, these receptors can mediate the nuclear import of protein VII fusions in vitro in permeabilized cells. Considered together, these data support the hypothesis that protein VII is a major NLS-containing adaptor for receptor-mediated import of adenovirus DNA and that multiple import pathways are utilized to promote efficient nuclear entry of the viral genome.
Collapse
Affiliation(s)
- Harald Wodrich
- Institut de Génétique Moléculaire de Montpellier, UMR 5535 CNRS, 1919 Route de Mende, 34293 Montpellier Cedex 05, France.
| | | | | | | | | | | |
Collapse
|
84
|
Burakov AV, Nadezhdina ES. Dynein and dynactin as organizers of the system of cell microtubules. Russ J Dev Biol 2006. [DOI: 10.1134/s1062360406050018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
85
|
Kelkar S, De BP, Gao G, Wilson JM, Crystal RG, Leopold PL. A common mechanism for cytoplasmic dynein-dependent microtubule binding shared among adeno-associated virus and adenovirus serotypes. J Virol 2006; 80:7781-5. [PMID: 16840360 PMCID: PMC1563724 DOI: 10.1128/jvi.00481-06] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During infection, adenovirus-associated virus (AAV) undergoes microtubule-dependent retrograde transport as part of a program of vectorial transport of viral genome to the nucleus. A microtubule binding assay was used to evaluate the hypothesis that cytoplasmic dynein mediates AAV interaction with microtubules. Binding of AAV serotype 2 (AAV2) was enhanced in a nucleotide-dependent manner by the presence of total cellular microtubule-associated proteins (MAPs) but not cytoplasmic dynein-depleted MAPs. Excess AAV2 capsid protein prevented microtubule binding by AAV serotypes 2, 5, and rh.10, as well as adenovirus serotype 5, indicating that similar binding sites are used by these viruses.
Collapse
Affiliation(s)
- Samir Kelkar
- Department of Genetic Medicine, Weill Medical College of Cornell University, 515 E. 71st Street, S-1000, New York, NY 10021, USA.
| | | | | | | | | | | |
Collapse
|
86
|
Warren JC, Rutkowski A, Cassimeris L. Infection with replication-deficient adenovirus induces changes in the dynamic instability of host cell microtubules. Mol Biol Cell 2006; 17:3557-68. [PMID: 16775012 PMCID: PMC1525226 DOI: 10.1091/mbc.e05-09-0850] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Adenovirus translocation to the nucleus occurs through a well characterized minus end-directed transport along microtubules. Here, we show that the adenovirus infection process has a significant impact on the stability and dynamic behavior of host cell microtubules. Adenovirus-infected cells had elevated levels of acetylated and detyrosinated microtubules compared with uninfected cells. The accumulation of modified microtubules within adenovirus-infected cells required active RhoA. Adenovirus-induced changes in microtubule dynamics were characterized at the centrosome and at the cell periphery in living cells. Adenovirus infection resulted in a transient enhancement of centrosomal microtubule nucleation frequency. At the periphery of adenovirus-infected cells, the dynamic instability of microtubules plus ends shifted toward net growth, compared with the nearly balanced growth and shortening observed in uninfected cells. In infected cells, microtubules spent more time in growth, less time in shortening, and underwent catastrophes less frequently compared with those in uninfected cells. Drug-induced inhibition of Rac1 prevented most of these virus-induced shifts in microtubule dynamic instability. These results demonstrate that adenovirus infection induces a significant stabilizing effect on host cell microtubule dynamics, which involve, but are not limited to, the activation of the RhoGTPases RhoA and Rac1.
Collapse
Affiliation(s)
- James C Warren
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015, USA.
| | | | | |
Collapse
|
87
|
McConnell MJ, Danthinne X, Imperiale MJ. Characterization of a permissive epitope insertion site in adenovirus hexon. J Virol 2006; 80:5361-70. [PMID: 16699016 PMCID: PMC1472126 DOI: 10.1128/jvi.00256-06] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 03/16/2006] [Indexed: 01/22/2023] Open
Abstract
A robust immune response is generated against components of the adenovirus capsid. In particular, a potent and long-lived humoral response is elicited against the hexon protein. This is due to the efficient presentation of adenovirus capsid proteins to CD4+ T cells by antigen-presenting cells, in addition to the highly repetitive structure of the adenovirus capsids, which can efficiently stimulate B-cell proliferation. In the present study, we take advantage of this immune response by inserting epitopes against which an antibody response is desired into the adenovirus hexon. We use a B-cell epitope from Bacillus anthracis protective antigen (PA) as a model antigen to characterize hypervariable region 5 (HVR5) of hexon as a site for peptide insertion. We demonstrate that HVR5 can accommodate a peptide of up to 36 amino acids without adversely affecting virus infectivity, growth, or stability. Viruses containing chimeric hexons elicited antibodies against PA in mice, with total immunoglobulin G (IgG) titers reaching approximately 1 x 10(3) after two injections. The antibody response contained both IgG1 and IgG2a subtypes, suggesting that Th1 and Th2 immunity had been stimulated. Coinjection of wild-type adenovirus and a synthetic peptide from PA produced no detectable antibodies, indicating that incorporation of the epitope into the capsid was crucial for immune stimulation. Together, these results indicate that the adenovirus capsid is an efficient vehicle for presenting B-cell epitopes to the immune system, making this a useful approach for the design of epitope-based vaccines.
Collapse
Affiliation(s)
- Michael J McConnell
- University of Michigan Medical School, 6304 Cancer Center, 1500 E. Medical Center Drive, Ann Arbor, MI 48109-0942, USA
| | | | | |
Collapse
|
88
|
Le LP, Le HN, Nelson AR, Matthews DA, Yamamoto M, Curiel DT. Core labeling of adenovirus with EGFP. Virology 2006; 351:291-302. [PMID: 16678874 PMCID: PMC1781517 DOI: 10.1016/j.virol.2006.03.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Revised: 12/15/2005] [Accepted: 03/21/2006] [Indexed: 11/27/2022]
Abstract
The study of adenovirus could greatly benefit from diverse methods of virus detection. Recently, it has been demonstrated that carboxy-terminal EGFP fusions of adenovirus core proteins Mu, V, and VII properly localize to the nucleus and display novel function in the cell. Based on these observations, we hypothesized that the core proteins may serve as targets for labeling the adenovirus core with fluorescent proteins. To this end, we constructed various chimeric expression vectors with fusion core genes (Mu-EGFP, V-EGFP, preVII-EGFP, and matVII-EGFP) while maintaining expression of the native proteins. Expression of the fusion core proteins was suboptimal using E1 expression vectors with both conventional CMV and modified (with adenovirus tripartite leader sequence) CMV5 promoters, resulting in non-labeled viral particles. However, robust expression equivalent to the native protein was observed when the fusion genes were placed in the deleted E3 region. The efficient Ad-wt-E3-V-EGFP and Ad-wt-E3-preVII-EGFP expression vectors were labeled allowing visualization of purified virus and tracking of the viral core during early infection. The vectors maintained their viral function, including viral DNA replication, viral DNA encapsidation, cytopathic effect, and thermostability. Core labeling offers a means to track the adenovirus core in vector targeting studies as well as basic adenovirus virology.
Collapse
Affiliation(s)
- Long P Le
- Division of Human Gene Therapy, Departments of Medicine, Pathology and Surgery, University of Alabama at Birmingham, 901 19th Street South, BMR2-502, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
89
|
Abstract
Microtubule-mediated transport of macromolecules and organelles (also known as "cargo") is essential for cells to function. Deficiencies in cytoplasmic transport are frequently associated with severe diseases and syndromes. Cytoplasmic transport also provides viruses with the means to reach their site of replication and is the route for newly assembled progeny to leave the infected cell. This parasitic relationship of viruses with the host cytoskeleton provides an excellent basis for cell biologists to unlock the secrets of cytoplasmic transport and unravel mechanisms of disease. Recent advances in live cell imaging and computational tracking of fluorescently labeled viruses are now revealing how complex the movements of single viruses are in infected cells. This review focuses on microtubule-based motility of viruses and highlights the mechanisms regulating cytoplasmic transport.
Collapse
Affiliation(s)
- Urs F Greber
- Zoologisches Institut der Universität Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | | |
Collapse
|
90
|
Abstract
Nonviral vectors continue to be attractive alternatives to viruses due to their low toxicity and immunogenicity, lack of pathogenicity, and ease of pharmacologic production. However, nonviral vectors also continue to suffer from relatively low levels of gene transfer compared to viruses, thus the drive to improve these vectors continues. Many studies on vector-cell interactions have reported that nonviral vectors bind and enter cells efficiently, but yield low gene expression, thus directing our attention to the intracellular trafficking of these vectors to understand where the obstacles occur. Here, we will review nonviral vector trafficking pathways, which will be considered here as the steps from cell binding to nuclear delivery. Studies on the intracellular trafficking of nonviral vectors has given us valuable insights into the barriers these vectors must overcome to mediate efficient gene transfer. Importantly, we will highlight the different approaches used by researchers to overcome certain trafficking barriers to gene transfer, many of which incorporate components from biological systems that have naturally evolved the capacity to overcome such obstacles. The tools used to study trafficking pathways will also be discussed.
Collapse
Affiliation(s)
- L K Medina-Kauwe
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | |
Collapse
|
91
|
Campos SK, Barry MA. Comparison of adenovirus fiber, protein IX, and hexon capsomeres as scaffolds for vector purification and cell targeting. Virology 2006; 349:453-62. [PMID: 16504233 DOI: 10.1016/j.virol.2006.01.032] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2005] [Revised: 12/22/2005] [Accepted: 01/18/2006] [Indexed: 11/17/2022]
Abstract
The direct genetic modification of adenoviral capsid proteins with new ligands is an attractive means to confer targeted tropism to adenoviral vectors. Although several capsid proteins have been reported to tolerate the genetic fusion of foreign peptides and proteins, direct comparison of cell targeting efficiencies through the different capsomeres has been lacking. Likewise, direct comparison of with one or multiple ligands has not been performed due to a lack of capsid-compatible ligands available for retargeting. Here we utilize a panel of metabolically biotinylated Ad vectors to directly compare targeted transduction through the fiber, protein IX, and hexon capsomeres using a variety of biotinylated ligands including antibodies, transferrin, EGF, and cholera toxin B. These results clearly demonstrate that cell targeting with a variety of high affinity receptor-binding ligands is only effective when transduction is redirected through the fiber protein. In contrast, protein IX and hexon-mediated targeting by the same set of ligands failed to mediate robust vector targeting, perhaps due to aberrant trafficking at the cell surface or inside targeted cells. These data suggest that vector targeting by genetic incorporation of high affinity ligands will likely be most efficient through modification of the adenovirus fiber rather than the protein IX and hexon capsomeres. In contrast, single-step monomeric avidin affinity purification of Ad vectors using the metabolic biotinylation system is most effective through capsomeres like protein IX and hexon.
Collapse
Affiliation(s)
- Samuel K Campos
- The Department of Biochemistry and Cell Biology, Rice University, Houston, TX 77005, USA
| | | |
Collapse
|
92
|
Citterio C, Jones HD, Pacheco-Rodriguez G, Islam A, Moss J, Vaughan M. Effect of protein kinase A on accumulation of brefeldin A-inhibited guanine nucleotide-exchange protein 1 (BIG1) in HepG2 cell nuclei. Proc Natl Acad Sci U S A 2006; 103:2683-8. [PMID: 16467138 PMCID: PMC1413798 DOI: 10.1073/pnas.0510571103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Brefeldin A-inhibited guanine nucleotide-exchange proteins, BIG1 and BIG2, are activators of ADP-ribosylation factor GTPases that are essential for regulating vesicular traffic among intracellular organelles. Biochemical analyses and immunofluorescence microscopy demonstrated BIG1 in nuclei as well as membranes and cytosol of serum-starved HepG2 cells. Within 20 min after addition of 8-Br-cAMP, BIG1 accumulated in nuclei, and this effect was blocked by protein kinase A (PKA) inhibitors H-89 and PKI, suggesting a dependence on PKA-catalyzed phosphorylation. BIG2 localization was not altered by cAMP, nor did BIG2 small interfering RNA influence nuclear accumulation of BIG1 induced by cAMP. Mutant BIG1 (S883A) in which Ala replaced Ser-883, a putative PKA phosphorylation site, did not move to the nucleus with cAMP addition, whereas replacement with Asp (S883D) resulted in nuclear accumulation of BIG1 without or with cAMP exposure, consistent with the mechanistic importance of a negative charge at that site. Mutation (712KPK714) of the nuclear localization signal inhibited BIG1 accumulation in nuclei, and PKA-catalyzed phosphorylation of S883, although necessary, was not sufficient for nuclear accumulation, as shown by the double mutation S883D/nuclear localization signal. A role for microtubules in cAMP-induced translocation of BIG1 is inferred from its inhibition by nocodazole. Thus, two more critical elements of BIG1 molecular structure were identified, as well as the potential function of microtubules in a novel PKA effect on BIG1 translocation.
Collapse
Affiliation(s)
- Carmen Citterio
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Heather D. Jones
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Gustavo Pacheco-Rodriguez
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Aminul Islam
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Joel Moss
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Martha Vaughan
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
93
|
Mastrobattista E, van der Aa MAEM, Hennink WE, Crommelin DJA. Artificial viruses: a nanotechnological approach to gene delivery. Nat Rev Drug Discov 2006; 5:115-21. [PMID: 16521330 DOI: 10.1038/nrd1960] [Citation(s) in RCA: 256] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanotechnology is a rapidly expanding multidisciplinary field in which highly sophisticated nanoscale devices are constructed from atoms, molecules or (macro)molecular assemblies. In the field of gene medicine, systems for delivering nucleic acids are being developed that incorporate virus-like functions in a single nanoparticle. Although their development is still in its infancy, it is expected that such artificial viruses will have a great impact on the advancements of gene therapeutics.
Collapse
Affiliation(s)
- Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands.
| | | | | | | |
Collapse
|
94
|
Abstract
The intracellular steps involved in viral infection, namely cytoplasmic trafficking and nuclear import, are critical events in the viral life cycle that have lagged behind other areas of viral research. This review examines recent advances in our understanding of these steps for viruses commonly employed as viral gene delivery vectors. Steps governing the cytoplasmic trafficking and nuclear import of Herpes Simplex virus, Human Immunodeficiency virus and Adenovirus are reviewed in this article.
Collapse
Affiliation(s)
- E M Campbell
- Department of Microbiology and Immunology, University of Illinois at Chicago, 835 S. Wolcott Rm. E-703, Chicago, IL 60612, USA
| | | |
Collapse
|
95
|
Abstract
Retroviruses are efficient vehicles for delivering transgenes in vivo. Their ability to integrate into the host genome, providing a permanent imprint of their genes in the host, is a key asset for gene therapy. Furthermore, the lentivirus subset of retroviruses can infect nondividing as well as dividing cells. This expands the cell types capable of gene therapy, driving the development of lentiviral vectors. However, the precise mechanisms used by different retroviruses to efficiently deliver their genes into cell nuclei remains largely unclear. Understanding these molecular mechanisms may reveal features to improve the efficacy of current retroviral vectors. Moreover, this knowledge may expose elements pliable to other gene therapy vehicles to improve their in vivo performance and circumvent the biosafety concerns of using retroviral vectors. Therefore, the mechanisms underlying the early trafficking of retroviral vectors in host cells are reviewed here, as understood from studying the native retroviruses. Events after virus entry up to nuclear delivery of the viral cDNA are discussed. Cellular obstacles faced by these retroviral vectors and how they advance beyond these barriers is emphasized.
Collapse
Affiliation(s)
- J L Anderson
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3093, USA
| | | |
Collapse
|
96
|
De Laporte L, Cruz Rea J, Shea LD. Design of modular non-viral gene therapy vectors. Biomaterials 2005; 27:947-54. [PMID: 16243391 PMCID: PMC2648393 DOI: 10.1016/j.biomaterials.2005.09.036] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Accepted: 09/26/2005] [Indexed: 01/08/2023]
Abstract
Gene delivery has numerous potential applications both clinically and for basic science research. Non-viral vectors represent the long-term future of gene therapy and biomaterials are a critical component for the development of efficient delivery systems. Biomaterial development combined with fundamental studies of virus function and cellular processes will enable the molecular level design of modular vectors. Vectors are being developed based on cationic polymers or lipids that contain functional groups to mediate appropriate interactions with the extracellular environment or to interface with specific cellular processes. This review describes recent progress on the development of biomaterials for non-viral vectors and highlights opportunities for future development. Ultimately, efficient vectors will expand the traditional applications of gene therapy within the clinic and may enable numerous other opportunities within diagnostics, biotechnology, and basic science research.
Collapse
Affiliation(s)
| | | | - Lonnie D. Shea
- Corresponding author. Tel.: 847 491 7043; fax: 847 491 3728. E-mail address: (L.D. Shea)
| |
Collapse
|
97
|
Döhner K, Nagel CH, Sodeik B. Viral stop-and-go along microtubules: taking a ride with dynein and kinesins. Trends Microbiol 2005; 13:320-7. [PMID: 15950476 DOI: 10.1016/j.tim.2005.05.010] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Revised: 04/20/2005] [Accepted: 05/20/2005] [Indexed: 11/21/2022]
Abstract
Incoming viral particles move from the cell surface to sites of viral transcription and replication. By contrast, during assembly and egress, subviral nucleoprotein complexes and virions travel back to the plasma membrane. Because diffusion of large molecules is severely restricted in the cytoplasm, viruses use ATP-hydrolyzing molecular motors of the host for propelling along the microtubules, which are the intracellular highways. Recent studies have revealed that, besides travelling inside endocytic or exocytic vesicles, viral proteins interact directly with dynein or kinesin motors. Understanding the molecular mechanisms of cytoplasmic viral transport will aid in the construction of viral vectors for human gene therapy and the search for new antiviral targets.
Collapse
Affiliation(s)
- Katinka Döhner
- Institute of Virology, Hannover Medical School, D-30623 Hannover, Germany
| | | | | |
Collapse
|
98
|
Nieznanski K, Nieznanska H, Skowronek KJ, Osiecka KM, Stepkowski D. Direct interaction between prion protein and tubulin. Biochem Biophys Res Commun 2005; 334:403-11. [PMID: 16004966 DOI: 10.1016/j.bbrc.2005.06.092] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Accepted: 06/20/2005] [Indexed: 02/04/2023]
Abstract
Recently published data show that the prion protein in its cellular form (PrP(C)) is a component of multimolecular complexes. In this report, zero-length cross-linking with 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) allowed us to identify tubulin as one of the molecules interacting with PrP(C) in complexes observed in porcine brain extracts. We found that porcine brain tubulin added to these extracts can be cross-linked with PrP(C). Moreover, we observed that the 34 kDa species identified previously as full-length diglycosylated prion protein co-purifies with tubulin. Cross-linking of PrP(C) species separated by Cu(2+)-loaded immobilized metal affinity chromatography confirmed that only the full-length protein but not the N-terminally truncated form (C1) binds to tubulin. By means of EDC cross-linking and cosedimentation experiments, we also demonstrated a direct interaction of recombinant human PrP (rPrP) with tubulin. The stoichiometry of cosedimentation implies that rPrP molecules are able to bind both the alpha- and beta-isoforms of tubulin composing microtubule. Furthermore, prion protein exhibits higher affinity for microtubules than for unpolymerized tubulin.
Collapse
Affiliation(s)
- Krzysztof Nieznanski
- Department of Muscle Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
99
|
Salman H, Abu-Arish A, Oliel S, Loyter A, Klafter J, Granek R, Elbaum M. Nuclear localization signal peptides induce molecular delivery along microtubules. Biophys J 2005; 89:2134-45. [PMID: 16040740 PMCID: PMC1366715 DOI: 10.1529/biophysj.105.060160] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many essential processes in eukaryotic cells depend on regulated molecular exchange between its two major compartments, the cytoplasm and the nucleus. In general, nuclear import of macromolecular complexes is dependent on specific peptide signals and their recognition by receptors that mediate translocation through the nuclear pores. Here we address the question of how protein products bearing such nuclear localization signals arrive at the nuclear membrane before import, i.e., by simple diffusion or perhaps with assistance of cytoskeletal elements or cytoskeleton-associated motor proteins. Using direct single-particle tracking and detailed statistical analysis, we show that the presence of nuclear localization signals invokes active transport along microtubules in a cell-free Xenopus egg extract. Chemical and antibody inhibition of minus-end directed cytoplasmic dynein blocks this active movement. In the intact cell, where microtubules project radially from the centrosome, such an interaction would effectively deliver nuclear-targeted cargo to the nuclear envelope in preparation for import.
Collapse
Affiliation(s)
- Hanna Salman
- Department of Materials and Interfaces, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Here we develop an integrative computational framework to model biophysical processes involved in viral gene delivery. The model combines reaction-diffusion-advection equations that describe intracellular trafficking with kinetic equations that describe transcription and translation of the exogenous DNA. It relates molecular-level trafficking events to whole-cell distribution of viruses. The approach makes use of the current understanding of cellular processes and data from single-particle single-cell imaging experiments. The model reveals two important parameters that characterize viral transport at the population level, namely, the effective velocity, V(eff), and the effective diffusion coefficient, D(eff). V(eff) measures virus's net movement rate and D(eff) represents the total dispersion rate. We employ the model to study the influence of microtubule-mediated movements on nuclear targeting and gene expression of adenoviruses of type 2 and type 5 in HeLa and A549 cells. Effects of microtubule organization and the presence of microtubule-destabilizing drugs on viral transport were analyzed and quantified. Model predictions agree well with experimental data available in literature. The paper serves as a guide for future theoretical and experimental efforts to understand viral gene delivery.
Collapse
Affiliation(s)
- Anh-Tuan Dinh
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, USA
| | | | | |
Collapse
|