1
|
Simon F, Thoma-Kress AK. Intercellular Transport of Viral Proteins. Results Probl Cell Differ 2024; 73:435-474. [PMID: 39242389 DOI: 10.1007/978-3-031-62036-2_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Viruses are vehicles to exchange genetic information and proteins between cells and organisms by infecting their target cells either cell-free, or depending on cell-cell contacts. Several viruses like certain retroviruses or herpesviruses transmit by both mechanisms. However, viruses have also evolved the properties to exchange proteins between cells independent of viral particle formation. This exchange of viral proteins can be directed to target cells prior to infection to interfere with restriction factors and intrinsic immunity, thus, making the target cell prone to infection. However, also bystander cells, e.g. immune cell populations, can be targeted by viral proteins to dampen antiviral responses. Mechanistically, viruses exploit several routes of cell-cell communication to exchange viral proteins like the formation of extracellular vesicles or the formation of long-distance connections like tunneling nanotubes. Although it is known that viral nucleic acids can be transferred between cells as well, this chapter concentrates on viral proteins of human pathogenic viruses covering all Baltimore classes and summarizes our current knowledge on intercellular transport of viral proteins between cells.
Collapse
Affiliation(s)
- Florian Simon
- Institute of Clinical and Molecular Virology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andrea K Thoma-Kress
- Institute of Clinical and Molecular Virology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
2
|
Hejtmánková A, Váňová J, Španielová H. Cell-penetrating peptides in the intracellular delivery of viral nanoparticles. VITAMINS AND HORMONES 2021; 117:47-76. [PMID: 34420585 DOI: 10.1016/bs.vh.2021.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-penetrating peptides (CPPs) are a promising tool for the intracellular delivery of cargo. Due to their ability to cross membranes while also cotransporting various cargoes, they offer great potential for biomedical applications. Several CPPs have been derived from viral proteins with natural roles in the viral replication cycle that require them to breach or fuse to cellular membranes. Additionally, the ability of viruses to cross membranes makes viruses and virus-based particles a convenient model for research on nanoparticle delivery and nanoparticle-mediated gene therapy. In this chapter, we aim to characterize CPPs derived from both structural and nonstructural viral proteins. Their function as enhancers of viral infection and transduction by viral nanoparticles as well as the main features of viral CPPs employed in intracellular cargo delivery are summarized to emphasize their potential use in nanomedicine.
Collapse
Affiliation(s)
- Alžběta Hejtmánková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jana Váňová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Hana Španielová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry of the CAS, Prague, Czech Republic.
| |
Collapse
|
3
|
Yu X, Li T, Xia Y, Lei J, Wang Y, Zhang L. Herpes simplex virus type 1 VP22-mediated intercellular delivery of PTEN increases the antitumor activity of PTEN in esophageal squamous cell carcinoma cells in vitro and in vivo. Oncol Rep 2016; 35:3034-40. [PMID: 27004535 DOI: 10.3892/or.2016.4694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/12/2016] [Indexed: 11/06/2022] Open
Abstract
In the past decade, studies have revealed that the phosphatase and tensin homolog (PTEN) protein, a tumor suppressor, comprises a potential biological marker and therapeutic target for esophageal squamous cell carcinoma (ESCC). As such, the delivery of the PTEN gene represents a powerful strategy for ESCC therapy. The tegument protein VP22 of herpes simplex virus type 1 (HSV-1) has been reported to act as a transporter of heterologous proteins across the host cell membrane, thereby enhancing the biological functions of these proteins. In the present study, the intercellular delivery and antitumor activity of the fusion protein PTEN-VP22 were examined in the esophageal squamous cell carcinoma cell line Eca109 both in vitro and in vivo. VP22-mediated PTEN intercellular delivery was confirmed in the Eca109 cells by western blot analysis and by quantitation of immunofluorescence. VP22 alone did not exert antiproliferative effects or induce cell cycle arrest, induction of apoptosis, blockage of the Akt and focal adhesion kinase (FAK) pathways, tumor growth inhibition, or antiangiogenic effects in Eca109 cells. However, compared with PTEN alone, PTEN-VP22 exerted significantly higher antiproliferative effects and induced cell cycle arrest at G1 stage, apoptosis and antiangiogenic effects in Eca109 cells. Together, our findings demonstrate that VP22 alone does not exert antitumor activity directly; however, this protein mediates the intercellular delivery of PTEN and thereby increases its intracellular concentration to achieve a therapeutic steady state, leading to an overall increase in the antitumor activity of PTEN. This study provides further experimental data to confirm the potential of VP22-based intercellular delivery strategies for enhancing the efficacy of gene therapy for cancer treatment.
Collapse
Affiliation(s)
- Xian Yu
- Department of Pharmacy, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Tingting Li
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| | - Yifan Xia
- Department of Orthopaedics, Chongqing General Hospital, Chongqing 400014, P.R. China
| | - Jun Lei
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| | - Yan Wang
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| | - Lijuan Zhang
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| |
Collapse
|
4
|
Elmer JJ, Christensen MD, Rege K. Applying horizontal gene transfer phenomena to enhance non-viral gene therapy. J Control Release 2013; 172:246-257. [PMID: 23994344 PMCID: PMC4258102 DOI: 10.1016/j.jconrel.2013.08.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 08/17/2013] [Accepted: 08/20/2013] [Indexed: 12/25/2022]
Abstract
Horizontal gene transfer (HGT) is widespread amongst prokaryotes, but eukaryotes tend to be far less promiscuous with their genetic information. However, several examples of HGT from pathogens into eukaryotic cells have been discovered and mimicked to improve non-viral gene delivery techniques. For example, several viral proteins and DNA sequences have been used to significantly increase cytoplasmic and nuclear gene delivery. Plant genetic engineering is routinely performed with the pathogenic bacterium Agrobacterium tumefaciens and similar pathogens (e.g. Bartonella henselae) may also be able to transform human cells. Intracellular parasites like Trypanosoma cruzi may also provide new insights into overcoming cellular barriers to gene delivery. Finally, intercellular nucleic acid transfer between host cells will also be briefly discussed. This article will review the unique characteristics of several different viruses and microbes and discuss how their traits have been successfully applied to improve non-viral gene delivery techniques. Consequently, pathogenic traits that originally caused diseases may eventually be used to treat many genetic diseases.
Collapse
Affiliation(s)
- Jacob J Elmer
- Department of Chemical Engineering, Villanova University, Villanova 19085, USA.
| | | | - Kaushal Rege
- Chemical Engineering, Arizona State University, Tempe 85287-6106, USA.
| |
Collapse
|
5
|
Jin GS, Zhu GD, Zhao ZG, Liu FS. VP22 enhances the expression of glucocerebrosidase in human Gaucher II fibroblast cells mediated by lentiviral vectors. Clin Exp Med 2011; 12:135-43. [DOI: 10.1007/s10238-011-0152-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/29/2011] [Indexed: 01/17/2023]
|
6
|
Moeini H, Omar AR, Rahim RA, Yusoff K. Improving the potency of DNA vaccine against chicken anemia virus (CAV) by fusing VP1 protein of CAV to Marek's Disease Virus (MDV) type-1 VP22 protein. Virol J 2011; 8:119. [PMID: 21401953 PMCID: PMC3315791 DOI: 10.1186/1743-422x-8-119] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 03/14/2011] [Indexed: 11/17/2022] Open
Abstract
Background Studies have shown that the VP22 gene of Marek's Disease Virus type-1 (MDV-1) has the property of movement between cells from the original cell of expression into the neighboring cells. The ability to facilitate the spreading of the linked proteins was used to improve the potency of the constructed DNA vaccines against chicken anemia virus (CAV). Methods The VP1 and VP2 genes of CAV isolate SMSC-1 were amplified and inserted into eukaryotic co-expression vector, pBudCE4.1 to construct pBudVP2-VP1. We also constructed pBudVP2-VP1/VP22 encoding CAV VP2 and the VP22 of MDV-1 linked to the CAV VP1. In vitro expression of the genes was confirmed by using RT-PCR, Western blot and indirect immunofluorescence. The vaccines were then tested in 2-week-old SPF chickens which were inoculated with the DNA plasmid constructs by the intramuscular route. After in vivo expression studies, immune responses of the immunized chickens were evaluated pre- and post-immunization. Results Chickens vaccinated with pBudVP2-VP1/VP22 exhibited a significant increase in antibody titers to CAV and also proliferation induction of splenocytes in comparison to the chickens vaccinated with pBudVP2-VP1. Furthermore, the pBudVP2-VP1/VP22-vaccinated group showed higher level of the Th1 cytokines IL-2 and IFN-γ. Conclusions This study showed that MDV-1 VP22 gene is capable of enhancing the potency of DNA vaccine against CAV when fused with the CAV VP1 gene.
Collapse
Affiliation(s)
- Hassan Moeini
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | | | | | | |
Collapse
|
7
|
Wu Y, Ren C, Gao Y, Hou B, Chen T, Zhang C. A novel method for promoting heterologous protein expression in Escherichia coli by fusion with the HIV-1 TAT core domain. Amino Acids 2010; 39:811-20. [PMID: 20213440 DOI: 10.1007/s00726-010-0534-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 02/12/2010] [Indexed: 11/29/2022]
Abstract
The human immunodeficiency virus type 1 (HIV-1) transactivator of transcription (TAT) protein, a member of the protein transduction domain (PTD) superfamily, can deliver heterologous proteins across most biomembranes without losing bioactivity. However, there is no report on whether the TAT core domain containing the sequence 'YGRKKRRQRRR' has other functions. As the TAT core domain is most basic (pI=12.8) and has biomembrane crossing ability, we hypothesized it might probably influence the protein expression level due to subcellular redistribution of target proteins in the cells. To address this issue, we constructed the prokaryotic expression vector pET28b-TAT-EGFP (using the vector pET28b-EGFP for control) containing the core domain coding region, and transformed the vector into E. coli BL21 (DE3) cells for expression of the enhanced green fluorescent protein (EGFP) with the inducer isopropyl-beta-D-thiogalactopyranoside (IPTG). Equal amount of the total proteins were fractionated using 15% SDS-PAGE and identified by western blot, and the plasmid copy number was assayed by Southern blot. In order to further study the subcellular localization of heterologous proteins in E. coli cells, the cytoplasmic and periplasmic components were extracted by chloroform and osmotic shock techniques. Interestingly, our data showed that the TAT core domain was not only able to promote the heterologous protein expression in E. coli, but also improve the yields and the solubility of heterologous proteins, while the plasmid copy number of TAT-containing clones and TAT-free clones was not affected by the TAT core domain. In addition, the TAT-tagged protein was mainly localized in the cytoplasm and also accumulated in the periplasmic space along with the time for protein expression, while in contrast, the TAT-free protein was mainly expressed in the periplasm and only a few in cytoplasm. A further examination on the distribution of the expressed proteins in cytoplasm and periplasm suggested that the TAT core domain might promote protein expression in the cytoplasm initially and then partially deliver them across the cytomembrane to the periplasmic space in a concentration-dependent manner. Taken together, our current data have provided a novel method for improving heterologous protein expression in prokaryotic cells by fusion with the TAT core domain, which will promote expression efficiency of bioactive proteins for protein engineering.
Collapse
Affiliation(s)
- Yonghong Wu
- State Key Laboratory of Proteomics, Beijing Institute of Radiation Medicine, Taiping Road 27, 100850, Beijing, China
| | | | | | | | | | | |
Collapse
|
8
|
Abstract
As an attractive alternative to conventional vaccines, DNA vaccines play a critical role in inducing protection against several infectious diseases. In this review, we discuss the advantages that DNA vaccines offer in comparison to conventional protein-based vaccines. We discuss strategies to improve the potency and efficacy of DNA vaccines. Specifically, we focus on the potential use of DNA-based vaccines to elicit broad-spectrum humoral and cellular immunity against influenza virus. Finally, we discuss the advances made in the use of DNA vaccines to prevent avian H5N1 influenza.
Collapse
|
9
|
Kim DW, Kim SY, Lee SH, Lee YP, Lee MJ, Jeong MS, Jang SH, Park J, Lee KS, Kang TC, Won MH, Cho SW, Kwon OS, Eum WS, Choi SY. Protein transduction of an antioxidant enzyme: subcellular localization of superoxide dismutase fusion protein in cells. BMB Rep 2008; 41:170-5. [PMID: 18315955 DOI: 10.5483/bmbrep.2008.41.2.170] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In protein therapy, it is important for exogenous protein to be delivered into the target subcellular localization. To transduce a therapeutic protein into its specific subcellular localization, we synthesized nuclear localization signal (NLS) and membrane translocation sequence signal (MTS) peptides and produced a genetic in-frame SOD fusion protein. The purified SOD fusion proteins were efficiently transduced into mammalian cells with enzymatic activities. Immunofluorescence and Western blot analysis revealed that the SOD fusion proteins successfully transduced into the nucleus and the cytosol in the cells. The viability of cells treated with paraquat was markedly increased by the transduced fusion proteins. Thus, our results suggest that these peptides should be useful for targeting the specific localization of therapeutic proteins in various human diseases.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Biomedical Science and the Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Evaluation of the VP22 gene adjuvant for enhancement of DNA vaccine against somatostatin in mice. Animal 2008; 2:1569-74. [DOI: 10.1017/s175173110800284x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
11
|
Chauhan A, Tikoo A, Kapur AK, Singh M. The taming of the cell penetrating domain of the HIV Tat: myths and realities. J Control Release 2006; 117:148-62. [PMID: 17196289 PMCID: PMC1859861 DOI: 10.1016/j.jconrel.2006.10.031] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Accepted: 10/20/2006] [Indexed: 01/08/2023]
Abstract
Protein transduction with cell penetrating peptides over the past several years has been shown to be an effective way of delivering proteins in vitro and now several reports have also shown valuable in vivo applications in correcting disease states. An impressive bioinspired phenomenon of crossing biological barriers came from HIV transactivator Tat protein. Specifically, the protein transduction domain of HIV Tat has been shown to be a potent pleiotropic peptide in protein delivery. Various approaches such as molecular modeling, arginine guanidinium head group structural strategy, multimerization of PTD sequence and phage display system have been applied for taming of the PTD. This has resulted in identification of PTD variants which are efficient in cell membrane penetration and cytoplasmic delivery. In spite of these state of the art technologies, the dilemma of low protein transduction efficiency and target specific delivery of PTD fusion proteins remains unsolved. Moreover, some misconceptions about PTD of Tat in the literature require considerations. We have assembled critical information on secretory, plasma membrane penetration and transcellular properties of Tat and PTD using molecular analysis and available experimental evidences.
Collapse
Affiliation(s)
- Ashok Chauhan
- Department of Neurology, Richard Johnson Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, 509 Pathology, Baltimore, MD 21287, USA.
| | | | | | | |
Collapse
|
12
|
Saha S, Yoshida S, Ohba K, Matsui K, Matsuda T, Takeshita F, Umeda K, Tamura Y, Okuda K, Klinman D, Xin KQ, Okuda K. A fused gene of nucleoprotein (NP) and herpes simplex virus genes (VP22) induces highly protective immunity against different subtypes of influenza virus. Virology 2006; 354:48-57. [PMID: 16945400 DOI: 10.1016/j.virol.2006.04.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 03/01/2006] [Accepted: 04/12/2006] [Indexed: 10/24/2022]
Abstract
We evaluated the immunogenicity and protective activity of plasmid DNA vaccines encoding the influenza virus NP gene (pNP) alone or in combination with the herpes simplex virus type 1 protein 22 gene (pVP22). Optimal immune responses were observed in BALB/c mice immunized with the combination of pVP22 plus pNP, as assessed by enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunospot (ELISPOT) and intracellular cytokine staining (ICCS). These mice also showed maximal resistance following challenge with the A/PR/8/34 (H1N1) and A/Udron/72 (H3N2) strains of influenza virus. The susceptibility of immunized mice to virus infection was significantly increased following depletion of either CD4+ or CD8+ T cells. These results indicate that a plasmid DNA vaccine encoding pVP22 plus NP induces a high level of cross-protective immunity against influenza virus subtypes.
Collapse
Affiliation(s)
- Sukumar Saha
- Department of Molecular Biodefense Research, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hudecz F, Bánóczi Z, Csík G. Medium-sized peptides as built in carriers for biologically active compounds. Med Res Rev 2006; 25:679-736. [PMID: 15952174 DOI: 10.1002/med.20034] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A growing number of oligopeptides of natural and/or synthetic origin have been described and considered as targeting structures for delivery bioactive compounds into various cell types. This review will outline the discovery of peptide sequences and the corresponding mid-sized oligopeptides with membrane translocating properties and also summarize de novo designed structures possessing similar features. Conjugates and chimera constructs derived from these sequences with covalently attached bioactive peptide, epitope, oligonucleotide, PNA, drug, reporter molecule will be reviewed. A brief note will refer to the present understanding on the uptake mechanism at the end of each section.
Collapse
Affiliation(s)
- Ferenc Hudecz
- Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös Loránd University, Budapest 112, POB 32, Hungary H-1518. hudecz@szerves,chem.elte.hu
| | | | | |
Collapse
|
14
|
Zavaglia D, Lin EH, Guidetti M, Pluquet O, Hainaut P, Favrot MC, Coll JL. Poor intercellular transport and absence of enhanced antiproliferative activity after non-viral gene transfer of VP22-P53 or P53-VP22 fusions into p53 null cell lines in vitro or in vivo. J Gene Med 2005; 7:936-44. [PMID: 15754307 DOI: 10.1002/jgm.741] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The herpes simplex virus type 1 (HSV-1) VP22 protein has the property to mediate intercellular trafficking of heterologous proteins fused to its C- or N-terminus. We have previously shown improved delivery and enhanced therapeutic effect in vitro and in vivo with a P27-VP22 fusion protein. In this report, we were interested in studying the spread and biological activity of VP22 fused to the P53 tumor suppressor. METHODS Expression of the VP22-P53 and P53-VP22 fusion proteins was shown by Western blot and intercellular spreading was monitored by immunofluorescence on transiently transfected cells. In vitro antiproliferative activity of wild-type (wt) P53 and P53-VP22 was assessed by proliferation assays and transactivating ability was studied by a reporter gene test and a gel-shift assay. Antitumor activity was also tested in vivo by intratumoral injections of naked DNA in a model of subcutaneous tumors implanted in nude mice. RESULTS Our results show that the C-terminal fusion or the N-terminal P53-VP22 fusion proteins are not able to spread as efficiently as VP22. Moreover, we demonstrate that VP22-P53 does not possess any transactivating ability. P53-VP22 has an antiproliferative activity, but this activity is not superior to the one of P53 alone, in vitro or in vivo. CONCLUSIONS Our study indicates that a gene transfer strategy using VP22 cannot be considered as a universal system to improve the delivery of any protein.
Collapse
Affiliation(s)
- David Zavaglia
- Groupe de Recherche sur le Cancer du Poumon, INSERM U578, Institut Albert Bonniot, 38706 La Tronche, France
| | | | | | | | | | | | | |
Collapse
|
15
|
Hakkarainen T, Wahlfors T, Meriläinen O, Loimas S, Hemminki A, Wahlfors J. VP22 does not significantly enhance enzyme prodrug cancer gene therapy as a part of a VP22-HSVTk-GFP triple fusion construct. J Gene Med 2005; 7:898-907. [PMID: 15759279 DOI: 10.1002/jgm.737] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND VP22 is a herpes simplex virus type 1 (HSV-1) tegument protein that has been suggested to spread from cell to cell, alone or as a part of fusion proteins. Creating controversy, some reports indicate that VP22 cannot facilitate significant intercellular spreading. To study the capacity of VP22 to cause spreading and enhance thymidine kinase/ganciclovir cancer gene therapy, we constructed a novel triple fusion protein containing VP22, HSV thymidine kinase and green fluorescent protein (VP22-Tk-GFP). This fusion protein has three functional domains in the same polypeptide, thus making it possible to reliably compare the causality between transduction rate and cell killing efficiency in vitro and in vivo. METHODS VP22-Tk-GFP was cloned into lenti- and adenoviral vectors and used for expression studies, analyses for VP22-mediated protein spreading, and to study the effect of VP22 to thymidine kinase/ganciclovir-mediated cytotoxicity. The function of VP22-Tk-GFP was also investigated in vivo. RESULTS The triple fusion protein was expressed correctly in vitro, but intercellular trafficking was not observed in any of the studied cell lines. However, under certain conditions, VP22-Tk-GFP sensitized cells more efficiently to ganciclovir than Tk-GFP. In vivo there was a trend for increased inhibition of tumor growth with VP22-Tk-GFP when ganciclovir was present, but the difference with Tk-GFP was not statistically significant. CONCLUSIONS Based on our results, VP22 fusion proteins do not seem to traffic intercellularly at detectable levels in most tumor cell types. Even though VP22 enhanced cytotoxicity in one cell line in vitro, the effect in vivo was modest. Therefore, our results do not support the utility of VP22 as an enhancer of enzyme prodrug cancer gene therapy.
Collapse
Affiliation(s)
- Tanja Hakkarainen
- A. I. Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Kuopio, FIN-70211 Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
16
|
Oehmig A, Fraefel C, Breakefield XO. Update on herpesvirus amplicon vectors. Mol Ther 2005; 10:630-43. [PMID: 15451447 DOI: 10.1016/j.ymthe.2004.06.641] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 06/17/2004] [Indexed: 12/29/2022] Open
Affiliation(s)
- Angelika Oehmig
- Department of Neurology, Massachusetts General Hospital, and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
17
|
Perkins SD, Flick-Smith HC, Garmory HS, Essex-Lopresti AE, Stevenson FK, Phillpotts RJ. Evaluation of the VP22 protein for enhancement of a DNA vaccine against anthrax. GENETIC VACCINES AND THERAPY 2005; 3:3. [PMID: 15842732 PMCID: PMC1087864 DOI: 10.1186/1479-0556-3-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Accepted: 04/20/2005] [Indexed: 11/23/2022]
Abstract
Background Previously, antigens expressed from DNA vaccines have been fused to the VP22 protein from Herpes Simplex Virus type I in order to improve efficacy. However, the immune enhancing mechanism of VP22 is poorly understood and initial suggestions that VP22 can mediate intercellular spread have been questioned. Despite this, fusion of VP22 to antigens expressed from DNA vaccines has improved immune responses, particularly to non-secreted antigens. Methods In this study, we fused the gene for the VP22 protein to the gene for Protective Antigen (PA) from Bacillus anthracis, the causative agent of anthrax. Protective immunity against infection with B. anthracis is almost entirely based on a response to PA and we have generated two constructs, where VP22 is fused to either the N- or the C-terminus of the 63 kDa protease-cleaved fragment of PA (PA63). Results Following gene gun immunisation of A/J mice with these constructs, we observed no improvement in the anti-PA antibody response generated. Following an intraperitoneal challenge with 70 50% lethal doses of B. anthracis strain STI spores, no difference in protection was evident in groups immunised with the DNA vaccine expressing PA63 and the DNA vaccines expressing fusion proteins of PA63 with VP22. Conclusion VP22 fusion does not improve the protection of A/J mice against live spore challenge following immunisation of DNA vaccines expressing PA63.
Collapse
Affiliation(s)
- Stuart D Perkins
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 OJQ, UK
| | - Helen C Flick-Smith
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 OJQ, UK
| | - Helen S Garmory
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 OJQ, UK
| | - Angela E Essex-Lopresti
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 OJQ, UK
| | - Freda K Stevenson
- Tenovus Laboratory, University of Southampton Hospital NHS Trust, Southampton, SO16 6YD, UK
| | - Robert J Phillpotts
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 OJQ, UK
| |
Collapse
|
18
|
Roy V, Qiao J, de Campos-Lima P, Caruso M. Direct evidence for the absence of intercellular trafficking of VP22 fused to GFP or to the herpes simplex virus thymidine kinase. Gene Ther 2005; 12:169-76. [PMID: 15483667 DOI: 10.1038/sj.gt.3302394] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The treatment of solid tumors by retroviral delivery of the herpes simplex virus thymidine kinase (TK) followed by ganciclovir (GCV) treatment has so far shown only limited success in patients. One major drawback in this approach is the lack of efficient in vivo gene delivery to cancer cells. Although, the transduction of every single tumor cell is not a requirement since the bystander effect (BE) mediated by gap junctions allows the diffusion of the toxic GCV metabolites from TK-expressing cells toward untransduced cells. To render the TK/GCV approach more potent, and independent of the level of gap junctions, we have tested the efficiency of a TK mutant (TK30) fused to VP22, a herpes simplex protein that seems to be capable of intercellular trafficking. We failed to detect an increase in the BE with cells expressing VP22 fused to TK30 versus cells containing TK30 alone, and this result forced us to reinvestigate the trafficking properties of VP22. Using very sensitive Western blot and fluorescence assays, we were not able to detect the spread of VP22 fused either to TK30 or GFP. These results indicate that VP22 cannot be used as a cargo to translocate TK30 or GFP.
Collapse
Affiliation(s)
- V Roy
- Le Centre de Recherche en Cancérologie de l'Université Laval, L'Hôtel-Dieu de Québec, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | | | | | | |
Collapse
|
19
|
Perkins SD, Hartley MG, Lukaszewski RA, Phillpotts RJ, Stevenson FK, Bennett AM. VP22 enhances antibody responses from DNA vaccines but not by intercellular spread. Vaccine 2005; 23:1931-40. [PMID: 15734065 DOI: 10.1016/j.vaccine.2004.10.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Accepted: 10/18/2004] [Indexed: 11/29/2022]
Abstract
In some species DNA vaccines elicit potent humoral and cellular immune responses. However, their performance in humans and non-human primates is less impressive. There are suggestions in the literature that an increase in the intercellular distribution of protein expressed from a DNA vaccine may enhance immunogenicity. We incorporated the Herpes Simplex Virus type 1 (HSV) VP22 gene, which encodes a protein that has been described as promoting intercellular spread, into a DNA vector in which it was fused to enhanced green fluorescent protein (EGFP). Following transfection of the plasmid DNA into mammalian cells, distribution of the fusion protein VP22-EGFP was not increased compared to EGFP alone. Furthermore, we found no evidence to suggest that VP22 was capable of mediating intercellular spread. However, when these constructs were used as DNA vaccines to immunise mice, antibody levels specific to EGFP were significantly enhanced when EGFP was fused to VP22. These data suggest that amplification of the immune response may occur via mechanisms other than VP22-mediated intercellular spread of antigen.
Collapse
Affiliation(s)
- Stuart D Perkins
- Biomedical Sciences Department, Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 OJQ, UK.
| | | | | | | | | | | |
Collapse
|
20
|
Greco O, Joiner MC, Doleh A, Scott SD. VP22-mediated intercellular transport for suicide gene therapy under oxic and hypoxic conditions. Gene Ther 2005; 12:974-9. [PMID: 15729368 DOI: 10.1038/sj.gt.3302482] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
During herpes simplex virus type 1 (HSV 1) infection, the tegument protein VP22 is exported from infected cells to the nuclei of surrounding uninfected cells. These intercellular transport characteristics have prompted the exploitation of VP22 fusion proteins for cancer gene therapy, with the goal of maximizing the bystander effect. Since solid tumors contain hypoxic cell populations that are often refractive to therapy, for efficient targeting, it would be optimal if VP22 functioned even at reduced oxygen concentrations. In the present work, VP22 activity under hypoxic conditions was examined for the first time. Plasmid-transfected human glioma U87-MG and U373-MG cells expressing VP22 fused to the green fluorescent protein (GFP) showed protein export to untransfected cells under tumor oxygenation conditions (0-5% O(2)). For suicide gene therapy, VP22 activity was demonstrated under hypoxia by coupling VP22 to the HSV thymidine kinase (HSVtk). In the presence of the prodrug ganciclovir, cell cultures expressing VP22-HSVtk showed a significant increase in toxicity compared with cells transfected with a construct containing HSVtk only, under all tested conditions. To allow effective suicide gene therapy and simultaneous visualization of therapeutic enzyme localization, a triple fusion protein GFP-HSVtk-VP22 was engineered. Functionality of all components was demonstrated under oxia and hypoxia.
Collapse
Affiliation(s)
- O Greco
- Department of Radiation Oncology, Wayne State University and Karmanos Cancer Institute, Hudson Webber CRC, Detroit, MI, USA
| | | | | | | |
Collapse
|
21
|
Zheng C, Babiuk LA, van Drunen Littel-van den Hurk S. Bovine herpesvirus 1 VP22 enhances the efficacy of a DNA vaccine in cattle. J Virol 2005; 79:1948-53. [PMID: 15650221 PMCID: PMC544085 DOI: 10.1128/jvi.79.3.1948-1953.2005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
For this study, the intercellular trafficking ability of bovine herpesvirus 1 (BHV-1) VP22 was applied to improve the efficacy of a DNA vaccine in calves. A plasmid encoding a truncated version of glycoprotein D (tgD) fused to VP22 was constructed. The plasmid encoding tgD-VP22 elicited significantly enhanced and more balanced immune responses than those induced by a plasmid encoding tgD. Furthermore, protection against a BHV-1 challenge was obtained in calves immunized with the plasmid encoding tgD-VP22, as shown by significant reductions in viral excretion. However, less significant protection was observed for animals vaccinated with the tgD-expressing plasmid, correlating with the lower level of immunity observed prechallenge. This is the first report of the use of VP22 as a transport molecule in the context of a DNA vaccine for a large animal species.
Collapse
Affiliation(s)
- Chunfu Zheng
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Rd., Saskatoon, SK S7N 5E3, Canada
| | | | | |
Collapse
|
22
|
Qiu Z, Zhu J, Harms JS, Friedrichsen J, Splitter GA. Bovine Herpesvirus VP22 Induces Apoptosis in Neuroblastoma Cells by Upregulating the Expression Ratio of Bax to Bcl-2. Hum Gene Ther 2005; 16:101-8. [PMID: 15703493 DOI: 10.1089/hum.2005.16.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Herpesvirus tegument protein VP22 has been shown to have biotherapeutic potential in tumor gene therapy. Some studies indicate that VP22 may enhance the transfer efficiency of therapeutic proteins by delivering them to more cells while trafficking. Our previous study showed that bovine herpesvirus VP22 (BVP22) enhanced equine herpesvirus thymidine kinase-ganciclovir (Etk-GCV) suicide gene therapy by an unknown intracellular effect. In this study, the interaction between BVP22 and host tumor cells was studied in neuroblastoma NXS2 cells. Cell cycle analysis was performed to determine whether BVP22 possesses biotherapeutic potential by altering the cell cycle, making cells more sensitive to therapeutic genes. As a result, the cell cycle was not affected by the transfection of BVP22 into NXS2 cells. However, cytotoxicity induced by BVP22 was observed in NXS2 cells on the second and third days after transient transfection. Further, analyses of caspase-3 activity and apoptosis suggested that BVP22 induces apoptosis in host tumor cells by upregulating the expression ratio of Bax to Bcl-2.
Collapse
Affiliation(s)
- Zhaohua Qiu
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
23
|
Sheridan PJ, Lawrie A, Crossman DC, Holt CM, Newman CM. VP22-mediated intercellular transport correlates with enhanced biological activity of MybEngrailed but not (HSV-I) thymidine kinase fusion proteins in primary vascular cells following non-viral transfection. J Gene Med 2005; 7:375-85. [PMID: 15543525 DOI: 10.1002/jgm.679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The intercellular transport properties of the herpes simplex virus (HSV) protein VP22 have been harnessed to enhance the effectiveness of viral gene transfer. We investigated the intercellular transport and biological effects of VP22 fused with the dominant negative c-Myb chimera, MybEngrailed (MybEn) and HSV-I thymidine kinase (TK), in primary vascular smooth muscle cells (VSMC) following non-viral transfection. MATERIALS AND METHODS Porcine VSMC transfected with plasmids encoding MybEn, TK and their respective N- and C-terminal VP22 fusion proteins were assayed for the extent and distribution of transgene expression (by immunohistochemistry), culture growth and apoptosis. RESULTS The N-terminal MybEn fusion with VP22 (MybEnVP22) and both TK fusions, but not VP22MybEn, exhibited intercellular spread from primary transfected to up to 200 surrounding cells. pMybEnVP22-transfected cultures exhibited growth inhibition and apoptosis rates that were 10.6 +/- 3.6 and 3.2 +/- 1.0 fold higher than in pMybEn-transfected cultures; pVP22MybEn-transfected cultures showed no difference in these parameters. pTK-transfected cultures underwent 60-70% cell death in the presence of ganciclovir despite <2% primary transfection, which was not increased in cultures transfected with plasmids encoding VP22-TK fusions. CONCLUSIONS The close correlation between immunocytochemical and biological assays suggests that intercellular transport is crucial to the enhanced biological activity of the MybEnVP22 fusion. The "intrinsic" bystander activity of TK was 4-fold greater than was "engineered" by VP22 fusion, probably reflecting the abundance of gap junctions between VSMC. VP22 fusion may enhance the efficiency of non-viral gene delivery when combined with the appropriate therapeutic transgene, target tissue and transfection method.
Collapse
Affiliation(s)
- Paul J Sheridan
- Cardiovascular Research Unit, Division of Clinical Sciences (North), Clinical Sciences Centre, University of Sheffield, Northern General Hospital, Sheffield S5 7AU, UK.
| | | | | | | | | |
Collapse
|
24
|
Eum WS, Kim DW, Hwang IK, Yoo KY, Kang TC, Jang SH, Choi HS, Choi SH, Kim YH, Kim SY, Kwon HY, Kang JH, Kwon OS, Cho SW, Lee KS, Park J, Won MH, Choi SY. In vivo protein transduction: biologically active intact pep-1-superoxide dismutase fusion protein efficiently protects against ischemic insult. Free Radic Biol Med 2004; 37:1656-69. [PMID: 15477017 DOI: 10.1016/j.freeradbiomed.2004.07.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2004] [Accepted: 07/22/2004] [Indexed: 11/20/2022]
Abstract
Reactive oxygen species (ROS) are implicated in reperfusion injury after transient focal cerebral ischemia. The antioxidant enzyme Cu,Zn-superoxide dismutase (SOD) is one of the major means by which cells counteract the deleterious effects of ROS after ischemia. Recently, we reported that denatured Tat-SOD fusion protein is transduced into cells and skin tissue. Moreover, PEP-1 peptide, which has 21 amino acid residues, is a known carrier peptide that delivers full-length native proteins in vitro and in vivo. In the present study, we investigated the protective effects of PEP-1-SOD fusion protein after ischemic insult. A human SOD gene was fused with PEP-1 peptide in a bacterial expression vector to produce a genetic in-frame PEP-1-SOD fusion protein. The expressed and purified fusion proteins were efficiently transduced both in vitro and in vivo with a native protein structure. Immunohistochemical analysis revealed that PEP-1-SOD injected intraperitoneally (i.p.) into mice can have access into brain neurons. When i.p.-injected into gerbils, PEP-1-SOD fusion proteins prevented neuronal cell death in the hippocampus caused by transient forebrain ischemia. These results suggest that the biologically active intact forms of PEP-1-SOD provide a more efficient strategy for therapeutic delivery in various human diseases related to this antioxidant enzyme or to ROS, including stroke.
Collapse
Affiliation(s)
- Won Sik Eum
- Department of Genetic Engineering, Research Institute for Bioscience and Biotechnology, Hallym University, 1-1 Okchon-Dong, Chunchon, Kangwon-Do, Korea, 200-702, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Garg S, Oran AE, Hon H, Jacob J. The hybrid cytomegalovirus enhancer/chicken beta-actin promoter along with woodchuck hepatitis virus posttranscriptional regulatory element enhances the protective efficacy of DNA vaccines. THE JOURNAL OF IMMUNOLOGY 2004; 173:550-8. [PMID: 15210816 DOI: 10.4049/jimmunol.173.1.550] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DNA vaccines represent a novel and powerful alternative to conventional vaccine approaches. They are extremely stable and can be produced en masse at low cost; more importantly, DNA vaccines against emerging pathogens or bioterrorism threats can be quickly constructed based solely upon the pathogen's genetic code. The main drawback of DNA vaccines is that they often induce lower immune responses than traditional vaccines, particularly in nonrodent species. Thus, improving the efficacy of DNA vaccines is a critical issue in vaccine development. In this study we have enhanced the efficacy of DNA vaccines by adopting strategies that increase gene expression. We generated influenza-hemagglutinin (HA)-encoding DNA vaccines that contain the hybrid CMV enhancer/chicken beta-actin (CAG) promoter and/or the mRNA-stabilizing post-transcriptional regulatory element from the woodchuck hepatitis virus (WPRE). Mice were immunized with these DNA vaccines, and the influenza-HA-specific cellular and humoral immune responses were compared with a conventional, HA-encoding DNA vaccine whose gene expression was driven by the CMV immediate-early promoter (pCMV-HA). CAG promoter-driven DNA vaccines elicited significantly higher humoral and cellular immune responses compared with the pCMV-HA vaccine. DNA vaccines consisting of both CAG and WPRE elements (pCAG-HA-WPRE) induced the highest level of protective immunity, such that immunization with 10-fold lower DNA doses prevented death in 100% of the mice upon lethal viral challenge, whereas all mice immunized with the conventional pCMV-HA vaccine succumbed to influenza infection.
Collapse
Affiliation(s)
- Sanjay Garg
- Department of Microbiology and Immunology, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | | | | | | |
Collapse
|
26
|
Pastori RL, Klein D, Ribeiro MM, Ricordi C. DELIVERY OF PROTEINS AND PEPTIDES INTO LIVE CELLS BY MEANS OF PROTEIN TRANSDUCTION DOMAINS: POTENTIAL APPLICATION TO ORGAN AND CELL TRANSPLANTATION. Transplantation 2004; 77:1627-31. [PMID: 15201657 DOI: 10.1097/01.tp.0000119589.12467.20] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Ricardo L Pastori
- Diabetes Research Institute, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
27
|
Qiu Z, Harms JS, Zhu J, Splitter GA. Bovine herpesvirus tegument protein VP22 enhances thymidine kinase/ganciclovir suicide gene therapy for neuroblastomas compared to herpes simplex virus VP22. J Virol 2004; 78:4224-33. [PMID: 15047837 PMCID: PMC374295 DOI: 10.1128/jvi.78.8.4224-4233.2004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Accepted: 12/12/2003] [Indexed: 11/20/2022] Open
Abstract
Herpesvirus tegument protein VP22 can enhance the effect of therapeutic proteins in gene therapy, such as thymidine kinase (tk) and p53; however, the mechanism is unclear or controversial. In this study, mammalian expression vectors carrying bovine herpesvirus 1 (BHV-1) VP22 (BVP22) or herpes simplex virus type 1 (HSV-1) VP22 (HVP22) and equine herpesvirus type 4 (EHV-4) tk (Etk) were constructed in order to evaluate and compare the therapeutic potentials of BVP22 and HVP22 to enhance Etk/ganciclovir (Etk/GCV) suicide gene therapy for neuroblastomas by GCV cytotoxicity assays and noninvasive bioluminescent imaging in vitro and in vivo. BVP22 enhanced Etk/GCV cytotoxicity compared to that with HVP22 both in vitro and in vivo. However, assays utilizing a mixture of parental and stably transfected cells indicated that the enhancement was detected only in transfected cells. Thus, the therapeutic potential of BVP22 and HVP22 in Etk/GCV suicide gene therapy in this tumor system is not due to VP22 delivery of Etk into surrounding cells but rather is likely due to an enhanced intracellular effect.
Collapse
Affiliation(s)
- Zhaohua Qiu
- Department of Animal Health and Biomedical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Friedrich C Luft
- Franz-Volhard-Klinik, Humboldt University of Berlin, Wiltbergstrasse 50, 13125, Berlin-Buch, Germany.
| |
Collapse
|
29
|
Kueltzo LA, Middaugh CR. Nonclassical transport proteins and peptides: an alternative to classical macromolecule delivery systems. J Pharm Sci 2003; 92:1754-72. [PMID: 12949995 DOI: 10.1002/jps.10448] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The number of peptides and proteins known to exhibit nonclassical transport activity has increased significantly in recent years. In most cases, these entities have been studied in relation to their ability to deliver high molecular weight compounds, including proteins and DNA, for the ultimate purpose of developing new drug delivery strategies. In this review, an overview of the various types of vectors is presented. The in vitro and in vivo delivery successes of this technology, as well as preliminary therapeutic efforts, are described. Although a comprehensive mechanism of nonclassical transport has not yet been clearly established, we propose a straightforward model based on the cationic nature of the vectors and the need for lack of highly organized structure. In this hypothesis we suggest that the movement of polycations is mediated by a network of extra- and intracellular polyanions while transport across the bilayer is facilitated by cation-pi interactions between the vectors' basic groups and aromatic amino acid side chains in the bilayer spanning helices of membrane proteins.
Collapse
Affiliation(s)
- Lisa A Kueltzo
- Department of Pharmaceutical Chemistry, University of Kansas, 2095 Constant Ave., Lawrence, Kansas 66047, USA
| | | |
Collapse
|
30
|
Rutjes SA, Bosma PJ, Rohn JL, Noteborn MHM, Wesseling JG. Induction of insolubility by herpes simplex virus VP22 precludes intercellular trafficking of N-terminal Apoptin-VP22 fusion proteins. J Mol Med (Berl) 2003; 81:558-65. [PMID: 12879152 DOI: 10.1007/s00109-003-0457-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2003] [Accepted: 05/23/2003] [Indexed: 10/26/2022]
Abstract
The herpes simplex virus protein VP22 has the intriguing ability to deliver proteins from an expressing cell to neighboring cells. Fusion of VP22 to Apoptin, a protein that induces apoptosis in tumor cells but not in normal cells, might enhance the delivery of Apoptin. To analyze this hypothesis two fusion proteins of VP22 and full-length Apoptin were constructed, namely VP22-VP3 and VP3-VP22, and their apoptosis-inducing ability and intercellular spreading behavior were analyzed by transfection in tumor cells. While both of the Apoptin-VP22 fusion proteins retained the capacity to kill tumor cells, neither of them showed intercellular trafficking. To determine whether the presence of a nuclear localization signal in the C-terminus of Apoptin caused nuclear retention of the fusion protein and the subsequent lack of intercellular spreading, VP22 was fused to the biologically active N-terminal part (residues 1-69) of Apoptin (VP3n), which lacks the nuclear localization signal. However, analysis of the VP3n-VP22 fusion constructs gave no evidence of intercellular transport. A more careful inspection of different fractions of cell lysates expressing Apoptin with or without fusion to VP22 revealed that both the full-length Apoptin protein and its fusion with VP22 are insoluble. Despite the fact that VP3n was found to be soluble on its own, which could make it amenable to transport by VP22, the VP3n-VP22 fusion proteins were present exclusively in the insoluble fraction. We hypothesize that the N-terminal multimerization domain of Apoptin cooperates with VP22 to facilitate aggregation with cellular proteins, thereby inducing insolubility. From these results we conclude that, depending on the fusion partner, VP22 can have a negative effect on the solubility of fusion proteins, which consequently precludes intercellular trafficking. Such properties should be taken into account when establishing new VP22-mediated protein transduction systems.
Collapse
Affiliation(s)
- Saskia A Rutjes
- Academic Medical Center, Laboratory of Experimental Hepatology, AMC Liver Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Stroh C, Held J, Samraj AK, Schulze-Osthoff K. Specific inhibition of transcription factor NF-kappaB through intracellular protein delivery of I kappaBalpha by the Herpes virus protein VP22. Oncogene 2003; 22:5367-73. [PMID: 12917639 DOI: 10.1038/sj.onc.1206544] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In many cancers, a high constitutive activation of transcription factor NF-kappaB has been implicated in tumor progression and apoptosis resistance, making NF-kappaB an attractive target for cancer therapy. Here, we describe the specific inhibition of NF-kappaB by the intracellular delivery of IkappaBalpha through VP22-mediated protein transduction. The Herpes virus protein VP22 has attracted great attention in gene therapy, because of its ability to migrate from an original expressing cell into surrounding recipient cells, resulting in high levels of protein transduction. To evaluate the use of VP22 as a vehicle for NF-kappaB inhibition, we expressed several versions of VP22-IkappaBalpha fusion proteins in baculovirus, bacteria, and mammalian cells. While we could not detect transcellular migration of different VP22-IkappaBalpha constructs, interestingly, baculovirally expressed VP22-IkappaBalpha was efficiently delivered into cells after exogenous administration. The purified and imported VP22-IkappaBalpha retained its function and efficiently inhibited both constitutive and inducible NF-kappaB activation. We further show that the 34 C-terminal amino acids of VP22 were sufficient for the import property, suggesting also that the ability of intercellular migration and cellular import are not linked to each other. Together, our results demonstrate that recombinant VP22 acts as an efficient vehicle for the exogenous delivery of IkappaBalpha and, moreover, might find applications to block NF-kappaB activation specifically.
Collapse
Affiliation(s)
- Christopher Stroh
- Institute of Molecular Medicine, University of Düsseldorf, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| | | | | | | |
Collapse
|
32
|
Weisbart RH, Miller CW, Chan G, Wakelin R, Ferreri K, Koeffler HP. Nuclear delivery of p53 C-terminal peptides into cancer cells using scFv fragments of a monoclonal antibody that penetrates living cells. Cancer Lett 2003; 195:211-9. [PMID: 12767530 DOI: 10.1016/s0304-3835(03)00151-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
scFv fragments of a monoclonal antibody that penetrates living cells and localizes in nuclei were designed as fusion proteins with C-terminal p53 peptides and tested for restoring p53 function in p53 mutant cancer cells. scFv fragments transported a 30-mer C-terminal peptide of p53 into cancer cells and induced cellular cytotoxicity in contrast to scFv fragments alone and other scFv-p53 fusion peptides. Cellular toxicity was not observed with scFv fragments containing a single mutation in VH that prevented antibody penetration. Our results demonstrate the potential efficacy of antibody scFv fragments as a nuclear delivery system in cancer cells.
Collapse
Affiliation(s)
- Richard H Weisbart
- Department of Medicine, Division of Rheumatology, Veterans Affairs Greater Los Angeles Health Care System, Sepulveda, CA 91343, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Zavaglia D, Favrot MC, Eymin B, Tenaud C, Coll JL. Intercellular trafficking and enhanced in vivo antitumour activity of a non-virally delivered P27-VP22 fusion protein. Gene Ther 2003; 10:314-25. [PMID: 12595890 DOI: 10.1038/sj.gt.3301904] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
VP22, a structural protein from herpes simplex virus type I, exhibits the unique property of intercellular trafficking. This protein is exported from primary expressing cells and subsequently imported into neighbouring cells. This property is conserved when VP22 is genetically fused to a protein, making it a promising tool to enhance the delivery of a gene product. We chose to study the intercellular transport and biological effect of a fusion protein between the putative tumour suppressor gene p27(Kip1) and VP22. We show that in vitro, P27VP22 is able to spread as efficiently as VP22. Functionality of the P27VP22 protein was demonstrated by its ability to inhibit cyclin/CDK2 complexes activity. In proliferation and clonogenicity assays, transfection with the P27VP22 plasmid resulted in a stronger cell growth inhibition when compared to transfection with the p27(Kip1) vector. In vivo, sub cutaneous tumours established in nude mice were injected with naked DNA encoding P27 or P27VP22. Our results show that P27VP22 can spread in vivo and that injections of the P27VP22 plasmid resulted in a significantly greater antitumour activity than injections of the P27 plasmid. This study confirms the usefulness of VP22-mediated delivery and suggests that P27VP22 may have applications in cancer gene therapy.
Collapse
Affiliation(s)
- D Zavaglia
- Groupe de Recherche sur le Cancer du Pumon, Equipe INSERM 9924, Institut Albert Bonnoit, La Tronche cedex, France
| | | | | | | | | |
Collapse
|
34
|
Cashman SM, Sadowski SL, Morris DJ, Frederick J, Kumar-Singh R. Intercellular trafficking of adenovirus-delivered HSV VP22 from the retinal pigment epithelium to the photoreceptors--implications for gene therapy. Mol Ther 2002; 6:813-23. [PMID: 12498777 DOI: 10.1006/mthe.2002.0806] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenovirus (Ad)-mediated gene transfer is a promising technology for therapy of a wide variety of genetic disorders of the retina. The tropism of Ad vectors limits their utility to cells that express the coxsackie-adenovirus receptor. Upon ocular delivery, Ad vectors primarily infect the retinal pigment epithelium (RPE) and the Müller cells of the retina. However, the most frequent blinding diseases such as retinitis pigmentosa and age-related macular degeneration are associated with the expression of mutant proteins in the photoreceptors. In this study we demonstrate that adenovirus-delivered heterologous proteins fused to the herpes simplex virus tegument protein VP22 can translocate from infected cells to uninfected cells in culture and in vivo. We tested three different ocular cell lines, specifically Y79, RPE-J, and Chang C. We show that there is a 3.25-fold increase in the number of Y79 cells that take up GFP mediated by the intercellular trafficking properties of VP22. Our data are based on FACS analysis of living cells and there was no need for cell fixation for the effect to be observed. When adenovirus expressing a VP22-GFP fusion was injected into the subretinal space of adult mice, the VP22-GFP fusion peptides translocated from the RPE to all of the other layers of the retina, including the outer nuclear layer, which contains the photoreceptor cell bodies. Our study has significant implications for a wide variety of diseases of the retina and other organ systems.
Collapse
Affiliation(s)
- Siobhan M Cashman
- Department of Ophthalmology, University of Utah, Salt Lake City, Utah 84112-5330, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
Tissue regeneration in humans is limited and excludes vitals organs like heart and brain. Transformation experiments with oncogenes like T antigen have shown that retrodifferentiation of the respective cells is possible but hard to control. To bypass the risk of cancer formation a protein therapy approach has been developed. The transient delivery of proteins rather than genes could still induce terminally-differentiated cells to reenter the cell cycle. This approach takes advantage of protein-transducing domains that mediate the transfer of cargo proteins into cells. The goal of this brief review is to outline the basics of protein transduction and to discuss potential applications for tissue regeneration.
Collapse
|
36
|
Zender L, Köck R, Eckhard M, Frericks B, Gösling T, Gebhardt T, Drobek S, Galanski M, Kühnel F, Manns M, Kubicka S. Gene therapy by intrahepatic and intratumoral trafficking of p53-VP22 induces regression of liver tumors. Gastroenterology 2002; 123:608-18. [PMID: 12145813 DOI: 10.1053/gast.2002.34756] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS VP22-mediated intercellular transport provides an approach to deliver functional chimeric proteins into a high percentage of target cells. The aim of this study was to evaluate the efficacy of p53/VP22 fusion protein in gene therapy of liver tumors. METHODS Expression vectors of N- and C-terminal fusion proteins of p53 and VP22 were subcloned and transcriptional properties of chimeric proteins were assessed by luciferase assays. Adenoviral vectors expressing p53 wild type (AdGFP/p53wt) and p53-VP22 (AdGFP/p53-VP22) were generated to investigate the VP22-mediated spreading in normal liver and in liver tumors in vivo by green fluorescent protein fluorescence and p53 immunohistochemistry. Gene therapy was investigated in subcutaneous and preclinical orthotopic animal tumor models after subcutaneous and intra-arterial administration of the adenoviruses, and tumor growth was assessed by direct calibration and magnetic resonance imaging. RESULTS p53-VP22 showed enhanced transcriptional activity compared with p53 wild type. VP22-mediated intercellular transport of p53 could be observed in the normal liver and in liver tumors in vivo and was correlated with increased antitumor efficacy of gene therapy and improved survival of the animals. CONCLUSIONS Fusion of VP22 to p53 strongly improves the results of p53 replacement gene therapy. Furthermore, the demonstrated VP22-mediated intercellular transport in the liver could be important for other strategies in liver gene therapy, providing a tool for enhancing the effect of gene therapy in liver diseases such as metabolic disorders or viral hepatitis.
Collapse
Affiliation(s)
- Lars Zender
- Department of Gastroenterology and Hepatology, Medical School Hannover, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Zender L, Kühnel F, Köck R, Manns M, Kubicka S. VP22-mediated intercellular transport of p53 in hepatoma cells in vitro and in vivo. Cancer Gene Ther 2002; 9:489-96. [PMID: 12032659 DOI: 10.1038/sj.cgt.7700465] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2002] [Indexed: 12/13/2022]
Abstract
The capacity of VP22 chimeric proteins to spread from the primary transduced cell to surrounding cells could improve gene therapy approaches, especially in cancer therapy. However, there are conflicting data about VP22-mediated intercellular trafficking in different studies. To assess the role of VP22 in gene therapy of hepatocellular carcinomas (HCCs) we constructed expression vectors for N- and C-terminal versions of VP22-p53 fusion proteins and investigated the VP22-mediated shuttle effect in hepatoma cells by cotransfection experiments. VP22-mediated trafficking was not detectable in hepatoma cells in vitro by fluorescence microscopy, but reporter gene transactivation assays demonstrated intercellular trafficking of functional VP22-p53 in vitro. For in vivo experiments, the recombinant adenoviruses Ad5CMVp53 and Ad5CMVp53-VP22 were constructed. In contrast to the in vitro experiments intercellular trafficking of VP22-p53 could be observed in subcutaneous tumors of hepatoma cells by fluorescence microscopy, indicating a stronger shuttle effect in solid tumors compared to cell culture experiments. Because spread of p53-VP22 in liver tumors was correlated with enhanced apoptosis of hepatoma cells VP22-mediated trafficking of potential therapeutic proteins may improve the results of gene therapy of HCCs.
Collapse
Affiliation(s)
- Lars Zender
- Department of Gastroenterology and Hepatology, Medical School Hannover, Hannover 30625, Germany
| | | | | | | | | |
Collapse
|
39
|
Abstract
For most patients with advanced or multifocal hepatocellular carcinoma (HCC) or with metastatic malignant liver disease treatment options are limited, resulting in a poor prognosis. Novel therapeutic strategies such as gene therapy are therefore urgently required. Gene therapeutic approaches use gene delivery systems (vectors) to introduce DNA constructs as therapeutic agents into living cells. Antitumour strategies include the reintroduction of tumour suppressor genes into tumour cells, the expression of foreign enzymes to render tumours susceptible to treatment with chemotherapeutic agents and the enhancement of tumour immunogenicity by expressing immunomodulatory genes or by genetic vaccination with tumour antigens. Furthermore, gene therapy may be also used for anti-angiogenesis to reduce tumour growth and metastatic potential. Other novel approaches aim at the development of genetically altered replication competent viruses, which selectively replicate in tumour cells inducing cell lysis. Although most clinical trials of antitumour gene therapy so far have failed to induce strong therapeutic effects, further improvement of antitumour gene therapy may finally result in potent clinical treatment options for patients with malignant liver tumours.
Collapse
Affiliation(s)
- Leonhard Mohr
- Department of Medicine II, University Hospital Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany.
| | | | | |
Collapse
|
40
|
Abstract
Intracellular delivery of macromolecules remains problematic because of the bioavailability restriction imposed by the cell membrane. Recent studies on protein transduction domains have circumvented this barrier, however, and have resulted in the delivery of peptides, full-length proteins, iron beads, liposomes, and radioactive isotopes into cells in culture and animal models in vivo.
Collapse
Affiliation(s)
- Jehangir S Wadia
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, UCSD School of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0686, USA
| | | |
Collapse
|