Skip to main content

Genetic incorporation of the protein transduction domain of Tat into Ad5 fiber enhances gene transfer efficacy

Abstract

Background

Human adenovirus serotype 5 (Ad5) has been widely explored as a gene delivery vector for a variety of diseases. Many target cells, however, express low levels of Ad5 native receptor, the Coxsackie-Adenovirus Receptor (CAR), and thus are resistant to Ad5 infection. The Protein Transduction Domain of the HIV Tat protein, namely PTDtat, has been shown to mediate protein transduction in a wide range of cells. We hypothesize that re-targeting Ad5 vector via the PTDtat motif would improve the efficacy of Ad5-mediated gene delivery.

Results

In this study, we genetically incorporated the PTDtat motif into the knob domain of Ad5 fiber, and rescued the resultant viral vector, Ad5.PTDtat. Our data showed the modification did not interfere with Ad5 binding to its native receptor CAR, suggesting Ad5 infection via the CAR pathway is retained. In addition, we found that Ad5.PTDtat exhibited enhanced gene transfer efficacy in all of the cell lines that we have tested, which included both low-CAR and high-CAR decorated cells. Competitive inhibition assays suggested the enhanced infectivity of Ad5.PTDtat was mediated by binding of the positively charged PTDtat peptide to the negatively charged epitopes on the cells' surface. Furthermore, we investigated in vivo gene delivery efficacy of Ad5.PTDtat using subcutaneous tumor models established with U118MG glioma cells, and found that Ad5.PTDtat exhibited enhanced gene transfer efficacy compared to unmodified Ad5 vector as analyzed by a non-invasive fluorescence imaging technique.

Conclusion

Genetic incorporation of the PTDtat motif into Ad5 fiber allowed Ad5 vectors to infect cells via an alternative PTDtat targeting motif while retaining the native CAR-mediated infection pathway. The enhanced infectivity was demonstrated in both cultured cells and in in vivo tumor models. Taken together, our study identifies a novel tropism expanded Ad5 vector that may be useful for clinical gene therapy applications.

Background

Human adenovirus serotype 5 (Ad5) has been widely exploited as a gene delivery vector, owing largely to its superior gene delivery efficacy, minor pathological effect on humans, and easy manipulation in vitro. Several problems, however, have been identified in the course of development and application of Ad5-based gene therapy protocols, one of which is the inefficient gene delivery into target cells [1–3]. It is known that infection of Ad5 is initiated by attachment of its capsid fiber protein to the cell surface coxsackievirus adenovirus receptor (CAR), which is followed by interaction of its penton base with αv integrins that triggers the internalization of the viruses [4–7]. Many target cells, such as malignant tumor cells, are found to express very low level of CAR, and thus are resistant to Ad5 infection. Therefore, strategies to re-direct Ad5 infection via alternative receptors would be useful for gene therapy applications.

Since fiber, the capsid protein extruding from the Ad virion surface, is an essential mediator of Ad5 infection, fiber modification has been explored as a means to re-direct Ad5 tropism [1]. Ad5 fiber is composed of an N-terminal tail that is attached to a penton base on the virion surface, a shaft domain consisting of 22 repeats of a 15-amino acid residue motif, and a C-terminal globular domain, named knob, which functions as a receptor binding domain. Because of the essential role of the fiber knob domain in mediating Ad5 infection, knob modification could be one of the most effective ways to re-direct Ad5 tropism. Indeed, both genetic and non-genetic strategies have been shown to successfully retarget Ad5 vectors. For example, bi-specific adapter proteins that bind both the knob domain and an alternative receptor expressed on the surface of the target cells have been employed to re-direct Ad5 infection [8–11]. In addition, genetic incorporation of RGD peptide and/or a polylysine epitope into the knob domain allowed Ad5 to infect cells through alternative receptors (cell surface integrins for RGD and negatively charged epitopes such as heparan sulfate proteoglycans for polylysine), thus greatly improving the gene delivery efficacy Ad5 vectors in many target cells [12–15].

Protein transduction domains (PTD) or Cell Penetrating Peptides (CPP) are a class of small peptides that can traverse the plasma membrane of many, if not all, mammalian cells [16–20]. Among these peptides, the PTD of the Tat protein (PTDtat) of human immunodeficiency viruses types 1 and 2 (HIV-1 and HIV-2) has been one of the most widely studied PTDs. PTDtat consists of 11 highly basic amino acid residues, YGRKKRRQRRR [21, 22]. The mechanism of how PTDtat crosses the cell membrane has been intensively studied, but controversies remain [23–26]. Nonetheless, it is commonly agreed upon that the interaction between the positive charge of the PTD domain and the negative epitopes, in particular, the heparan sulfate proteoglycans expressed on cell membranes, plays an essential role in the internalization of PTDtat fusion proteins [17, 20, 27]. Further studies suggest that the interaction between PTDtat and heparan sulfate is specified by both charge and structure of the peptide and the proteoglycans [17, 27–30].

Given the potential importance of the PTDs in drug delivery, much interest has been generated in exploiting this system as a tool to deliver therapeutic molecules or particles into mammalian cells. PTDs have already been widely used in the field of protein therapy whereby PTDs are fused to the protein of interest, and used to deliver the heterologous protein into cultured cells [17, 20, 31]. Interestingly, it has been demonstrated in several mouse studies that PTDtat fusion proteins can be delivered into different tissues in vivo following systemic administration, and therapeutic benefits have been observed [32–35]. In addition, PTDs have been used to deliver other large molecules or particles including plasmids, liposomes, nanoparticles, phages and viruses, with variable efficiency [36–41]. In these applications, PTDs were conjugated to the vehicle of interest by incubation in coupling solutions. In other words, the coating of the vehicle was not based on genetic modification, but on ionic or other interactions between the peptides and the vehicle.

Because of the potency of PTDtat in mediating cellular uptake of small and large molecules, in this study, we attempted to re-direct Ad5 infection via the PTDtat pathway. Previous studies have demonstrated pre-treatment of Ad particles with chemically synthesized PTDs or bi-specific adaptor proteins composed of the extracellular domain of CAR and PTDs improved Ad infection [37, 42]. Nonetheless, intrinsic to these non-genetic modification strategies, the efficiency of retargeting depended on the affinity and stability of protein-protein interactions, and thus may be highly variable in different systems. In addition, a large amount of peptide or adaptor protein is seen to be required for in vivo investigations. Our study was designed to retarget Ad5 vectors to the PTDtat pathway using a genetic capsid modification strategy. We genetically incorporated the sequences encoding the PTDtat peptide into the 3' end of the Ad5 fiber gene, rescued the modified viruses, and characterized them in detail. Our data demonstrated that genetic modification of Ad5 fiber with the PTDtat motif greatly improved the efficacy of gene delivery in both cultured cells and in tumor models. Our study thus identified a novel tropism expanded Ad5 vector that may be useful for clinical gene therapy applications, especially for applications involving gene delivery into low-CAR expressing cells.

Results

Development of PTDtat-modified Ad5 vector – Ad5.PTDtat

As the receptor binding domain, the knob of the Ad5 fiber has been shown to be an effective site for incorporating foreign targeting motifs [12–15]. In this study, we genetically incorporated the PTDtat epitope into the C-terminal end of the fiber knob domain (Fig. 1). The Ad5 genome contains about 36 kilobases (kb) and is too large for direct modification using conventional cloning techniques. To achieve our goal, we therefore established a bacteria-based homologous recombination system for Ad5 fiber modification [15]. Using this system, the nucleotide sequences encoding PTDtat were incorporated into the 3'end of the fiber gene, immediately before the stop code. The modified Ad5 (Ad5.PTDtat) and the unmodified control (Ad5) were both replication deficient as their E1 region, which is essential for Ad5 replication, was replaced with a CMV promoter-driven green fluorescence protein (GFP) reporter gene. The viruses were rescued in 293 cells stably expressing Ad-E1 genes, and purified with CsCl gradient ultracentrifugation. The yield of Ad5.PTDtat total viral particles (VPs) and the ratio of VPs : plaque formation units (pfu) were in the same range as that of unmodified Ad5 viruses, suggesting that the modification did not interfere with virus formation (data not shown). The modification was confirmed by both polymerase chain reaction (PCR) and sequence analysis of the modified region of the viral genome using viral DNA from purified Ad5 and Ad5.PTDtat viruses (data not shown).

Figure 1
figure 1

Diagram of PTD tat modified Ad5 vector. (A) PTDtat peptide incorporated into the fiber knob domain. (B) Structural diagram of Ad5 and Ad5.PTDtat vector. The PTDtat motif was incorporated at the C-terminal end of the fiber.

CAR-binding activity of Ad5.PTDtat

Unmodified Ad5 viruses interact with their native receptor CAR via the fiber knob domain. We thus examined whether incorporation of PTDtat into the knob domain interfered with the Ad5-CAR interaction. An enzyme-linked immunosorbent assay (ELISA) was employed in this regard. In the assay, Ad5.PTDtat or Ad5 viral particles were immobilized in the wells of a 96-well maxi-sorp plate, and incubated with varying amounts of recombinant extracellular domain of CAR (sCAR) protein. After extensive washing, binding of sCAR to the viruses were assessed with an anti-CAR antibody and corresponding secondary antibody conjugated to alkaline phosphatase (AP). The OD405 readings resulting from the color reaction with an AP substrate correspond to the binding activity of sCAR to the viruses. As shown in Fig. 2, binding of sCAR to Ad5.PTDtat is similar to that of unmodified Ad5, suggesting the genetically modified vector Ad5.PTDtat maintained its ability to interact with the Ad5 native receptor, CAR.

Figure 2
figure 2

Ad5.PTD tat showed similar CAR-binding activity to unmodified Ad5 vector in an ELISA-based binding assay. In the experiment, 109 VPs of each viral vector were immobilized in the wells of a 96-well ELISA plate, and incubated with increasing concentrations of recombinant sCAR (extracellular domain of CAR, i.e. soluble CAR). The binding activity was detected by AP activity conjugated to detection antibodies.

Cell-binding activities of Ad5.PTDtat

The fiber knob domain of Ad is responsible for Ad5 binding to its target cells, which is the initial step in viral infection. Ad5.PTDtat was designed to re-direct Ad5 infection. We thus examined whether PTDtat modification had any effect on Ad5 binding to cells. To distinguish viruses bound to cells from viruses internalized into the cells, we performed a cell binding assay at 4°C since Ad internalization occurs through receptor-mediated endocytosis which is energy dependent, and is thus inhibited at 4°C [5, 7]. In the assay, Ad5.PTDtat or control Ad5 was incubated with cells expressing different levels of CAR at 4°C for 1 hour, and the bound viral particles were examined by a quantitative PCR assay which assessed the viral genome copies in the cell lysates. We found that Ad5.PTDtat exhibited a significant higher cell-binding activity in almost all of the cells we examined, including both high-CAR and low-CAR containing cells. Shown in Fig. 3 are results obtained in two representative cell lines: high-CAR expressing Hela cells, and low-CAR expressing U118MG cells [43, 44].

Figure 3
figure 3

PTD tat modification promoted Ad5 binding to cell surfaces. Binding of Ad5 and Ad5.PTDtat were examined in both high-CAR expressing Hela cells (A) and low-CAR expressing U118MG cells (B) at 4°C. The amount of viruses associated with the cells was determined by quantitative PCR after DNA isolation from the cell lysate, and the viral copy numbers were normalized to actin DNA in the samples. The * indicates p < 0.05 and ** indicates p < 0.01 as analyzed by the Student's t-test.

Enhanced gene transfer efficacy of Ad5.PTDtat

We further investigated the gene transfer efficacy of Ad5.PTDtat in a variety of cultured cells using the reporter GFP protein. Ad5.PTDtat vector or unmodified Ad5 was used to infect cells at different multiplicities of infection (MOIs). Two days after infection, we evaluated the transgene expression using a fluorescent microscope and a fluorescent plate reader. We found that Ad5.PTDtat showed more efficient gene delivery than unmodified Ad5 in all of the cells tested (Fig. 4). In particular, Ad5.PTDtat exhibited significantly higher gene transfer efficacy than unmodified Ad5 in the cells expressing low or medium levels of CAR such as RD cells, U118MG cells, and D65MG cells [43, 44]. In high-CAR expressing cells that are readily accessible to unmodified Ad5 vector, Ad5.PTDtat also showed enhanced infectivity, presumably because Ad5.PTDtat maintained the CAR-mediated infection pathway while gaining extra targeting activity through the PTDtat pathway (Fig. 4).

Figure 4
figure 4

Ad5.PTD tat exhibited enhanced gene transfer efficacy in a variety of tumor cells. Gene transfer efficacy was evaluated by use of a GFP reporter that was carried in the E1 region of each vector. In the assay, tumor cells expressing varying levels of CAR were infected with either Ad5 or Ad5.PTDtat at an MOI of 100 or 500 VPs/cell, and GFP expression was examined by fluorescence microscopy and a fluorescence plate reader. (A) Representative fluorescence images of low-CAR containing cells (RD), medium-CAR containing cells (D65MG) and high-CAR expressing cells (Hela) that were infected with Ad5 or Ad5.PTDtat at an MOI of 500 VPs/cell. (B) GFP expression in a variety of cells infected with either Ad5 or Ad5.PTDtat was quantified using a fluorescence plate reader.

Identification of pathways mediating Ad5.PTDtatinfection

Ad5.PTDtat showed enhanced gene delivery efficacy compared to unmodified Ad5 vectors. To confirm that this expanded tropism was mediated by the genetically incorporated targeting motif PTDtat, we performed a gene transfer assay in the presence of competitive inhibitors. It has been shown that the interaction between the positively charged PTDtat and the negatively charged cell surface epitopes such as heparan sulfate proteoglycans is essential for PTDtat mediated protein transduction. Heparin, the structural analogue of heparan sulfate, would thus be expected to inhibit PTDtat mediated infection. In addition, recombinant knob protein was used to block the native CAR-mediated Ad5 infection because it compete with Ad5 vectors for cell surface CAR. In low-CAR containing U118MG cells [44], due to the paucity of CAR, unmodified Ad5 showed poor gene transfer efficacy, and neither knob nor heparin had any effect on Ad5-mediated transgene expression (Fig. 5A). In contrast, Ad5.PTDtat exhibited efficient gene delivery into U118MG cells, which was completely inhibited by heparin, but not by the recombinant knob protein (Fig. 5A). These data demonstrated Ad5.PTDtat infected low-CAR expressing cells mainly through the incorporated PTDtat motif. In high-CAR containing A549 cells [43], infection of unmodified Ad5 was completely blocked by recombinant knob protein while heparin had little effect, confirming that unmodified Ad5 mainly infected cells through the CAR pathway (Fig. 5B). On the other hand, Ad5.PTDtat-mediated gene transfer was partially blocked by either knob or heparin, but completely blocked in the presence of both knob and heparin, suggesting Ad5.PTDtat could infect cells via both CAR and the PTDtat motif (Fig. 5B).

Figure 5
figure 5

Competitive inhibition assay showing the enhanced gene transfer efficacy of Ad5.PTD tat was mediated by the PTD tat motif. In this assay, recombinant knob protein (50 μg/ml) was used to block CAR-mediated viral infection, and heparin (100 μg/ml) was used to block PTDtat mediated infection. Infections were performed at an MOI of 100 VPs/cell. (A) In low-CAR expressing U118MG cells that were resistant to unmodified Ad5 vector, Ad5.PTDtat mediated efficient gene delivery and the efficacy was completely inhibited by heparin, while recombinant knob had little effect, suggesting the enhanced infectivity of Ad5.PTDtat in low-CAR expressing cells resulted from the PTDtat motif. (B) In high-CAR expressing A549 cells, Ad5.PTDtat mediated gene delivery was partially inhibited with either knob or heparin, while being completely inhibited in the presence of both inhibitors, suggesting Ad5.PTDtat infected high-CAR expressing cells via both CAR and PTDtat pathways.

In vivo gene transfer efficacy of Ad5.PTDtat

We next examined whether the infectivity-enhanced vector Ad5.PTDtat could deliver enhanced gene transfer efficacy in vivo. Since Ad5.PTDtat showed more profound infectivity enhancement for low-CAR expressing tumor cells in vitro, we assessed the in vivo gene delivery efficacy of the Ad5 vectors using tumor models established with low-CAR containing U118MG cells. After the tumors were established subcutaneously in athymic (nude) mice, PBS, unmodified Ad5, or Ad5.PTDtat vectors were injected into the tumors. The gene delivery efficacy of each vector was analyzed by non-invasive fluorescence imaging that detected GFP expression in live mice. As shown in Fig. 6A, Ad5.PTDtat-infected tumors showed more intensive green fluorescence signals than Ad5-infected tumors, while no signal was detected in PBS-injected tumors. Quantitative analysis of the green fluorescence signals revealed that Ad5.PTDtat-mediated GFP expression was significantly higher than that of unmodified Ad5 vector in the tumors (p < 0.01) (Fig. 6B). These data suggest the infectivity-enhanced Ad5.PTDtat vector could be a useful vector for in vivo gene delivery into tumors, which is essential for cancer gene therapy.

Figure 6
figure 6

PTD tat modification of Ad5 fiber enhanced in vivo gene delivery efficacy of the vector. In vivo gene delivery of Ad5.PTDtat was examined using a non-invasive fluorescence imaging technique in low-CAR expressing tumor models. 1010 VPs of Ad5 or Ad5.PTDtat were injected into the subcutaneous U118MG tumors, and in vivo green fluorescence images were acquired at different days post viral injection. (A) Representative in vivo images from PBS, Ad5, or Ad5.PTDtat injected mouse tumor models at day 7 after vector administration. The colors representing different intensities of signal are shown on the color bar. Ad5.PTDtat infection resulted in more intensive GFP signals than unmodified Ad5 vectors. (B) Quantitative analysis of the GFP intensity in the tumor model of each group. The * marks significant differences (p < 0.01) as analyzed by the Student's t-test.

Discussion

In this study, we sought to improve the gene transfer efficacy of Ad 5 vectors by genetic modification of the fiber knob domain with a PTDtat motif. Our data demonstrated the success of this strategy. The fiber modified Ad5 vector, Ad5.PTDtat, not only exhibited enhanced gene delivery efficiency of Ad5 vectors in low-CAR cells that are resistant to unmodified Ad5 infection, but also in high-CAR cells that are permissive to Ad5 infection. The enhanced infectivity of Ad5.PTDtat was found to be mediated by targeting of PTDtat to the negatively charged epitopes such as heparan sulfate containing proteoglycans on cell surface. In addition, we found PTDtat mediated Ad5.PTDtat infection is additive to native CAR-mediated infection as assessed by competitive inhibition assays, which was not unexpected since Ad5.PTDtat maintained full CAR-binding activity. More significantly, the enhanced gene delivery efficacy of Ad5.PTDtat was demonstrated in vivo using low-CAR U118MG tumor models, and employment of a recently developed non-invasive optical imaging system allowed us to visually detect the enhanced gene delivery in vivo.

As a cell penetrating peptide, PTDtat is capable of traversing the plasma membrane of mammalian cells. Since the initial description that PTDtat is responsible for the ability of the HIV Tat protein to enter mammalian cells, PTDtat has attracted tremendous interest as a drug delivery vehicle [16–20]. Further interest has been stimulated by the observation that PTDs can facilitate systemic delivery of biologically active recombinant proteins in vivo [32–35, 37]. Since inefficient gene delivery into target cells has been one of the major limitations in Ad5-mediated gene therapy, in this study, we attempted to employ PTDtat peptide to facilitate Ad5 mediated gene delivery. Employment of PTDs to facilitate virus infection has been investigated previously, but only using non-genetic methods [37, 42]. In particular, chemically synthesized PTDs or bi-specific adaptor proteins consisting of PTDs and the extracelluar domain of CAR have been used to coat Ad vectors. These strategies too resulted in enhanced gene delivery [37, 42]. Compared to the non-genetic methods, our genetically PTDtat modified vector has major advantages for two major reasons: 1) genetic modification allows stable interaction between Ad5 and the PTDtat targeting epitope, thus reducing the volatility associated with the affinity and stability of protein-protein interactions in the presence of different environmental factors. This is critical especially for in vivo applications; and 2) genetic modification does not require production of peptides or fusion proteins other than the viral vector, while large amounts of high quality protein/peptide production is required for non-genetic strategies (in addition to high quality production of the viral vectors), which is especially important for in vivo studies.

One issue associated with PTDtat-mediated protein delivery is the inefficient release of PTDtat fusion proteins from the endosomal compartments [24, 45–48]. It has been demonstrated that a large proportion of the PTDtat fusion protein remains trapped in non-cytosolic compartments even though it is efficiently taken up by the cells. This apparently would compromise the therapeutic effect of the fusion protein. In our study, we examined the distribution of Ad5.PTDtat particles in cells at various time points (from 0.5 hour to 4 hours) following addition of the viruses to the cells by immunofluorescent staining, and found that the distribution of Ad5.PTDtat inside the cells was similar to that of unmodified Ad5 vectors (data not shown). This indicates endosomal trapping is not significant, if any present at all, with Ad5.PTDtat infection of cells. In addition, the enhanced gene delivery mediated by Ad5.PTDtat confirmed that the virions were able to efficiently escape the endosomal compartment.

The potential utility of the infectivity-enhanced Ad5.PTDtat vector in cancer gene therapy was initially investigated in this study using low-CAR expressing tumor models. Indeed, many tumor cells have been shown to express very low levels of CAR, which is partially responsible for the low efficacy of Ad5 mediated cancer gene therapy in in vivo studies, especially in clinical trials [1–3]. The ability of Ad5.PTDtat to improve the gene delivery efficacy is attributable to the PTDtat motif, which binds to the negatively charged motifs expressed on cell surface, in particular, heparan sulfate containing proteoglycans that are widely expressed in a variety of cells including tumor cells [49–51]. In addition to cancer gene therapy, Ad5.PTDtat may also be applied in other gene therapy applications where infectivity-enhancement is beneficial. Infectivity-enhanced vectors will not only allow efficient gene delivery into low-CAR target cells, but also allow use of a reduced amount of viral vectors, thus reducing vector-associated toxicity.

Previous studies have developed several other infectivity-enhanced vectors, which include Ad5 vectors modified with RGD, polylysine, or knobs from other Ad serotypes [13–15, 52]. Since each of the modified vectors uses a unique extra targeting motif, the enhanced gene delivery efficacy in a specific cell type depends on the expression of individual receptors on its cell surface. Similar to PTDtat, the polylysine epitope, which is composed of a stretch of lysine residues, is highly basic, and can utilize heparan sulfate as its receptor. Nonetheless, the interaction between PTDtat and heparan sulfate is not only based on ionic intereactions, but also on the specific structures of the peptide and the proteoglycans [27–29]. Therefore, the choice of an infectivity-enhanced vector needs to be determined for each specific application involving gene delivery enhancement.

Conclusion

Our data showed that a genetically modified Ad5 vector, Ad5.PTDtat, maintained the ability to interact with its native receptor CAR, and delivered transgenes into both high-CAR and low-CAR cells more efficiently than the unmodified Ad5 vector. Our data further showed Ad5.PTDtat infected cells via both CAR and PTDtat pathways. More significantly, Ad5.PTDtat exhibited enhanced gene delivery in vivo in a tumor model, and thus may be useful for gene therapy applications involving low gene delivery efficacy.

Methods

Cell culture

The human embryonic kidney 293 cells stably transformed with Ad-E1 DNA, human lung carcinoma A549 cells, human cervix adenocarcinoma Hela cells, human embryonic rhabdomyosarcoma RD cells, and human glioma D65MG and U118MG cells were all obtained from the American Type Culture Collection (ATCC, Manassas, VA). The 293 cells, A549 cells and U118MG cells were cultured in Dulbecco's modified Eagle's medium/Ham's F12 medium (DMEM/F12) containing 10% fetal bovine serum (FBS) and 2 mM L-glutamine. Hela cells were cultured cultured in minimum essential Eagle medium (MEM) containing 10% FBS and 2 mM L-glutamine. Both RD and D65MG cells were cultured in DMEM containing 10% FBS and 2 mM L-glutamine. All of the cells were maintained at 37°C in a 5% CO2 humidified incubator.

Generation of the Ad5.PTDtatvector

Genetic modification of the Ad5 vector with PTDtat was achieved using our previously established fiber modification system [15]. In brief, the fiber shuttle vector containing a unique SnaBI restriction site immediately in front of the stop code of the fiber gene, named pNEB.PK.SnaBI, was used to generate a PTDtat modification. The sense and antisense oligonucleotides encoding the PTDtat motif, 5'-phos-ACT TTT TCA TAC ATT GCG CAA GAA GGC GGT GGA GGG TAT GGC AGG AAG AAG CGG AGA CAG CGA CGA AGA TAA TAA A-3' (sense) and 5'-phos-TTT ATT ATC TTC GTC GCT GTC TCC GCT TCT TCC TGC CAT ACC CTC CAC CGC CTT CTT GCG CAA TGT ATG AAA AAG T -3' (antisense), were annealed and cloned into the fiber shuttle vector pNEB.PK.SnaBI. This resulted in the fiber modified shuttle vector pNEB.PK.PTDtat. In order to incorporate the modified fiber into an Ad5 genome, pNEB.PK.PTDtat was linearized and recombined in Escherichia coli (E. coli) BJ5183 with a linearized Ad5 backbone plasamid pVK50 that contained the CMV promoter driven GFP reporter gene in its E1 region. After the positive recombinant plasmid, designated pAd5.PTDtat, was identified, stable and high quality plasmid was obtained from E. coli DH5α after re-transformation of the construct. The modification was confirmed by sequencing analysis.

The modified virus Ad5.PTDtat was rescued and purified as previously described [53]. In brief, the pAd5.PTDtat plasmid was digested with PacI (to release the viral genome), purified, and transfected into 293 cells stably expressing the complementary E1 genes. After the virus plaques formed, they were amplified in 293 cells, and purified utilizing a standard CsCl gradient protocol. The viral particle (VP) titer was determined using a conversion factor of 1.1 × 1012 VPs/ml per absorbance unit at 260 nm.

ELISA

The ELISA binding assay was performed essentially as described [15]. In brief, 109 VPs of either Ad5 or Ad5.PTDtat in 100 μl of 100 mM carbonate buffer (pH 9.5) was immobilized in each well of a 96-well maxisorp plate (Nunc, Roskilde, Denmark) by overnight incubation at 4°C. Following extensive washes with Tris-buffered saline (TBS) containing 0.05% Tween 20 (TBS-Tween), and blocking with 2% bovine serum albumin (BSA) in TBS-Tween, the viruses were incubated with varying amounts of purified recombinant sCAR. The binding of sCAR to the viruses was detected by incubation with anti-CAR antibody (Santa Cruz Biotechnology Inc., Santa Cruz, CA), followed by an AP-conjugated secondary antibody incubation. AP activity reflecting the amount of bound sCAR was determined using a color reaction with p-nitrophenyl phosphate (Sigma, St. Louis, MO) as recommended by the manufacturer. The absorbance at 405 nm (OD405) was obtained using PowerWaveHT 340 microplate reader (BioTek Instruments Inc., Winooski, VT).

Cell binding assay

Cells were cultured in 6-well plates until they were confluent. The plate was then cooled down on ice, and incubated with Ad5 or Ad5.PTDtat at an MOI of 5000 VPs/cell for one hour at 4°C. After washing cells twice with cold phosphate buffered saline (PBS) on ice, the cells were collected by incubation with Versene (0.53 mM EDTA). After two more washes with PBS, the cells were lysed and processed to isolate DNA (Qiagen Inc., Valencia, CA). The viral copy number in the DNA samples were obtained by quantitative PCR using primers designed for the E4 region of adenoviral genome. The data were normalized against actin DNA in each sample.

Gene transfer assay

Gene transfer efficacy of the viral vectors was assessed with the use of GFP reporter. In the assay, cells were plated in 24-well plates with a density of 105 cells per well the day before infection. Then the cells were infected with Ad5 or Ad5.PTDtat at MOIs of 100 or 500 VPs/cell as described previously [53]. Two days later, GFP expression was examined by fluorescence microscopy and quantified by a Synergy HT fluorescence plate reader (BioTek Instruments Inc., Winooski, VT).

Competitive inhibition assays

Low-CAR U118MG cells or high-CAR A549 cells were plated in 24-well plates at a density of 105 cells per well the day before infection. Viruses equivalent to an MOI of 100 VPs/cell were used for each infection. To block cell surface CAR, recombinant knob protein was pre-incubated with cells at a final concentration of 50 μg/ml prior to viral infection [54], and to block the PTDtat epitope, the viruses were pre-incubated with 100 μg/ml of heparin [15, 54]. Two hours after infection, the cells were washed with PBS, and refreshed with complete media containing 10% FBS. The cells were cultured for two days in the humidified 37°C, 5% CO2 incubator, and GFP microscopy was performed to examine the transgene expression.

In vivogene delivery

The subcutaneous tumors were established in athymic nude mice using 1 × 107 U118MG cells per tumor per mouse. After the tumors developed to ~0.5 cm in diameter, PBS or 1010 VPs of Ad5 or Ad.PTDtat were injected into each tumor (n = 6). GFP expression was analyzed at 3, 7, and 10 days post infection using a custom-built non-invasive optical imaging system described previously [55]. The mice were placed in the imaging chamber under anesthesia with 3% isoflurane. Green fluorescence images were acquired at f/8 with 20-second exposure using a combination of excitation filter HQ487/15× and emission filter D535/30m (Chroma Technology, Rockingham, VT) supported by WinView32 software (Roper Scientific Inc., Trenton, NJ). All of the procedures involving animals were approved by the Institutional Animal Care and Use Committee of the University of Alabama at Birmingham and performed according to their guidelines.

References

  1. Glasgow JN, Everts M, Curiel DT: Transductional targeting of adenovirus vectors for gene therapy. Cancer Gene Ther. 2006, 13 (9): 830-844. 10.1038/sj.cgt.7700928.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  2. Hedley SJ, Chen J, Mountz JD, Li J, Curiel DT, Korokhov N, Kovesdi I: Targeted and shielded adenovectors for cancer therapy. Cancer Immunol Immunother. 2006, 55 (11): 1412-1419. 10.1007/s00262-006-0158-2.

    Article  PubMed  CAS  Google Scholar 

  3. Rein DT, Breidenbach M, Curiel DT: Current developments in adenovirus-based cancer gene therapy. Future Oncol. 2006, 2 (1): 137-143. 10.2217/14796694.2.1.137.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  4. Bai M, Harfe B, Freimuth P: Mutations that alter an Arg-Gly-Asp (RGD) sequence in the adenovirus type 2 penton base protein abolish its cell-rounding activity and delay virus reproduction in flat cells. J Virol. 1993, 67 (9): 5198-5205.

    PubMed  CAS  PubMed Central  Google Scholar 

  5. Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A, Hong JS, Horwitz MS, Crowell RL, Finberg RW: Isolation of a common receptor for Coxsackie B viruses and adenoviruses 2 and 5. Science. 1997, 275 (5304): 1320-1323. 10.1126/science.275.5304.1320.

    Article  PubMed  CAS  Google Scholar 

  6. Louis N, Fender P, Barge A, Kitts P, Chroboczek J: Cell-binding domain of adenovirus serotype 2 fiber. J Virol. 1994, 68 (6): 4104-4106.

    PubMed  CAS  PubMed Central  Google Scholar 

  7. Wickham TJ, Mathias P, Cheresh DA, Nemerow GR: Integrins alpha v beta 3 and alpha v beta 5 promote adenovirus internalization but not virus attachment. Cell. 1993, 73 (2): 309-319. 10.1016/0092-8674(93)90231-E.

    Article  PubMed  CAS  Google Scholar 

  8. Dmitriev I, Kashentseva E, Rogers BE, Krasnykh V, Curiel DT: Ectodomain of coxsackievirus and adenovirus receptor genetically fused to epidermal growth factor mediates adenovirus targeting to epidermal growth factor receptor-positive cells. J Virol. 2000, 74 (15): 6875-6884. 10.1128/JVI.74.15.6875-6884.2000.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  9. Li HJ, Everts M, Pereboeva L, Komarova S, Idan A, Curiel DT, Herschman HR: Adenovirus tumor targeting and hepatic untargeting by a coxsackie/adenovirus receptor ectodomain anti-carcinoembryonic antigen bispecific adapter. Cancer Res. 2007, 67 (11): 5354-5361. 10.1158/0008-5472.CAN-06-4679.

    Article  PubMed  CAS  Google Scholar 

  10. Tang Y, Han T, Everts M, Zhu ZB, Gillespie GY, Curiel DT, Wu H: Directing adenovirus across the blood-brain barrier via melanotransferrin (P97) transcytosis pathway in an in vitro model. Gene Ther. 2007, 14 (6): 523-532. 10.1038/sj.gt.3302888.

    Article  PubMed  CAS  Google Scholar 

  11. Watkins SJ, Mesyanzhinov VV, Kurochkina LP, Hawkins RE: The 'adenobody' approach to viral targeting: specific and enhanced adenoviral gene delivery. Gene Ther. 1997, 4 (10): 1004-1012. 10.1038/sj.gt.3300511.

    Article  PubMed  CAS  Google Scholar 

  12. Belousova N, Krendelchtchikova V, Curiel DT, Krasnykh V: Modulation of adenovirus vector tropism via incorporation of polypeptide ligands into the fiber protein. J Virol. 2002, 76 (17): 8621-8631. 10.1128/JVI.76.17.8621-8631.2002.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Dmitriev I, Krasnykh V, Miller CR, Wang M, Kashentseva E, Mikheeva G, Belousova N, Curiel DT: An adenovirus vector with genetically modified fibers demonstrates expanded tropism via utilization of a coxsackievirus and adenovirus receptor-independent cell entry mechanism. J Virol. 1998, 72 (12): 9706-9713.

    PubMed  CAS  PubMed Central  Google Scholar 

  14. Wickham TJ, Roelvink PW, Brough DE, Kovesdi I: Adenovirus targeted to heparan-containing receptors increases its gene delivery efficiency to multiple cell types. Nat Biotechnol. 1996, 14 (11): 1570-1573. 10.1038/nbt1196-1570.

    Article  PubMed  CAS  Google Scholar 

  15. Wu H, Seki T, Dmitriev I, Uil T, Kashentseva E, Han T, Curiel DT: Double modification of adenovirus fiber with RGD and polylysine motifs improves coxsackievirus-adenovirus receptor-independent gene transfer efficiency. Hum Gene Ther. 2002, 13 (13): 1647-1653. 10.1089/10430340260201734.

    Article  PubMed  CAS  Google Scholar 

  16. Deshayes S, Morris MC, Divita G, Heitz F: Cell-penetrating peptides: tools for intracellular delivery of therapeutics. Cell Mol Life Sci. 2005, 62 (16): 1839-1849. 10.1007/s00018-005-5109-0.

    Article  PubMed  CAS  Google Scholar 

  17. Fittipaldi A, Giacca M: Transcellular protein transduction using the Tat protein of HIV-1. Adv Drug Deliv Rev. 2005, 57 (4): 597-608. 10.1016/j.addr.2004.10.011.

    Article  PubMed  CAS  Google Scholar 

  18. Joliot A, Prochiantz A: Transduction peptides: from technology to physiology. Nat Cell Biol. 2004, 6 (3): 189-196. 10.1038/ncb0304-189.

    Article  PubMed  CAS  Google Scholar 

  19. Snyder EL, Dowdy SF: Cell penetrating peptides in drug delivery. Pharm Res. 2004, 21 (3): 389-393. 10.1023/B:PHAM.0000019289.61978.f5.

    Article  PubMed  CAS  Google Scholar 

  20. Wadia JS, Dowdy SF: Transmembrane delivery of protein and peptide drugs by TAT-mediated transduction in the treatment of cancer. Adv Drug Deliv Rev. 2005, 57 (4): 579-596. 10.1016/j.addr.2004.10.005.

    Article  PubMed  CAS  Google Scholar 

  21. Ruben S, Perkins A, Purcell R, Joung K, Sia R, Burghoff R, Haseltine WA, Rosen CA: Structural and functional characterization of human immunodeficiency virus tat protein. J Virol. 1989, 63 (1): 1-8.

    PubMed  CAS  PubMed Central  Google Scholar 

  22. Vives E, Brodin P, Lebleu B: A truncated HIV-1 Tat protein basic domain rapidly translocates through the plasma membrane and accumulates in the cell nucleus. J Biol Chem. 1997, 272 (25): 16010-16017. 10.1074/jbc.272.25.16010.

    Article  PubMed  CAS  Google Scholar 

  23. Ferrari A, Pellegrini V, Arcangeli C, Fittipaldi A, Giacca M, Beltram F: Caveolae-mediated internalization of extracellular HIV-1 tat fusion proteins visualized in real time. Mol Ther. 2003, 8 (2): 284-294. 10.1016/S1525-0016(03)00122-9.

    Article  PubMed  CAS  Google Scholar 

  24. Fischer R, Kohler K, Fotin-Mleczek M, Brock R: A stepwise dissection of the intracellular fate of cationic cell-penetrating peptides. J Biol Chem. 2004, 279 (13): 12625-12635. 10.1074/jbc.M311461200.

    Article  PubMed  CAS  Google Scholar 

  25. Fittipaldi A, Ferrari A, Zoppe M, Arcangeli C, Pellegrini V, Beltram F, Giacca M: Cell membrane lipid rafts mediate caveolar endocytosis of HIV-1 Tat fusion proteins. J Biol Chem. 2003, 278 (36): 34141-34149. 10.1074/jbc.M303045200.

    Article  PubMed  CAS  Google Scholar 

  26. Richard JP, Melikov K, Vives E, Ramos C, Verbeure B, Gait MJ, Chernomordik LV, Lebleu B: Cell-penetrating peptides. A reevaluation of the mechanism of cellular uptake. J Biol Chem. 2003, 278 (1): 585-590. 10.1074/jbc.M209548200.

    Article  PubMed  CAS  Google Scholar 

  27. Tyagi M, Rusnati M, Presta M, Giacca M: Internalization of HIV-1 tat requires cell surface heparan sulfate proteoglycans. J Biol Chem. 2001, 276 (5): 3254-3261. 10.1074/jbc.M006701200.

    Article  PubMed  CAS  Google Scholar 

  28. Maccarana M, Casu B, Lindahl U: Minimal sequence in heparin/heparan sulfate required for binding of basic fibroblast growth factor. J Biol Chem. 1993, 268 (32): 23898-23905.

    PubMed  CAS  Google Scholar 

  29. Rusnati M, Tulipano G, Spillmann D, Tanghetti E, Oreste P, Zoppetti G, Giacca M, Presta M: Multiple interactions of HIV-I Tat protein with size-defined heparin oligosaccharides. J Biol Chem. 1999, 274 (40): 28198-28205. 10.1074/jbc.274.40.28198.

    Article  PubMed  CAS  Google Scholar 

  30. Spillmann D, Witt D, Lindahl U: Defining the interleukin-8-binding domain of heparan sulfate. J Biol Chem. 1998, 273 (25): 15487-15493. 10.1074/jbc.273.25.15487.

    Article  PubMed  CAS  Google Scholar 

  31. Nagahara H, Vocero-Akbani AM, Snyder EL, Ho A, Latham DG, Lissy NA, Becker-Hapak M, Ezhevsky SA, Dowdy SF: Transduction of full-length TAT fusion proteins into mammalian cells: TAT-p27Kip1 induces cell migration. Nat Med. 1998, 4 (12): 1449-1452. 10.1038/4042.

    Article  PubMed  CAS  Google Scholar 

  32. Asoh S, Ohsawa I, Mori T, Katsura K, Hiraide T, Katayama Y, Kimura M, Ozaki D, Yamagata K, Ohta S: Protection against ischemic brain injury by protein therapeutics. Proc Natl Acad Sci U S A. 2002, 99 (26): 17107-17112. 10.1073/pnas.262460299.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  33. Cao G, Pei W, Ge H, Liang Q, Luo Y, Sharp FR, Lu A, Ran R, Graham SH, Chen J: In Vivo Delivery of a Bcl-xL Fusion Protein Containing the TAT Protein Transduction Domain Protects against Ischemic Brain Injury and Neuronal Apoptosis. J Neurosci. 2002, 22 (13): 5423-5431.

    PubMed  CAS  Google Scholar 

  34. Orii KO, Grubb JH, Vogler C, Levy B, Tan Y, Markova K, Davidson BL, Mao Q, Orii T, Kondo N, Sly WS: Defining the pathway for Tat-mediated delivery of beta-glucuronidase in cultured cells and MPS VII mice. Mol Ther. 2005, 12 (2): 345-352. 10.1016/j.ymthe.2005.02.031.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  35. Schwarze SR, Ho A, Vocero-Akbani A, Dowdy SF: In vivo protein transduction: delivery of a biologically active protein into the mouse. Science. 1999, 285 (5433): 1569-1572. 10.1126/science.285.5433.1569.

    Article  PubMed  CAS  Google Scholar 

  36. Eguchi A, Akuta T, Okuyama H, Senda T, Yokoi H, Inokuchi H, Fujita S, Hayakawa T, Takeda K, Hasegawa M, Nakanishi M: Protein transduction domain of HIV-1 Tat protein promotes efficient delivery of DNA into mammalian cells. J Biol Chem. 2001, 276 (28): 26204-26210. 10.1074/jbc.M010625200.

    Article  PubMed  CAS  Google Scholar 

  37. Gratton JP, Yu J, Griffith JW, Babbitt RW, Scotland RS, Hickey R, Giordano FJ, Sessa WC: Cell-permeable peptides improve cellular uptake and therapeutic gene delivery of replication-deficient viruses in cells and in vivo. Nat Med. 2003, 9 (3): 357-362. 10.1038/nm835.

    Article  PubMed  CAS  Google Scholar 

  38. Ignatovich IA, Dizhe EB, Pavlotskaya AV, Akifiev BN, Burov SV, Orlov SV, Perevozchikov AP: Complexes of plasmid DNA with basic domain 47-57 of the HIV-1 Tat protein are transferred to mammalian cells by endocytosis-mediated pathways. J Biol Chem. 2003, 278 (43): 42625-42636. 10.1074/jbc.M301431200.

    Article  PubMed  CAS  Google Scholar 

  39. Lewin M, Carlesso N, Tung CH, Tang XW, Cory D, Scadden DT, Weissleder R: Tat peptide-derivatized magnetic nanoparticles allow in vivo tracking and recovery of progenitor cells. Nat Biotechnol. 2000, 18 (4): 410-414. 10.1038/74464.

    Article  PubMed  CAS  Google Scholar 

  40. Sandgren S, Cheng F, Belting M: Nuclear targeting of macromolecular polyanions by an HIV-Tat derived peptide. Role for cell-surface proteoglycans. J Biol Chem. 2002, 277 (41): 38877-38883. 10.1074/jbc.M205395200.

    Article  PubMed  CAS  Google Scholar 

  41. Torchilin VP, Rammohan R, Weissig V, Levchenko TS: TAT peptide on the surface of liposomes affords their efficient intracellular delivery even at low temperature and in the presence of metabolic inhibitors. Proc Natl Acad Sci U S A. 2001, 98 (15): 8786-8791. 10.1073/pnas.151247498.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  42. Kuhnel F, Schulte B, Wirth T, Woller N, Schafers S, Zender L, Manns M, Kubicka S: Protein transduction domains fused to virus receptors improve cellular virus uptake and enhance oncolysis by tumor-specific replicating vectors. J Virol. 2004, 78 (24): 13743-13754. 10.1128/JVI.78.24.13743-13754.2004.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Seki T, Dmitriev I, Suzuki K, Kashentseva E, Takayama K, Rots M, Uil T, Wu H, Wang M, Curiel DT: Fiber shaft extension in combination with HI loop ligands augments infectivity for CAR-negative tumor targets but does not enhance hepatotropism in vivo. Gene Ther. 2002, 9 (16): 1101-1108. 10.1038/sj.gt.3301815.

    Article  PubMed  CAS  Google Scholar 

  44. Van Houdt WJ, Wu H, Glasgow JN, Lamfers ML, Dirven CM, Gillespie GY, Curiel DT, Haviv YS: Gene delivery into malignant glioma by infectivity-enhanced adenovirus: in vivo versus in vitro models. Neuro Oncol. 2007, 9 (3): 280-290. 10.1215/15228517-2007-017.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  45. Albarran B, To R, Stayton PS: A TAT-streptavidin fusion protein directs uptake of biotinylated cargo into mammalian cells. Protein Eng Des Sel. 2005, 18 (3): 147-152. 10.1093/protein/gzi014.

    Article  PubMed  CAS  Google Scholar 

  46. Al-Taei S, Penning NA, Simpson JC, Futaki S, Takeuchi T, Nakase I, Jones AT: Intracellular traffic and fate of protein transduction domains HIV-1 TAT peptide and octaarginine. Implications for their utilization as drug delivery vectors. Bioconjug Chem. 2006, 17 (1): 90-100. 10.1021/bc050274h.

    Article  PubMed  CAS  Google Scholar 

  47. Loison F, Nizard P, Sourisseau T, Le Goff P, Debure L, Le Drean Y, Michel D: A ubiquitin-based assay for the cytosolic uptake of protein transduction domains. Mol Ther. 2005, 11 (2): 205-214. 10.1016/j.ymthe.2004.10.010.

    Article  PubMed  CAS  Google Scholar 

  48. Wadia JS, Stan RV, Dowdy SF: Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med. 2004, 10 (3): 310-315. 10.1038/nm996.

    Article  PubMed  CAS  Google Scholar 

  49. Blackhall FH, Merry CL, Davies EJ, Jayson GC: Heparan sulfate proteoglycans and cancer. Br J Cancer. 2001, 85 (8): 1094-1098. 10.1054/bjoc.2001.2054.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  50. Davies EJ, Blackhall FH, Shanks JH, David G, McGown AT, Swindell R, Slade RJ, Martin-Hirsch P, Gallagher JT, Jayson GC: Distribution and clinical significance of heparan sulfate proteoglycans in ovarian cancer. Clin Cancer Res. 2004, 10 (15): 5178-5186. 10.1158/1078-0432.CCR-03-0103.

    Article  PubMed  CAS  Google Scholar 

  51. Steck PA, Moser RP, Bruner JM, Liang L, Freidman AN, Hwang TL, Yung WK: Altered expression and distribution of heparan sulfate proteoglycans in human gliomas. Cancer Res. 1989, 49 (8): 2096-2103.

    PubMed  CAS  Google Scholar 

  52. Kanerva A, Mikheeva GV, Krasnykh V, Coolidge CJ, Lam JT, Mahasreshti PJ, Barker SD, Straughn M, Barnes MN, Alvarez RD, Hemminki A, Curiel DT: Targeting adenovirus to the serotype 3 receptor increases gene transfer efficiency to ovarian cancer cells. Clin Cancer Res. 2002, 8 (1): 275-280.

    PubMed  CAS  Google Scholar 

  53. Wu H, Dmitriev I, Kashentseva E, Seki T, Wang M, Curiel DT: Construction and characterization of adenovirus serotype 5 packaged by serotype 3 hexon. J Virol. 2002, 76 (24): 12775-12782. 10.1128/JVI.76.24.12775-12782.2002.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  54. Glasgow JN, Kremer EJ, Hemminki A, Siegal GP, Douglas JT, Curiel DT: An adenovirus vector with a chimeric fiber derived from canine adenovirus type 2 displays novel tropism. Virology. 2004, 324 (1): 103-116. 10.1016/j.virol.2004.03.028.

    Article  PubMed  CAS  Google Scholar 

  55. Le LP, Le HN, Dmitriev IP, Davydova JG, Gavrikova T, Yamamoto S, Curiel DT, Yamamoto M: Dynamic monitoring of oncolytic adenovirus in vivo by genetic capsid labeling. J Natl Cancer Inst. 2006, 98 (3): 203-214.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank Dr. Joel N. Glasgow for providing recombinant knob protein and Minghui Wang for assistance in quantitative PCR analysis. This work was supported by the NIH brain SPORE grant P50 CA097247 and the Juvenile Diabetes Research Foundation grants 1-2005-71 and 5-2007-660.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hongju Wu.

Additional information

Competing interests

The author(s) declare that they have no competing interests.

Authors' contributions

TH participated in the generation and in vitro characterization of the adenoviral vectors. YT carried out in vitro and in vivo gene transfer assays. HU performed immunohistochemistry studies. LEP participated in cell culture and tumor model establishment. GPS helped in immunohistochemical studies and in the preparation of the manuscript. JLC assisted in the design of the study and manuscript preparation. HW conceived of the study, participated in its design and coordination, and drafted the manuscript. All authors read and approved the final manuscript.

Tie Han, Yizhe Tang contributed equally to this work.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Han, T., Tang, Y., Ugai, H. et al. Genetic incorporation of the protein transduction domain of Tat into Ad5 fiber enhances gene transfer efficacy. Virol J 4, 103 (2007). https://doi.org/10.1186/1743-422X-4-103

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1743-422X-4-103

Keywords