key: cord-0719457-hnaw4pyf authors: Liu, Qiang; Zhou, Shuya; Fan, Changfa; Huang, Weijin; Li, Qianqian; Liu, Susu; Wu, Xi; Li, Baowen; Wang, Youchun title: Biodistribution and residence time of adenovector serotype 5 in normal and immunodeficient mice and rats detected with bioluminescent imaging date: 2017-06-15 journal: Sci Rep DOI: 10.1038/s41598-017-03852-0 sha: a30cef8b3428aa804aa2cbf0ec0303827bb09514 doc_id: 719457 cord_uid: hnaw4pyf As concerns increase about adenovirus type 5 (Ad5) being a safe gene transfer vector, it is important to evaluate its distribution, residence time, and possible toxicity in immunodeficient populations. To characterize the potential risk associated with different Ad5 vector delivery modes, we used immunocompetent and immunodeficient Rag2 (−/−) animals to establish mouse and rat models that could be monitored with bioluminescent imaging following intramuscular or intravascular infection with an engineered replication-incompetent Ad5 virus carrying the firefly luciferase gene (Ad5-Fluc). The Ad5 vector was less well-tolerated by Rag2 (−/−) animals than by wildtype ones, with delayed residence time, wider virus dissemination, less weight gain, and relatively severe pathological changes. In intravascularly Ad5-Fluc-infected Rag2 (−/−) mice, systemic virus dissemination extended from the abdomen to the limbs and head on day 9 post-infection. Additionally, significant increases in plasma TNF-α and IFN-γ, which may be important factors in the heightened immunopathology in the liver and brain, were detected in the Rag2 (−/−) mice 30 days after intravascular delivery. The Ad5 vector was better tolerated after intramuscular delivery than after intravascular delivery. Ad5-Fluc/Rag2 (−/−) mice and rats can be used as reliable models of an immunodeficient population in which to evaluate the safety of Ad5-vectored vaccines or gene therapy products. associated with different modes of Adv5 delivery, especially in immunodeficient hosts, and to identify the mechanisms underlying the resulting immunopathology. Therefore, we monitored the biodistribution and metabolism of replication-incompetent Ad5 expressing firefly luciferase (Ad5-Fluc) in real time in living mice and rats with or without immunodeficiency. The immunodeficient Rag2 −/− rats were generated previously to resemble immunodeficient human populations 9 . Here, both Rag2 −/− mice and Rag2 −/− rats were used for Ad5 vector safety evaluation. Like Rag2 −/− rats, Rag2 −/− mice are defective in the recombination machinery required for the development of both B and T cells, but these animals can produce NK cells, and no other innate immune cells are affected by the Rag2 −/− mutation 11 . The immunodeficient Rag2 −/− mice were more sensitive to Ad5 viral infection than the immunocompetent mice; when luminescent signals were detected with the BLI method, brain tissue sections showed pathological findings only in the Rag2 −/− mice. Additionally, a tumour necrosis factor α (TNF-α)-and interferon γ (IFN-γ)-related cytokine storm occurred in the Rag2 −/− mice that were infected intravascularly with Adv5, and the level of the cytokine storm correlated with the pathological changes in the liver and brain. To test and verify the T and B cell development in homozygous Rag2-knockout mice, we used flow cytometry to identify various white blood cell populations in the peripheral blood. We detected significantly reduced numbers of T helper cells (CD3 + /CD4 + ) in the Rag2 −/− mice (0.18%) compared with the wildtype mice. The development of cytotoxic T cells (CD3 + /CD8 + ) was also blocked in the Rag2 −/− mice (0.09%). Additionally, we failed to detect CD3 − /CD19 + B cells in the peripheral blood (0.35%) of the Rag2 −/− mice. These results indicate that no mature T or B cells were produced after the Rag2 gene was knocked out. However, we observed that the number of CD3 − NK1.1 + natural killer (NK) cells increased in the Rag2 −/− mice (77.3%) compared with the wildtype mice (11.1%) ( Supplementary Fig. S1 ). This increase in NK cells may be a compensation mechanism to strengthen the immune system when mature T and B cells are abolished by a Rag2 gene knockout. Adv5 is commonly administered as a vaccine vector by the intramuscular (IM) route and as gene delivery vector in gene therapy by the intravascular (IV) route. To document the distribution and kinetics of Adv5 in both immunocompetent C57BL/6 and immunodeficient Rag2 −/− mouse models, the animals were inoculated with 5 × 10 10 plaque-forming units (pfu)/kg Adv5 by the IM or IV route. Because Ad5-Fluc expresses the bioluminescent luciferase protein, BLI was used to monitor the progress, sites, and degree of Adv5 expression in real time, based on the detected luciferase bioluminescence. In vivo luminescent signals were visible at the inoculation sites of both IM and IV injections and in the abdomen as early as 6 h after administration (Fig. 1a,b, left) , suggesting that the bioluminescent signals generated by Ad5-Fluc are strong enough to show the distribution of Adv5, even in the very early stage of its dissemination. By day 9, the peak luminescence intensity and greatest systemic dissemination were observed, extending from the abdomen to the limbs and head in the Rag2 −/− mice infected via the IV route. However, we observed a rapid damping of luciferase expression in the wildtype C57BL/6 mice, and Adv5 was almost cleared by day 20 post-inoculation, regardless of its route of delivery (IM or IV). Interestingly, the luciferase expression from Adv5-Fluc that was injected IM persisted and increased continuously in the Rag2 −/− mice until the end of the experiment (day 35), but no systemic dissemination was observed (Fig. 1a, left) . Without clearance of the Adv5-infected cells by T cells and B cells, the luciferase signal was significantly stronger and more persistent in the Rag2 −/− mice, regardless of the inoculation route (P < 0.01; Fig. 1a ,b, right). These data indicate that T cells and B cells are essential for the clearance of Adv5 administered by either the IM or IV route. To determine the bioluminescence distributions in vivo, various organs or tissues from the Adv5-inoculated mice were dissected and analysed after the perfusion of luciferin on day 1 post-inoculation. In the IM inoculation group, bioluminescent signals were detected in the tissues around the inoculation site, including the lymph node, muscle, and skin, and in the liver, reflecting the dissemination kinetics from the muscle to the liver (Fig. 1c) . Judging from the live imaging, the biodistribution of IV-inoculated Adv5 was wider in the Rag2 −/− mice than in the C57BL/6 mice, especially in the limbs and mouth. However, the bioluminescent signals did not differ in any of the dissected organs other than the limbs and mouth, and luminescence was detected in the liver, spleen, and lung (Fig. 1d ). This indicates that Adv5 inoculated into the blood by IV administration accumulates rapidly in the liver and then spreads to other organs. Bioluminescent imaging of Adv5 infection in Rag2 −/− and SD rats. Rag2 −/− rats, generated by applying the transcription activator-like effector nucleases (TALENs) technology, were observed to have no mature T or B cells; however, these animals had more NK cells than SD rats ( Supplementary Fig. S2 ). After the administration of 5 × 10 10 pfu/kg Adv5 via the IM or IV route, the initial local and systemic dissemination of Adv5 in rats were similar to those in mice. The Rag2 −/− rats were more susceptible to Adv5 than the immunocompetent SD rats, displaying stronger luminescent signals and longer residence times (Fig. 2a,b , left). There were clear differences between the IV and IM delivery routes in terms of the biodistribution and residence time. After infection by the IM route, the Adv5 distribution was wider in the Rag2 −/− rats than in the SD rats. However, the Adv5 distribution patterns were similar in the two rat strains after Adv5 administration by the IV route. Notably, the luciferase signal was significantly stronger and more persistent in the Rag2 −/− rats infected by IV route than that in SD rats (P < 0.05; Fig. 2b, right) . Notably, only small proportions of the Rag2 −/− and SD rats infected by the IV route displayed infected spleens, but all of the rats displayed infected livers (Fig. 2c ). Upregulated pro-inflammatory cytokine responses in Adv5-inoculated Rag2 −/− mice. Vaccine antigen clearance is predominantly dependent on the adaptive immune responses. Therefore, the striking difference between the Adv5 profiles of the wildtype and immunodeficient mice might be related to the magnitudes of their cellular immune responses. The cytokine dysregulation increases the risks associated with Adv5 inoculation in immunodeficient mice. We examined the magnitude of the cellular response induced in the immunodeficient Rag2 −/− mice and the wildtype mice. Notably, the Rag2 −/− mice showed no clearance of the Adv5-infected cells in vivo, at least at 35 days after IM or IV injection. Many pro-inflammatory cytokines in the blood, including the inflammatory cytokines interleukin (IL)-12p70, TNF-α, IFN-γ, IL-6, IL-10, granulocyte/macrophage colony-stimulating factor (GM-CSF), MIP-1α, MIP-1β, IL-1β, and chemokine monocyte chemoattractant protein 1 (MCP-1), were measured 1 day before and 30 days after inoculation. The cytokine levels showed little change in the C57BL/6 mice after Adv5 inoculation. Although the level of MIP-1α expression by C57BL/6 mice trended lower at 30 days post-inoculation, this difference did not reach statistical significance. Interestingly, the Rag2 −/− mice had significantly lower expression levels of MIP-1α and MIP-1β than the C57BL/6 mice (P < 0.05; Fig. 3h ,i), possibly due to the lack of mature T and B lymphocytes in these mice. As expected, the levels of TNF-α and IFN-γ were significantly higher in the post-Ad5-Fluc-inoculated Rag2 −/− mice than in the pre-Ad5-Fluc-inoculated Rag2 −/− mice (P < 0.05; Fig. 3b ,c). However, although the MCP-1 levels tended to be higher in these mice after Adv5 inoculation, this difference did not reach statistical significance. When we compared the effects of IV and IM administration, although the levels of TNF-α and IFN-γ trend higher in the IV group than in the IM group, especially in the Rag2 −/− mice, these differences are not statistically significant. Combined with the compensatory NK cell increase in the Rag2 −/− mice that was detected with flow cytometry, these data suggest that the innate immune response was increased and prolonged in the Rag2 −/− mice. . Bioluminescent images of a representative mouse from each group are shown at 6 h, 1 day, 3 days, 6 days, 9 days, 20 days, and 35 days post-infection. Relative bioluminescence intensity is shown in pseudo-colour, with red and blue representing the strongest and weakest photon fluxes, respectively. Mean (±standard error of the mean) values for total flux at different time points are shown. The differences between subgroups were calculated with a paired t-test. (c,d) Rag2 −/− and C57BL/6 mice inoculated with Ad5-Fluc by the IM route (c) or IV route (d) were imaged on day 1 post-inoculation. Different organs were dissected and tested for Fluc transgene expression with BLI: (1) thymus, (2) heart, (3) liver, (4) spleen, (5) kidney, (6) lung, (7) lymph node, (8) muscle, (9) skin, (10) ovary/testis, (11) brain, and (12) intestine. Scientific RepoRts | 7: 3597 | DOI:10.1038/s41598-017-03852-0 Adv5 inoculation caused pathological changes in the brains of Rag2 −/− , but not of C57BL/6 mice. To further investigate the potential risk of Adv5 inoculation in C57BL/6 and Rag2 −/− mice, their body weights after Adv5 administration were monitored, and any pathological changes were analysed. No significant differences were detected between the body weight gains in the IV-infected, IM-infected, and naïve C57BL/6 mice (P = 0.12; Fig. 4a ). However, the body weight gain in the IV-infected Rag2 −/− mice was significantly lower than those in the other groups (P < 0.05; Fig. 4b) . A systemic histopathological examination was performed in many tissues, including the brain, heart, liver, spleen, lung, kidney, intestine, thymus, muscle, and testis/ovary. No pathological changes were observed in the naïve C57BL/6 mice (Fig. 4c) or naïve Rag2 −/− mice (Fig. 4d ) or in the immunocompetent or immunodeficient mice inoculated with Adv5 via the IM route (Fig. 4e,f) . Therefore, the IM route is relatively safe for use in the inoculation of Adv5-based vaccines. However, the IV-inoculated groups displayed inflammatory signs in the liver (Fig. 4g,h, upper) . Moreover, glial nodules formed and oligodendrocytes increased in the brains of the immunodeficient Rag2 −/− mice that were inoculated with Adv5 via the IV route. In contrast, these changes were not found in the brains of C57BL/6 mice infected via the IV route (Fig. 4g,h, lower) . ALT is a cytosolic enzyme that mainly exists in the liver, while AST is primarily present in mitochondria and cytoplasm in the liver. Once hepatocytes are damaged, ALT and AST will leak into circulation and the levels of these enzymes will increase in serum. IV administration of Adv5 to the Rag2 −/− mice increased serum alanine aminotransferase (ALT, Fig. 4i ) and aspartate aminotransferase (AST, Fig. 4j ) activities compared to that in control mice (P < 0.05). But there were no significant differences within the IM inoculated C57BL/6, Rag2 −/− and naïve mice (Fig. 4i,j) . Here, we show that BLI is a valuable tool with which to visualize the distribution of Adv5 and the clearance of Adv5-infected cells after the virus administered by the IM or IV route to living mice and rats with or without immunodeficiency. This is a significant advance toward the development of Adv5-based therapies and vaccine improvement, and it extends our understanding of Adv5 immunopathology. In the context of preclinical drug safety evaluation, rats are believed to have a higher similarity to humans, providing predictable useful experimental data, and pharmaceutical companies use rats for a large proportion of their mandatory toxicity testing 12 . Rag2 −/− rats, generated with the TALENs technology, were previously used to evaluate the toxicity and biodistribution of Adv5 in an immunodeficient population 9 . In this study, we found that Adv5 was more widely disseminated in the Rag2-deficient mice than in the Rag2-deficient rats, which suggests that these two rodent species Rag2 −/− mice inoculated via IV (h), was performed systematically on day 35 after inoculation with 5 × 10 10 pfu/ kg Ad5-Fluc. Paraffin-embedded tissue sections were stained with haematoxylin and eosin, and arrows indicate the lesion sites. Scale bar, 20 μm. Effects on the activities of serum (i) alanine aminotransferase (ALT) and (j) aspartate aminotransferase (AST). *Indicates that there is a significant (P < 0.05) difference between the treatment group and the control group. differently predict the outcome in humans. However, in both species, the distributions and residence times in immunodeficient animals were different from those in immunocompetent ones, suggesting that Adv5 should be used with caution in various groups of immunosuppressed patients because considerably more serious adverse effects can be expected in these patients. Consistent with this, adenovirus is ubiquitous and causes generally mild, self-limiting infections in immunocompetent adults, but it can lead to a serious, often life-threatening illness in solid-organ transplant recipients and AIDS patients 13, 14 . It was shown that after its IV administration, the Adv5 half-life was 2-5 min in normal mice and that more than 99% of Adv5 was cleared from the blood within 1 h. Most of the virus accumulated in the liver and was eliminated by Kupffer cells within 24 h 15 . In this study, the peak level of transgene expression was significantly higher and the kinetics of viral clearance was significantly slower in the T and B cell-deficient mice than in the wildtype mice after infection via either the IV or IM route. Therefore, we speculate that the T and B cells play important roles in the clearance of the Ad5 virus. Until now, the specific role of B cells during primary infection has been somewhat controversial. Although mice lacking both CD8 + T cells and B cells died after influenza infection, mice lacking CD4 + T cells and B cells survived 16 . However, Lee et al. showed that mice lacking B cells succumbed to influenza H1N1 (PR8) infection despite the infiltration of a larger number of CD8 + T cells 17 . Conventional assays of host-pathogen interactions involving replication-competent and replication-defective human adenoviruses have been performed with PCR, Southern blotting, and immunohistochemical methods; however, these required large numbers of animals to be euthanized to obtain statistically meaningful data at multiple time points 18 . Here, to simplify this process of viral vector evaluation, we used BLI to monitor the dissemination of Adv5 and dynamically localize it in unexpected anatomical sites in the same animals by measuring the firefly luciferase expressed by Ad5-Fluc. We and others have also shown a significant correlation between photoemission and viral load in vivo and have demonstrated the many advantages of BLI, including the ability to image each animal repeatedly, which is both ethically and economically advantageous 19, 20 . We found that Adv5 spread to the liver within 1 day of its IM administration. As reported previously, when Adv5 is delivered into muscle, it may infect the antigen-presenting cells that reside there, which then traffic it to the liver 21 . However, Adv5 can escape from the perivascular space into the systemic circulation via the vasa vasorum or through the lymphatics after its IM injection 22 . In contrast, after its IV administration, Adv5 rapidly binds to circulating platelets, causing their activation/aggregation and subsequent entrapment in the liver sinusoids. These virus-platelet aggregates are taken up by Kupffer cells and degraded 23 . We anticipate the future development of imaging strategies that detect not only the pathogen but also the cellular and tissue-wide reactions of the host. The study by Kadurugamuwa et al. that used multicolour bioluminescence to track bacterial spread and identify host neuronal injury was a key step towards the goal of monitoring both the host and the pathogen 24 . Current evidence suggests that when Adv5 is used in gene therapy or vaccination, the innate immune responses are responsible for its dose-limiting toxicity and that the adaptive immune responses, which eliminate the infected cells, limit the duration of transgene expression 25 . We found that the Rag2 −/− mice displayed significantly higher levels of the cytokines TNF-α and IFN-γ after Adv5 inoculation than the immunocompetent mice. Positive correlations between viral immunopathology and TNF-α or IFN-γ levels have also been reported in clinical cases and animal models of severe acute respiratory syndrome (SARS) and H5N1 infection 26, 27 . This release or synthesis of TNF, in turn, plays a major causative role in the pathological changes associated with IV adenovirus infusion as well as a role in the induction of the antiviral immune response 28, 29 . The IFN-γ produced by T and NK cells plays an important role in cell-mediated immunity against a broad spectrum of intracellular pathogens, and it also activates antigen presentation and primes TNF-α release by macrophages 30 . The inflammation associated with a cytokine storm has been shown to begin at a local site and to spread throughout the body via the systemic circulation 31 . Therefore, we speculate that the brain pathology in the immunodeficient mice was not a direct toxic effect of adenovector delivery, but more likely a secondary effect of inflammation and the immune response to the adenovector, because TNF-α and IFN-γ are considered the key cytokines that drive pro-inflammatory activity 32 . Notably, hepatic injury was detected in both the Rag2 −/− and wildtype C57BL/6 mice. Muruve showed that neutrophils acutely infiltrate the liver 1-6 h after the injection of Adv5. Immune effector cells are recruited to the sites of injury in response to chemokines, such as MCP-1, MIP-1α, MIP-1β, and RANTES. These signals directly correlate with the infiltration of inflammatory cells and the degree of hepatic injury 33 . Similar results were reported by Lieber, who reported a biphasic increase in serum TNF, with the first peak contributed by macrophages shortly after adenovirus administration and the second major peak contributed by lymphocytes after several days 34 . However, MIP-1α and MIP-1β decreased significantly after Adv5 infection in the Rag2 −/− mice. MIP-1α and MIP-1β are produced by macrophages and activate human granulocytes, including neutrophils, eosinophils, and basophils, which may lead to acute neutrophilic inflammation 35 . Further research is required to determine whether or not the high-level expression of cytokines such as TNF-α and IFN-γ and the low levels of MIP-1α and MIP-1β in Adv5-infected mice are the causes or effects of the observed pathological changes. Moreover, the dynamics of cytokine expression during infection in vivo should also be investigated to detect any transient changes in cytokine levels. It is a pity that the data on cytokine response and histopathology were not conducted in rats, we believe that the conclusions will be more robust complementing with additional data in rats, and we will pay attention to this problem in the future study. The current goal of vaccinologists and gene therapists is to create an Adv5 that avoids being detected by the immune system and has no deleterious side effects. We believe that the focus should be on two types of approaches. The first is dosage control. Some findings indicate that the hepatotropism of Adv5 is dose-limiting, and a certain threshold dose of injected Adv5 can saturate the capacity of Kupffer cells for adenovirus uptake, leading to the systemic dissemination of Adv5 particles. This then results in acute hepatocellular damage and the potential infection of other cell types that secrete pro-inflammatory cytokines, thus exacerbating the systemic toxicity of the vector 36 . The second approach involves modifying the method or route of delivery, using for instance, hydrodynamic injection, ultrasound-guided intramyocardial gene delivery, or other techniques to locally deliver low vector doses directly into the target organs, reducing the systemic dissemination of the vector, the inflammatory responses, and the immunotoxicity 37 . Cytokine flare, reduced body weight gain, and pathological damage suggest that there is a possible risk involved in the use of Adv5 in gene therapy via the IV route, especially in immunodeficient populations. Although useful in preclinical studies, in general, the predictions from rodent studies have limited applicability to human outcomes. A single animal model can never recapitulate all of the aspects of human viral-vector-based gene therapies and vaccines because each model has its own advantages and disadvantages. Adenovirus-vectored vaccines have been constructed based on various different systems. In this study, recombinant Adv5 was generated using a Cre/loxP-based system 38 , which has been used in several vaccines, including an HIV vaccine 39 , and in gene therapy 40 . Notably, the promoter and overall design of the Ad5 expression cassette has a major impact on Adv5 toxicity and distribution in vivo. Therefore, the safety of recombinant Ad5 products generated with other construction systems must be evaluated in future studies. Additionally, the relative data should be obtained from older animals because gene therapy is predominantly administered to adults with degenerated tissues/cells. In summary, in this study, we tested the residence time, biodistribution, and possible toxicity of Adv5 in both immunodeficient and immunocompetent mice and rats, with doses equivalent to the higher end of a reasonable range in humans. We found that the ideal route of administration was IM, by which even immunodeficient animals were successfully and safely treated with a high dose of the vaccine. Adverse effects were observed when the animals were treated by the IV route, which has been used in previous gene therapy applications. Therefore, in accord with previous reports, we believe that Ad5 vectors are generally safe for vaccine and gene therapy applications. However, future research should focus on any adverse effects in recipients with congenital immunodeficient backgrounds or virus-induced immunodeficiency. Finally, visualizing Ad5-Fluc infection in immunocompetent and immunodeficient mice and rats is an additional or alternative way to study adenoviral infection because it is convenient, sensitive, and requires the use of fewer animals. Construction of Ad5-Fluc virus. Using the firefly luciferase (Fluc) gene derived from pLUCF (kindly provided by John T. Schiller, National Cancer Institute, Bethesda, MD, USA), a replication-incompetent Ad5-Fluc was constructed by deleting the E1 and E3 viral genes using the Cre/loxP-based system 38 . Briefly, PCR was used to construct a recombinant shuttle plasmid, pDC316-Fluc. HEK293 cells were co-transfected with an expression plasmid containing structural genes (pBHGlox-E1,3Cre) and the reporter plasmid for Ad5-Fluc construction using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA). The recombinant adenovirus containing the Fluc gene (Ad5-Fluc) was acquired by Cre-lox recombination in HEK293 cells. Animal experiments. The mice and rats used in this study were housed and handled strictly in accordance with the guidelines set by the Association for the Assessment and Accreditation of Laboratory Animal Care (AAALAC; Frederick, MD, USA). The study protocol was approved by the Animal Care and Use Committee of the National Institute for Food and Drug Control (NIFDC; Beijing, China). Four-week-old Rag2 −/− mice and Rag2 −/− rats, wildtype C57BL/6 mice, and SD rats were obtained from the Institute for Laboratory Animal Resources, NIFDC. Eight groups each of mice (ten per group) and rats (four per group) received a single dose of Ad5-Fluc at 5 × 10 10 pfu/kg (suspended in 100 μl of PBS) by IM inoculation or 5 × 10 10 pfu/kg (suspended in 200 μl of PBS) by IV (tail vein) injection. This dose is equivalent to the higher end of a reasonable range in humans 41, 42 . The mice and rats were examined with BLI at 6 h, 1 day, 3 days, 6 days, 9 days, 20 days, and 35 days post-infection. Ten additional Rag2 −/− and normal animals mice were used as negative controls. To test the immunopathology of Ad5-Fluc in mice, their body weights were monitored and recorded. The mice and rats were humanely euthanized with isoflurane to analyse the biodistribution of Ad5-Fluc in their tissues on day 1 post-infection. BLI analysis. Bioluminescent imaging was performed with the IVIS Lumina Series III Imaging System (PerkinElmer, Baltimore, MD, USA), as described previously 43 . In brief, the mice and rats were anesthetized with successive intraperitoneal injections of pelltobarbitalum natricum (240 mg/kg body weight) and the substrate d-luciferin (50 mg/kg body weight; Xenogen-Caliper Corp., Alameda, CA, USA). The mice and rats were imaged 10 min later. To image individual tissues, mice and rats were dissected rapidly, and the bioluminescence was quantified by using the IVIS system to analyse the tissues ex vivo within 3 min of dissection. Luminescence was acquired with an exposure time of 60 s, and regions of interest (ROIs) were analysed with the Living Image software (Caliper Life Sciences, Baltimore, MD, USA). The relative intensities of the emitted light are presented as pseudo-coloured images, with colours ranging from red (most intense) to blue (least intense). The signals emitted from different ROIs in the body were measured and are presented as the total flux in photons/s. All data are presented as mean values ± SEM. IL-1β, and MCP-1, were quantified with the Cytometric Bead Array (BD Biosciences) on a FACSCalibur cytometer (BD Biosciences); this method has been used previously to analyze the immunopathology of different viral infections in humans 44-46 . Individual cytokine concentrations (pg/ml) were computed with standard reference curves in the CellQuest ™ (BD Biosciences) and FCAP Array software, according to the manufacturer's protocols. The inflammatory cytokines were measured simultaneously with different capture beads. Each capture bead in the array had a unique fluorescence and was coated with a capture antibody specific for a single analyte. A combination of different beads was mixed with a sample or standard with a mixture of detection antibodies Tissues, including the heart, lung, liver, spleen, kidney, intestine, thymus, skin, muscle, and brain, were dissected from the mice on day 35 post-infection, fixed in 10% formalin, and embedded in paraffin. Paraffin sections (2 μm thick) were stained with haematoxylin and eosin, and pathological changes were observed under a light microscope. Plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities were measured using an enzyme-linked immune sorbent assay An inventory of shedding data from clinical gene therapy trials Intravascular adenoviral agents in cancer patients: lessons from clinical trials Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine Adenovirus-based vaccines for fighting infectious diseases and cancer: progress in the field Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer Vaccination coverage in immunosuppressed patients: results of a regional health services research study Pandemic potential of a strain of influenza A (H1N1): early findings Infections in the immunosuppressed host Bioluminescent imaging of vaccinia virus infection in immunocompetent and immunodeficient rats as a model for human smallpox Visualizing influenza virus infection in living mice RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement Laboratory animals: the Renaissance rat Ganciclovir inhibits human adenovirus replication and pathogenicity in permissive immunosuppressed Syrian hamsters Hexadecyloxypropyl-cidofovir, CMX001, prevents adenovirus-induced mortality in a permissive, immunosuppressed animal model Blood clearance rates of adenovirus type 5 in mice Th cell-deficient mice control influenza virus infection more effectively than Th-and B cell-deficient mice: evidence for a Th-independent contribution by B cells to virus clearance CD4 T cell-independent antibody response promotes resolution of primary influenza infection and helps to prevent reinfection Analysis of Adenovirus Sequestration in the Liver, Transduction of Hepatic Cells, and Innate Toxicity after Injection of Fiber-Modified Vectors A safe and sensitive enterovirus A71 infection model based on human SCARB2 knock-in mice Luciferase imaging for evaluation of oncolytic adenovirus replication in vivo Adenoviral vectors persist in vivo and maintain activated CD8+ T cells: implications for their use as vaccines Biodistribution of adenoviral vector to nontarget tissues after local in vivo gene transfer to arterial wall using intravascular and periadventitial gene delivery methods Adenovirus-platelet interaction in blood causes virus sequestration to the reticuloendothelial system of the liver Reduction of astrogliosis by early treatment of pneumococcal meningitis measured by simultaneous imaging, in vivo, of the pathogen and host response Implications of the innate immune response to adenovirus and adenoviral vectors Interferon-mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia Analysis of adenovirus sequestration in the liver, transduction of hepatic cells, and innate toxicity after injection of fiber-modified vectors Toxicity of a first-generation adenoviral vector in rhesus macaques Acute hepatotoxicity of oncolytic adenoviruses in mouse models is associated with expression of wild-type E1a and induction of TNF-alpha Incidence and outcomes of acute lung injury Cutting edge: pulmonary immunopathology mediated by antigen-specific expression of TNF-alpha by antiviral CD8+ T cells Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo The role of Kupffer cell activation and viral gene expression in early liver toxicity after infusion of recombinant adenovirus vectors Multiplex bead analysis of vitreous and serum concentrations of inflammatory and proangiogenic factors in diabetic patients The role of capsid-endothelial interactions in the innate immune response to adenovirus vectors Pseudo-hydrodynamic delivery of helper-dependent adenoviral vectors into non-human primates for liverdirected gene therapy A high-efficiency Cre/loxP-based system for construction of adenoviral vectors Potent specific immune responses induced by prime-boost-boost strategies based on DNA, adenovirus, and Sendai virus vectors expressing gag gene of Chinese HIV-1 subtype B Targeted gene therapy toward astrocytoma using a Cre/loxP-based adenovirus system Safety and Immunogenicity of the MRKAd5 gag HIV Type 1 Vaccine in a Worldwide Phase 1 Study of Healthy Adults Phase I trial of adenovirus-mediated p53 gene therapy for recurrent glioma: biological and clinical results Application of bioluminescence imaging to the prediction of lethality in vaccinia virus-infected mice CD40 ligand enhances dengue viral infection of dendritic cells: a possible mechanism for T cell-mediated immunopathology Gamma interferon plays a crucial early antiviral role in protection against West Nile virus infection Measles virus infection in adults induces production of IL-10 and is associated with increased CD4+ CD25+ regulatory T cells We are grateful to Dr John T. Schiller from the National Cancer Institute, Bethesda, MA, USA, for providing the pLUCF plasmid and to Dr Yanqiu Li from PerkinElmer Inc. for providing the IVIS Lumina Series III machine and technical assistance. We thank Yanxian Yu and Xuebo Shi from BD Biosciences for help with the flow cytometric analysis. We also thank Dr Katie Oakley for language editing. This study was supported by grants from the National Science and Technology Major Project (2012ZX10004701-001) and the National Science and Technology Support Project (2014BAI02B01). The funding body had no role in the study design, data collection or analysis, decision to publish, or preparation of the manuscript. Statistical analysis. All graphs were generated with Prism 5.0 software (GraphPad, San Diego, CA, USA).The differences in the various cytokine levels in the pre-and post-infection subgroups and the differences in the bioluminescent intensities in the Rag2 −/− and normal subgroups were calculated with paired t-tests. P values < 0.05 were considered statistically significant. Supplementary information accompanies this paper at doi:10.1038/s41598-017-03852-0Competing Interests: The authors declare that they have no competing interests.Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.