AAV2-mediated transfer of the human aquaporin-1 cDNA restores fluid secretion from irradiated miniature pig parotid glands

R Gao, X Yan, C Zheng, C M Goldsmith, S Afione, B Hai, J Xu, J Zhou, C Zhang, J A Chiorini, B J Baum, S Wang, R Gao, X Yan, C Zheng, C M Goldsmith, S Afione, B Hai, J Xu, J Zhou, C Zhang, J A Chiorini, B J Baum, S Wang

Abstract

Previously (Shan et al, 2005), we reported that adenoviral vector-mediated transfer of the human aquaporin-1 (hAQP1) cDNA to minipig parotid glands following irradiation (IR) transiently restored salivary flow to near normal levels. This study evaluated a serotype 2, adeno-associated viral (AAV2) vector for extended correction of IR (single dose; 20 Gy)-induced, parotid salivary hypofunction in minipigs. At 16 weeks following the IR parotid salivary flow decreased by 85-90%. AAV2hAQP1 administration at week 17 transduced only duct cells and resulted in a dose-dependent increase in salivary flow to approximately 35% of pre-IR levels (to approximately 1 ml per 10 min) after 8 weeks (peak response). Administration of a control AAV2 vector or saline was without effect. Little change was observed in clinical chemistry and hematology values after AAV2hAQP1 delivery. Vector-treated animals generated high anti-AAV2 neutralizing antibody titers by week 4 (approximately 1:1600) and significant elevations in salivary (approximately 15%), but not serum, granulocyte macrophage colony-stimulating factor levels. Following vector administration, salivary [Na(+)] was dramatically increased, from approximately 10 to approximately 55 mM (at 4 weeks) and finally to 39 mM (8 weeks). The findings demonstrate that localized delivery of AAV2hAQP1 to IR-damaged parotid glands leads to increased fluid secretion from surviving duct cells, and may be useful in providing extended relief of salivary hypofunction in previously irradiated patients.

Conflict of interest statement

The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
Effect of irradiation on parotid saliva flow rate in minipigs. One parotid gland of animals (n=9) was irradiated with 20 Gy, as described in the Materials and Methods. Parotid saliva flow rates were thereafter measured at weeks 4 and 16. Data shown are the mean±SD of salivary flow rates for the targeted glands (open bars; note that for the 16 week value the SD is too small to be seen) and the contralateral glands (black bars). Data were analyzed using a one-way analysis of variance (F=72.69, p

Figure 2

Effect on saliva flow rates…

Figure 2

Effect on saliva flow rates of AAV2hAQP1 administration to minipig parotid glands after…

Figure 2
Effect on saliva flow rates of AAV2hAQP1 administration to minipig parotid glands after irradiation. At week 17 following 20 Gy irradiation, either AAV2hAQP1 (A, B), AAV2LacZ (B) or saline buffer (B) were administered to a single parotid gland. Panel A shows the effect of different doses of AAV2hAQP1 on parotid saliva flow rates in irradiated glands (n=2/group; data are mean values ±SD obtained 12 weeks following vector administration; note that for the 3×1011 vp dose the SD is too small to be seen). The data in Panel A was not analyzed for statistical significance, because there were only two animals/group. Panel B shows salivary flow rates at different times following administration of AAV2hAQP1, AAV2LacZ or saline buffer to irradiated parotid glands (n=3/group; data are mean values ±SD). Time zero corresponds to week 17 following irradiation. Note that for the 8-week time-point with buffer and AAV2LacZ treatment the SDs are too small to be seen. Data were analyzed using a one-way analysis of variance (F=27.88, p<0.001), followed by the Tukey test. All of the parotid flow rates for the AAV2hAQP1-treated minipigs are significantly different from those of the buffer- and AAV2LacZ-treated animals at weeks 4 (p<0.01 vs buffer; p<0.05 vs AAV2LacZ), 6 (p<0.001) and 8 (p<0.001). There were no significant differences in parotid flow rates between the buffer- and AAV2LacZ-treated animals at every time-point.

Figure 3

AAV2-mediated transgene expression in minipig…

Figure 3

AAV2-mediated transgene expression in minipig parotid glands. Representative results of Western blot analyses…

Figure 3
AAV2-mediated transgene expression in minipig parotid glands. Representative results of Western blot analyses are shown. 10 ug of crude membrane protein was subjected to SDS–gel electrophoresis and Western blotting, as described in Materials and Methods. Lane 1, positive control membranes from rat kidney; lane 2, membranes from an AAV2hAQP1-transduced, irradiated minipig parotid gland (8 weeks after 1×1011 vp was delivered) showing AQP1 expression; lane 3, membranes from an AAV2LacZ-transduced, irradiated minipig parotid gland (8 weeks after 1×1011 vp was delivered) showing little AQP1 expression; lane 4, membranes from saline-infused, irradiated minipig parotid gland, also showing little AQP1expression; lane 5, membranes from the non-irradiated, contralateral side, showing AQP1 expression normally present in minipig parotid glands, which is derived from microvascular endothelial cells,,. The migration position of all immunopositive protein bands represents the nonglycosylated AQP1 monomer (28 kDa).
Figure 2
Figure 2
Effect on saliva flow rates of AAV2hAQP1 administration to minipig parotid glands after irradiation. At week 17 following 20 Gy irradiation, either AAV2hAQP1 (A, B), AAV2LacZ (B) or saline buffer (B) were administered to a single parotid gland. Panel A shows the effect of different doses of AAV2hAQP1 on parotid saliva flow rates in irradiated glands (n=2/group; data are mean values ±SD obtained 12 weeks following vector administration; note that for the 3×1011 vp dose the SD is too small to be seen). The data in Panel A was not analyzed for statistical significance, because there were only two animals/group. Panel B shows salivary flow rates at different times following administration of AAV2hAQP1, AAV2LacZ or saline buffer to irradiated parotid glands (n=3/group; data are mean values ±SD). Time zero corresponds to week 17 following irradiation. Note that for the 8-week time-point with buffer and AAV2LacZ treatment the SDs are too small to be seen. Data were analyzed using a one-way analysis of variance (F=27.88, p<0.001), followed by the Tukey test. All of the parotid flow rates for the AAV2hAQP1-treated minipigs are significantly different from those of the buffer- and AAV2LacZ-treated animals at weeks 4 (p<0.01 vs buffer; p<0.05 vs AAV2LacZ), 6 (p<0.001) and 8 (p<0.001). There were no significant differences in parotid flow rates between the buffer- and AAV2LacZ-treated animals at every time-point.
Figure 3
Figure 3
AAV2-mediated transgene expression in minipig parotid glands. Representative results of Western blot analyses are shown. 10 ug of crude membrane protein was subjected to SDS–gel electrophoresis and Western blotting, as described in Materials and Methods. Lane 1, positive control membranes from rat kidney; lane 2, membranes from an AAV2hAQP1-transduced, irradiated minipig parotid gland (8 weeks after 1×1011 vp was delivered) showing AQP1 expression; lane 3, membranes from an AAV2LacZ-transduced, irradiated minipig parotid gland (8 weeks after 1×1011 vp was delivered) showing little AQP1 expression; lane 4, membranes from saline-infused, irradiated minipig parotid gland, also showing little AQP1expression; lane 5, membranes from the non-irradiated, contralateral side, showing AQP1 expression normally present in minipig parotid glands, which is derived from microvascular endothelial cells,,. The migration position of all immunopositive protein bands represents the nonglycosylated AQP1 monomer (28 kDa).

References

    1. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics 2009. CA Cancer J Clin. 2009;59:225–249.
    1. Vissink A, Jansma J, Spijkervet FK, Burlage FR, Coppes RP. Oral sequelae of head and neck radiotherapy. Crit Rev Oral Biol Med. 2003;14:199–212.
    1. Vissink A, Burlage FR, Spijkervet FK, Jansma J, Coppes RP. Prevention and treatment of the consequences of head and neck radiotherapy. Crit Rev Oral Biol Med. 2003;14:213–225.
    1. Langendijk JA, Doornaert P, Verdonck-de Leeuw IM, Leemans CR, Aaronson NK, Slotman BJ. Impact of late treatment-related toxicity on quality of life among patients with head and neck cancer treated with radiotherapy. J Clin Oncol. 2008;26:3770–3776.
    1. Ho KF, Farnell DJ, Routledge JA, Burns MP, Sykes AJ, Slevin NJ, et al. Developing a CTCAEs patient questionnaire for late toxicity after head and neck radiotherapy. Eur J Cancer. 2009;45:1992–1998.
    1. Brizel DM, Overgaard J. Does amifostine have a role in chemoradiation treatment? Lancet Oncol. 2003;4:378–381.
    1. Vergeer MR, Doornaert P, Reitveld DH, Leemans CR, Slotman BJ, Langendijk JA. Intensity-modulated radiotherapy reduces radiation-induced morbidity and improves health-related quality of life: results of a non-randomized prospective study using a standardized follow-up program. Int J Radiat Oncol Biol Phys. 2009;74:1–8.
    1. Cotrim AP, Sowers A, Mitchell JB, Baum BJ. Prevention of irradiation-induced salivary hypofunction by microvessel protection in mouse salivary glands. Mol Ther. 2007;15:2101–2106.
    1. Epperly MW, Melendez JA, Zhang X, Nie S, Pearce L, Peterson J, et al. Mitochondrial targeting of a catalase transgene product by plasmid liposomes increases radioresistance in vitro and in vivo. Radiat Res. 2009;171:588–595.
    1. Cox JD, Steitz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for research and treatment of cancer. Int J Radiat Oncol Biol Phys. 1995;31:1341–1346.
    1. Baum BJ, Zheng C, Cotrim AP, McCullagh L, Goldsmith CM, Brahim JS, et al. Aquaporin-1 gene transfer to correct radiation-induced salivary hypofunction. In: Beitz E, editor. Aquaporins Handbook of Experimental Pharmacology. Vol. 190. Berlin Heidelberg: Springer-Verlag; 2009. pp. 403–418.
    1. Preston GM, Agre P. Isolation of the cDNA for erythrocyte integral membrane protein of 28 kilodaltons: member of an ancient channel family. Proc Natl Acad Sci U S A. 1991;88:11110–11114.
    1. Delporte C, O'Connell BC, He X, Lancaster HE, O'Connell AC, Agre P, et al. Increased fluid secretion after adenoviral-mediated transfer of the aquaporin-1 cDNA to irradiated rat salivary glands. Proc Natl Acad Sci USA. 1997;94:3268–3273.
    1. Shan Z, Li J, Zheng C, Liu X, Fan Z, Zhang C, et al. Increased fluid secretion after adenoviral-mediated transfer of the human aquaporin-1 cDNA to irradiated miniature pig parotid glands. Mol Ther. 2005;11:444–451.
    1. Zheng C, Goldsmith CM, Mineshiba F, Chiorini JA, Kerr A, Wenk ML, et al. Toxicity and biodistribution of a first-generation recombinant adenoviral vector, encoding aquaporin-1, after retroductal delivery to a single rat submandibular gland. Hum Gene Ther. 2006;17:1122–1133.
    1. http://www.clinicaltrials.gov/ct/show/NCT00372320?order=

    1. Kagami H, Atkinson JC, Michalek SM, Handelman B, Yu S, Baum BJ, et al. Repetitive adenovirus administration to the parotid gland: role of immunological barriers and induction of oral tolerance. Hum Gene Ther. 1998;9:305–313.
    1. Braddon VR, Chiorini JA, Wang S, Kotin RM, Baum BJ. Adenoassociated virus-mediated transfer of a functional water channel into salivary epithelial cells in vitro and in vivo. Hum Gene Ther. 1998;9:2777–2785.
    1. Voutetakis A, Kok MR, Zheng C, Bossis I, Wang J, Cotrim AP, et al. Reengineered salivary glands are stable endogenous bioreactors for systemic gene therapeutics. Proc Natl Acad Sci U S A. 2004;101:3053–3058.
    1. Voutetakis A, Zheng C, Mineshiba F, Cotrim AP, Goldsmith CM, Schmidt M, et al. Adeno-associated virus serotype 2-mediated gene transfer to the parotid glands of nonhuman primates. Hum Gene Ther. 2007;18:142–150.
    1. Cotrim AP, Hyodo F, Matsumoto KI, Sowers AL, Cook JA, Baum BJ, et al. Differential radiation protection of salivary glands versus tumor by Tempol with accompanying tissue assessment of Tempol by magnetic resonance imaging. Clin Cancer Res. 2007;13:4928–4933.
    1. Li J, Shan Z, Ou G, Liu X, Zhang C, Baum BJ, et al. Structural and functional characteristics of irradiation damage to parotid glands in the miniature pig. Int J Radiat Oncol Biol Phys. 2005;62:1510–1516.
    1. Hai B, Yan X, Voutetakis A, Zheng C, Cotrim AP, Shan Z, et al. Long-term transduction of miniature pig parotid glands using serotype 2 adeno-associated viral vectors. J Gene Med. 2009;11:506–514.
    1. Li J, Nielsen S, Dai Y, Lazowski KW, Christensen EI, Tabak LA, et al. Examination of rat salivary glands for the presence of the aquaporin CHIP. Pflugers Arch. 1994;428:455–460.
    1. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ, et al. Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nat Med. 2006;12:342–347.
    1. Voutetakis A, Zheng C, Cotrim AP, Mineshiba F, Afione S, Roescher N, et al. AAV5-mediated gene transfer to the parotid glands of non-human primates. Gene Ther. 2010;17:50–60.
    1. Nakamoto T, Srivastava A, Romanenko VG, Ovitt CE, Perez-Cornejo P, Arreola J, et al. Functional and molecular characterization of the fluid secretion mechanism in human parotid acinar cells. Am J Physiol Regul Integr Comp Physiol. 2007;292:R2380–R2390.
    1. Catalan MA, Nakamoto T, Melvin JE. The salivary gland fluid secretion mechanism. J Med Invest. 2009;56(Suppl):192–196.
    1. Voutetakis A, Zheng C, Wang J, Goldsmith CM, Afione S, Chiorini JA, et al. Gender differences in serotype 2 adeno-associated virus biodistribution after administration to rodent salivary glands. Hum Gene Ther. 2007;18:1109–1118.
    1. Kok MR, Voutetakis A, Yamano S, Wang J, Cotrim A, Katano H, et al. Immune responses following salivary gland administration of recombinant adeno-associated virus serotype 2 vectors. J Gene Med. 2005;7:432–441.
    1. Li J, Zheng C, Zhang X, Liu X, Zhang C, Goldsmith CM, et al. Developing a convenient large animal model for gene transfer to salivary glands in vivo. J Gene Med. 2004;6:55–63.
    1. Yan JX, Wait R, Berkelman T, Harry RA, Westbrook JA, Wheeler CH, Dunn MJ. A modified silver staining protocol for visualization of proteins compatible with matrix-assisted laser desorption/ionization and electrospray ionization-mass spectrometry. Electrophoresis. 2000;21:3666–3672.
    1. Katano H, Kok MR, Cotrim AP, Yamano S, Schmidt M, Afione S, et al. Enhanced transduction of mouse salivary glands with AAV5-based vectors. Gene Ther. 2006;13:594–601.

Source: PubMed

3
Iratkozz fel