Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Lentiviral gene therapy vectors encoding VIP suppressed diabetes-related inflammation and augmented pancreatic beta-cell proliferation

Abstract

Type 1 diabetes (T1DM) is an autoimmune condition in which the immune system attacks and destroys insulin-producing beta cells in the pancreas leading to hyperglycemia. Vasoactive intestinal peptide (VIP) manifests insulinotropic and anti-inflammatory properties, which are useful for the treatment of diabetes. Because of its limited half-life due to DPP-4-mediated degradation, constant infusions or multiple injections are needed to observe any therapeutic benefit. Since gene therapy has the potential to treat genetic diseases, an HIV-based lentiviral vector carrying VIP gene (LentiVIP) was generated to provide a stable VIP gene expression in vivo. The therapeutic efficacy of LentiVIP was tested in a multiple low-dose STZ-induced animal model of T1DM. LentiVIP delivery into diabetic animals reduced hyperglycemia, improved glucose tolerance, and prevented weight loss. Also, a decrease in serum CRP levels, and serum oxidant capacity, but an increase in antioxidant capacity were observed in LentiVIP-treated animals. Restoration of islet cell mass was correlated with an increase in pancreatic beta-cell proliferation. These beneficial results suggest the therapeutic effect of LentiVIP is due to the repression of diabetes-induced inflammation, its insulinotropic properties, and VIP-induced beta-cell proliferation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Experimental animal model of Type 1 diabetes. STZ (30 mg/kg) was injected IP into Wistar or Sprague Dawley rats (n = 8) for 5 consecutive days.
Fig. 2: Construction of pLentiVIP transfer plasmid for lentivirus vector production using Gateway Technology.
Fig. 3: In vitro expression and functional analysis of the LentiVIP vector.
Fig. 4: Therapeutic efficacy of LentiVIP injection in STZ-induced rat model of diabetes.
Fig. 5: Improved glucose tolerance in LentiVIP-injected rats correlated with higher serum VIP levels.
Fig. 6: VIP restores beta-cell loss in STZ-induced diabetes.
Fig. 7: LentiVIP suppresses diabetes-induced inflammation.

Similar content being viewed by others

References

  1. Michels A, Zhang L, Khadra A, Kushner JA, Redondo MJ, Pietropaolo M. Prediction and prevention of type 1 diabetes: update on success of prediction and struggles at prevention. Pediatr Diabetes. 2015;16:465–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Krzewska A, Ben-Skowronek I. Effect of associated autoimmune diseases on type 1 diabetes mellitus incidence and metabolic control in children and adolescents. Biomed Res Int. 2016;2016:6219730.

    PubMed  PubMed Central  Google Scholar 

  3. Cerna M. Epigenetic regulation in etiology of type 1 diabetes mellitus. Int J Mol Sci. 2019;21:1–17.

    Google Scholar 

  4. Lee HS, Hwang JS. Genetic aspects of type 1 diabetes. Ann Pediatr Endocrinol Metab. 2019;24:143–8.

    PubMed  PubMed Central  Google Scholar 

  5. DiMeglio LA, Evans-Molina C, Oram RA. Type 1 diabetes. Lancet. 2018;391:2449–62.

    PubMed  PubMed Central  Google Scholar 

  6. Padgett LE, Broniowska KA, Hansen PA, Corbett JA, Tse HM. The role of reactive oxygen species and proinflammatory cytokines in type 1 diabetes pathogenesis. Ann N Y Acad Sci. 2013;1281:16–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Lu J, Liu J, Li L, Lan Y, Liang Y. Cytokines in type 1 diabetes: mechanisms of action and immunotherapeutic targets. Clin Transl Immunology. 2020;9:e1122.

    PubMed  PubMed Central  Google Scholar 

  8. Nepom GT, Ehlers M, Mandrup-Poulsen T. Anti-cytokine therapies in T1D: concepts and strategies. Clin Immunol. 2013;149:279–85.

    CAS  PubMed  Google Scholar 

  9. Mastrandrea L, Yu J, Behrens T, Buchlis J, Albini C, Fourtner S, et al. Etanercept treatment in children with new-onset type 1 diabetes: pilot randomized, placebo-controlled, double-blind study. Diabetes Care. 2009;32:1244–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Rosenzwajg M, Churlaud G, Mallone R, Six A, Derian N, Chaara W, et al. Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun. 2015;58:48–58.

    CAS  PubMed  Google Scholar 

  11. Sanlioglu AD, Karacay B, Balci MK, Griffith TS, Sanlioglu S. Therapeutic potential of VIP vs PACAP in diabetes. J Mol Endocrinol. 2012;49:R157–67.

    CAS  PubMed  Google Scholar 

  12. Sragovich S, Amram N, Yeheskel A, Gozes I. VIP/PACAP-based drug development: the ADNP/NAP-derived mirror peptides SKIP and D-SKIP exhibit distinctive in vivo and in silico effects. Front Cell Neurosci. 2019;13:589.

    CAS  PubMed  Google Scholar 

  13. Hannibal J, Fahrenkrug J. Pituitary adenylate cyclase-activating polypeptide in intrinsic and extrinsic nerves of the rat pancreas. Cell Tissue Res. 2000;299:59–70.

    CAS  PubMed  Google Scholar 

  14. Yada T, Sakurada M, Ishihara H, Nakata M, Shioda S, Yaekura K, et al. Pituitary adenylate cyclase-activating polypeptide (PACAP) is an islet substance serving as an intra-islet amplifier of glucose-induced insulin secretion in rats. J Physiol. 1997;505:319–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Ganea D, Hooper KM, Kong W. The neuropeptide vasoactive intestinal peptide: direct effects on immune cells and involvement in inflammatory and autoimmune diseases. Acta Physiol (Oxf). 2015;213:442–52.

    CAS  Google Scholar 

  16. Martinez C, Juarranz Y, Gutierrez-Canas I, Carrion M, Perez-Garcia S, Villanueva-Romero R, et al. A clinical approach for the use of VIP axis in inflammatory and autoimmune diseases. Int J Mol Sci. 2019;21:1–44.

    CAS  Google Scholar 

  17. Lodde BM, Delporte C, Goldsmith CM, Tak PP, Baum BJ. A recombinant adenoviral vector encoding functional vasoactive intestinal peptide. Biochem Biophys Res Commun. 2004;319:189–92.

    CAS  PubMed  Google Scholar 

  18. Lodde BM, Mineshiba F, Wang J, Cotrim AP, Afione S, Tak PP, et al. Effect of human vasoactive intestinal peptide gene transfer in a murine model of Sjogren’s syndrome. Ann Rheum Dis. 2006;65:195–200.

    CAS  PubMed  Google Scholar 

  19. Herrera JL, Fernandez-Montesinos R, Gonzalez-Rey E, Delgado M, Pozo D. Protective role for plasmid DNA-mediated VIP gene transfer in non-obese diabetic mice. Ann N Y Acad Sci. 2006;1070:337–41.

    CAS  PubMed  Google Scholar 

  20. Zhou Y, Zhang CY, Duan JX, Li Q, Yang HH, Sun CC, et al. Vasoactive intestinal peptide suppresses the NLRP3 inflammasome activation in lipopolysaccharide-induced acute lung injury mice and macrophages. Biomed Pharmacother. 2020;121:109596.

    CAS  PubMed  Google Scholar 

  21. Zhang YF, Zhang J, Sun CC, Tang CY, Sun GY, Luo WJ, et al. Vasoactive intestinal peptide inhibits the activation of murine fibroblasts and expression of interleukin 17 receptor C. Cell Biol Int. 2019;43:770–80.

    CAS  PubMed  Google Scholar 

  22. Sun GY, Yang HH, Guan XX, Zhong WJ, Liu YP, Du MY, et al. Vasoactive intestinal peptide overexpression mediated by lentivirus attenuates lipopolysaccharide-induced acute lung injury in mice by inhibiting inflammation. Mol Immunol. 2018;97:8–15.

    CAS  PubMed  Google Scholar 

  23. Tasyurek HM, Eksi YE, Sanlioglu AD, Altunbas HA, Balci MK, Griffith TS, et al. HIV-based lentivirus-mediated vasoactive intestinal peptide gene delivery protects against DIO animal model of Type 2 diabetes. Gene Ther. 2018;25:269–83.

    CAS  PubMed  Google Scholar 

  24. Sanlioglu AD, Griffith TS, Omer A, Dirice E, Sari R, Altunbas HA, et al. Molecular mechanisms of death ligand-mediated immune modulation: a gene therapy model to prolong islet survival in type 1 diabetes. J Cell Biochem. 2008;104:710–20.

    CAS  PubMed  Google Scholar 

  25. Sanlioglu AD, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Clinical utility of insulin and insulin analogs. Islets. 2013;5:67–78.

    PubMed  PubMed Central  Google Scholar 

  26. Sanlioglu AD, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Insulin gene therapy from design to beta cell generation. Expert Rev Mol Med. 2012;14:e18.

    PubMed  Google Scholar 

  27. Tasyurek HM, Altunbas HA, Balci MK, Griffith TS, Sanlioglu S. Therapeutic potential of lentivirus-mediated glucagon-like peptide-1 gene therapy for diabetes. Hum Gene Ther. 2018;29:802–15.

    CAS  PubMed  Google Scholar 

  28. Chen YG, Mathews CE, Driver JP. The role of NOD mice in type 1 diabetes research: lessons from the past and recommendations for the future. Front Endocrinol. 2018;9:51.

    CAS  Google Scholar 

  29. Dirice E, Kahraman S, Elpek GO, Aydin C, Balci MK, Omer A, et al. TRAIL and DcR1 expressions are differentially regulated in the pancreatic islets of STZ- versus CY-applied NOD mice. Exp Diabetes Res. 2011;2011:625813.

    PubMed  PubMed Central  Google Scholar 

  30. Rosignoli F, Torroba M, Juarranz Y, Garcia-Gomez M, Martinez C, Gomariz RP, et al. VIP and tolerance induction in autoimmunity. Ann N Y Acad Sci. 2006;1070:525–30.

    CAS  PubMed  Google Scholar 

  31. Kahraman S, Aydin C, Elpek GO, Dirice E, Sanlioglu AD, Diabetes-resistant NOR. mice are more severely affected by streptozotocin compared to the diabetes-prone NOD mice: correlations with liver and kidney GLUT2 expressions. J Diabetes Res. 2015;2015:450128.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Roep BO, Atkinson M, von Herrath M. Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol. 2004;4:989–97.

    CAS  PubMed  Google Scholar 

  33. Atkinson MA, Leiter EH. The NOD mouse model of type 1 diabetes: as good as it gets? Nat Med. 1999;5:601–4.

    CAS  PubMed  Google Scholar 

  34. Tasyurek MH, Altunbas HA, Canatan H, Griffith TS, Sanlioglu S. GLP-1-mediated gene therapy approaches for diabetes treatment. Expert Rev Mol Med. 2014;16:e7.

    PubMed  Google Scholar 

  35. Wang Z, Gleichmann H. GLUT2 in pancreatic islets: crucial target molecule in diabetes induced with multiple low doses of streptozotocin in mice. Diabetes. 1998;47:50–6.

    CAS  PubMed  Google Scholar 

  36. Muller A, Schott-Ohly P, Dohle C, Gleichmann H. Differential regulation of Th1-type and Th2-type cytokine profiles in pancreatic islets of C57BL/6 and BALB/c mice by multiple low doses of streptozotocin. Immunobiology. 2002;205:35–50.

    CAS  PubMed  Google Scholar 

  37. O’Brien BA, Harmon BV, Cameron DP, Allan DJ. Beta-cell apoptosis is responsible for the development of IDDM in the multiple low-dose streptozotocin model. J Pathol. 1996;178:176–81.

    PubMed  Google Scholar 

  38. Li Z, Karlsson FA, Sandler S. Islet loss and alpha cell expansion in type 1 diabetes induced by multiple low-dose streptozotocin administration in mice. J Endocrinol. 2000;165:93–9.

    CAS  PubMed  Google Scholar 

  39. Dirice E, Sanlioglu AD, Kahraman S, Ozturk S, Balci MK, Omer A, et al. Adenovirus-mediated TRAIL gene (Ad5hTRAIL) delivery into pancreatic islets prolongs normoglycemia in streptozotocin-induced diabetic rats. Hum Gene Ther. 2009;20:1177–89.

    CAS  PubMed  Google Scholar 

  40. Akbarzadeh A, Norouzian D, Mehrabi MR, Jamshidi S, Farhangi A, Verdi AA, et al. Induction of diabetes by Streptozotocin in rats. Indian J Clin Biochem. 2007;22:60–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gale EA. The discovery of type 1 diabetes. Diabetes. 2001;50:217–26.

    CAS  PubMed  Google Scholar 

  42. In’t Veld P. Insulitis in human type 1 diabetes: The quest for an elusive lesion. Islets. 2011;3:131–8.

    PubMed  PubMed Central  Google Scholar 

  43. Cecil RL. A study of the pathological anatomy of the pancreas in ninety cases of diabetes mellitus. J Exp Med. 1909;11:266–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Doniach I, Morgan AG. Islets of Langerhans in juvenile diabetes mellitus. Clin Endocrinol. 1973;2:233–48.

    CAS  Google Scholar 

  45. Campbell-Thompson M, Fu A, Kaddis JS, Wasserfall C, Schatz DA, Pugliese A, et al. Insulitis and beta-cell mass in the natural history of type 1 diabetes. Diabetes. 2016;65:719–31.

    CAS  PubMed  Google Scholar 

  46. Kato I, Suzuki Y, Akabane A, Yonekura H, Tanaka O, Kondo H, et al. Transgenic mice overexpressing human vasoactive intestinal peptide (VIP) gene in pancreatic beta cells. Evidence for improved glucose tolerance and enhanced insulin secretion by VIP and PHM-27 in vivo. J Biol Chem. 1994;269:21223–8.

    CAS  PubMed  Google Scholar 

  47. Asnicar MA, Koster A, Heiman ML, Tinsley F, Smith DP, Galbreath E, et al. Vasoactive intestinal polypeptide/pituitary adenylate cyclase-activating peptide receptor 2 deficiency in mice results in growth retardation and increased basal metabolic rate. Endocrinology. 2002;143:3994–4006.

    CAS  PubMed  Google Scholar 

  48. Hultgardh-Nilsson A, Nilsson J, Jonzon B, Dalsgaard CJ. Growth-inhibitory properties of vasoactive intestinal polypeptide. Regul Pept. 1988;22:267–74.

    CAS  PubMed  Google Scholar 

  49. Maruno K, Absood A, Said SI. VIP inhibits basal and histamine-stimulated proliferation of human airway smooth muscle cells. Am J Physiol. 1995;268:L1047–51.

    CAS  PubMed  Google Scholar 

  50. Martin B, Shin YK, White CM, Ji S, Kim W, Carlson OD, et al. Vasoactive intestinal peptide-null mice demonstrate enhanced sweet taste preference, dysglycemia, and reduced taste bud leptin receptor expression. Diabetes. 2010;59:1143–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Yamamoto K, Hashimoto H, Tomimoto S, Shintani N, Miyazaki J, Tashiro F, et al. Overexpression of PACAP in transgenic mouse pancreatic beta-cells enhances insulin secretion and ameliorates streptozotocin-induced diabetes. Diabetes. 2003;52:1155–62.

    CAS  PubMed  Google Scholar 

  52. Edvell A, Lindstrom P. Initiation of increased pancreatic islet growth in young normoglycemic mice (Umea +/?). Endocrinology. 1999;140:778–83.

    CAS  PubMed  Google Scholar 

  53. Yamamoto J, Imai J, Izumi T, Takahashi H, Kawana Y, Takahashi K, et al. Neuronal signals regulate obesity induced beta-cell proliferation by FoxM1 dependent mechanism. Nat Commun. 2017;8:1930.

    PubMed  PubMed Central  Google Scholar 

  54. Xu G, Stoffers DA, Habener JF, Bonner-Weir S. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes. 1999;48:2270–6.

    CAS  PubMed  Google Scholar 

  55. Muriach M, Flores-Bellver M, Romero FJ, Barcia JM. Diabetes and the brain: oxidative stress, inflammation, and autophagy. Oxid Med Cell Longev. 2014;2014:102158.

    PubMed  PubMed Central  Google Scholar 

  56. Jha JC, Ho F, Dan C, Jandeleit-Dahm K. A causal link between oxidative stress and inflammation in cardiovascular and renal complications of diabetes. Clin Sci. 2018;132:1811–36.

    CAS  Google Scholar 

  57. Snell-Bergeon JK, West NA, Mayer-Davis EJ, Liese AD, Marcovina SM, D’Agostino RB Jr., et al. Inflammatory markers are increased in youth with type 1 diabetes: the SEARCH case-control study. J Clin Endocrinol Metab. 2010;95:2868–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Castelblanco E, Hernandez M, Castelblanco A, Gratacos M, Esquerda A, Mollo A, et al. Low-grade inflammatory marker profile may help to differentiate patients with LADA, classic adult-onset type 1 diabetes, and type 2 diabetes. Diabetes Care. 2018;41:862–8.

    CAS  PubMed  Google Scholar 

  59. Tekula S, Khurana A, Anchi P, Godugu C. Withaferin-A attenuates multiple low doses of Streptozotocin (MLD-STZ) induced type 1 diabetes. Biomed Pharmacother. 2018;106:1428–40.

    CAS  PubMed  Google Scholar 

  60. Pietropaolo M, Barinas-Mitchell E, Kuller LH. The heterogeneity of diabetes: unraveling a dispute: is systemic inflammation related to islet autoimmunity? Diabetes. 2007;56:1189–97.

    CAS  PubMed  Google Scholar 

  61. Milone MC, O’Doherty U. Clinical use of lentiviral vectors. Leukemia. 2018;32:1529–41.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Olgun HB, Tasyurek HM, Sanlioglu AD, Sanlioglu S. High-titer production of HIV-based lentiviral vectors in roller bottles for gene and cell therapy. Methods Mol Biol. 2019;1879:323–45.

    CAS  PubMed  Google Scholar 

  63. Olgun HB, Tasyurek HM, Sanlioglu AD, Sanlioglu S. High-grade purification of third-generation HIV-based lentiviral vectors by anion exchange chromatography for experimental gene and stem cell therapy applications. Methods Mol Biol. 2019;1879:347–65.

    CAS  PubMed  Google Scholar 

  64. He Y, Shi B, Zhao X, Sui J. Sphingosine-1-phosphate induces islet beta-cell proliferation and decreases cell apoptosis in high-fat diet/streptozotocin diabetic mice. Exp Ther Med. 2019;18:3415–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Malik A, Morya RK, Bhadada SK, Rana S. Type 1 diabetes mellitus: complex interplay of oxidative stress, cytokines, gastrointestinal motility and small intestinal bacterial overgrowth. Eur J Clin Investig. 2018;48:e13021.

    Google Scholar 

  66. Yaribeygi H, Atkin SL, Sahebkar A. A review of the molecular mechanisms of hyperglycemia-induced free radical generation leading to oxidative stress. J Cell Physiol. 2019;234:1300–12.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by the Akdeniz University Scientific Research Administration Division and “the gene therapy for diabetes” project funded by The Scientific and Technological Research Council of Turkey (TUBITAK) under Grant No: 215S820.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Salih Sanlioglu.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Erendor, F., Sahin, E.O., Sanlioglu, A.D. et al. Lentiviral gene therapy vectors encoding VIP suppressed diabetes-related inflammation and augmented pancreatic beta-cell proliferation. Gene Ther 28, 130–141 (2021). https://doi.org/10.1038/s41434-020-0183-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41434-020-0183-3

Search

Quick links