We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Flow vibration-doubled concentric system coupled with low ratio amine to produce bile acid-macrocapsules of β-cells

    Armin Mooranian

    Biotechnology & Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia

    ,
    Rebecca Negrulj

    Biotechnology & Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia

    &
    Hani Al-Salami

    *Author for correspondence:

    E-mail Address: hani.al-salami@curtin.edu.au

    Biotechnology & Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia

    Published Online:https://doi.org/10.4155/tde-2015-0001

    Background: Pancreatic β-cell microencapsulation using sodium alginate (SA), polylornithine (PLO) copolymers, and ultrasoluble hydrogels, polystyrenes and polyallamines (PAA), has been heavily studied. However, long-term success remains limited due to poor macrocapsules’ physical properties and cell functions. Our study aimed to incorporate percentages of PAA and ursodeoxycholic acid, into SA and PLO dispersion mixture and examine best microencapsulating methods and best macrocapsules containing β-cells. Methods/results: Microencapsulating parameters were examined and the Flow-Vibrational Nozzle built-in system was screened and found to be most efficient at high frequency (1900 Hz). Macrocapsules were produced with or without ursodeoxycholic acid in percentages: 0.018SA:0.01PLO:0.005PAA:0.04ursodeoxycholic acid (up to 100% H2O). Using the refined microencapsulation method with vibrational frequency of 1900 Hz, macrocapsules with ursodeoxycholic acid had optimized cell viability and biological functions and ameliorated inflammatory biomarkers. Conclusion: High frequency and air-pressure with Flow-Vibrational encapsulation using the mixture: 0.018SA:0.01PLO:0.005PAA:0.04ursodeoxycholic acid resulted in better cell biology suggesting potentials in β-cell transplantation.

    References

    • 1 Fakhoury M, Negrulj R, Mooranian A et al. Inflammatory bowel disease: clinical aspects and treatments. J. Inflamm. Res. 7, 113–120 (2014).
    • 2 Negrulj R, Mooranian A, Al-Salami H. Potentials and limitations of bile acids in Type 2 diabetes mellitus: applications of microencapsulation as a novel oral delivery system. J. Endocrinol. Diabetes Mellit. 1(2), 49–59 (2013).
    • 3 De Vos P, Lazarjani HA, Poncelet D, Faas MM. Polymers in cell encapsulation from an enveloped cell perspective. Adv. Drug Deliv. Rev. 67–68, 15–34 (2014).
    • 4 Rokstad AM, Lacik I, De Vos P, Strand BL. Advances in biocompatibility and physico–chemical characterization of microspheres for cell encapsulation. Adv. Drug Deliv. Rev. 67–68, 111–130 (2014).
    • 5 Shapiro AM, Ricordi C, Hering BJ et al. International trial of the Edmonton protocol for islet transplantation. N. Engl. J. Med. 355(13), 1318–1330 (2006).
    • 6 Goss JA, Soltes G, Goodpastor SE et al. Pancreatic islet transplantation: the radiographic approach. Transplantation 76(1), 199–203 (2003).
    • 7 De Vos P, Faas MM, Strand B, Calafiore R. Alginate-based macrocapsules for immunoisolation of pancreatic islets. Biomaterials 27(32), 5603–5617 (2006).
    • 8 Chang TMS. Therapeutic applications of polymeric artificial cells. Nat. Rev. Drug Discov. 4(3), 221–235 (2005).
    • 9 Orive G, Maria Hernández R, RodríGuez Gascón A et al. History, challenges and perspectives of cell microencapsulation. Trends Biotechnol. 22(2), 87–92 (2004).
    • 10 Schmidt JJ, Rowley J, Kong HJ. Hydrogels used for cell-based drug delivery. J. Biomed. Mater. Res A. 87(4), 1113–1122 (2008).
    • 11 Bhatia SR, Khattak SF, Roberts SC. Polyelectrolytes for cell encapsulation. Curr. Opin. Colloid Interface Sci. 10(1), 45–51 (2005).
    • 12 Mooranian A, Negrulj R, Chen-Tan N et al. Advanced bile acid-based multi-compartmental microencapsulated pancreatic beta-cells integrating a polyelectrolyte-bile acid formulation, for diabetes treatment. Artif. Cells Nanomed. Biotechnol. 44(2), 588–595 (2016).
    • 13 Mooranian A, Negrulj R, Arfuso F, Al-Salami H. Characterization of a novel bile acid-based delivery platform for microencapsulated pancreatic β-cells. Artif. Cells Nanomed. Biotechnol. 44(1), 194–200 (2016).
    • 14 Martinez-Moya P, Romero-Calvo I, Requena P et al. Dose-dependent antiinflammatory effect of ursodeoxycholic acid in experimental colitis. Int. Immunopharmacol. 15(2), 372–380 (2013).
    • 15 Mooranian A, Negrulj R, Mathavan S et al. An advanced microencapsulated system: a platform for optimized oral delivery of antidiabetic drug-bile acid formulations. Pharm. Dev. Technol. 20(6), 702–709 (2015).
    • 16 Mooranian A, Negrulj R, Arfuso F, Al-Salami H. Multicompartmental, multilayered probucol macrocapsules for diabetes mellitus: formulation characterization and effects on production of insulin and inflammation in a pancreatic beta-cell line. Artif. Cells Nanomed. Biotechnol. doi:10.3109/21691401.2015.1069299, 1–12 (2015) (Epub ahead of print).
    • 17 Mooranian A, Negrulj R, Arfuso F, Al-Salami H. The effect of a tertiary bile acid, taurocholic acid, on the morphology and physical characteristics of microencapsulated probucol: potential applications in diabetes: a characterization study. Drug Deliv. Transl. Res. 5(5), 511–522 (2015).
    • 18 Mooranian A, Negrulj R, Mikov M, Golocorbin-Kon S, Arfuso F, Al-Salami H. Novel chenodeoxycholic acid-sodium alginate matrix in the microencapsulation of the potential antidiabetic drug, probucol. An in vitro study. J. Microencapsul. 32(6), 589–597 (2015).
    • 19 Negrulj R, Mooranian A, Chen-Tan N et al. Swelling, mechanical strength, and release properties of probucol macrocapsules with and without a bile acid, and their potential oral delivery in diabetes. Artif. Cells Nanomed. Biotechnol. doi:10.3109/21691401.2015.1024845, 1–8 (2015).
    • 20 Mooranian A, Negrulj R, Arfuso F, Al-Salami H. Characterization of a novel bile acid-based delivery platform for microencapsulated pancreatic β-cells. Artif. Cells Nanomed. Biotechnol. 44(1), 194–200 (2016).
    • 21 Mooranian A, Negrulj R, Al-Salami H. The effects of ionic gelation – Vibrational Jet Flow technique in fabrication of macrocapsules incorporating beta-cell: applications in Type 1 diabetes. Curr. Diabetes Rev. doi:10.2174/1573399812666151229101756#sthash.HD4rFRaQ.dpuf (2015).
    • 22 Mooranian A, Negrulj R, Al-Salami H. The incorporation of water-soluble gel matrix into bile acid-based macrocapsules for the delivery of viable beta-cells of the pancreas, in diabetes treatment: biocompatibility and functionality studies. Drug Deliv. Transl. Res. 6(1), 17–23 (2016).
    • 23 Mooranian A, Negrulj R, Al-Salami H. Viability and topographical analysis of microencapsulated β-cells exposed to a biotransformed tertiary bile acid: an ex vivo study. Int. J. Nano. Biomaterials (2016) (In Press).
    • 24 Mooranian A, Negrulj R, Al-Salami H. The influence of stabilized deconjugated ursodeoxycholic acid on polymer-hydrogel system of transplantable NIT-1 cells. Pharm. Res. (2016) (In Press).
    • 25 Mooranian A, Negrulj R, Al-Salami H. The incorporation of water-soluble gel matrix into bile acid-based macrocapsules for the delivery of viable beta-cells of the pancreas, in diabetes treatment: biocompatibility and functionality studies. Drug Deliv. Transl. Res. 6(1), 17–23 (2016).
    • 26 Mooranian A, Negrulj R, Morahan G, Jamieson E, Al-Salami H. Designing anti-diabetic beta-cells macrocapsules using polystyrenic sulfonate, polyallylamine and a tertiary bile acid: morphology, bioenergetics and cytokine analysis. Biotechnol. Prog. doi:10.1002/btpr.2223 (2016) (Epub ahead of print).
    • 27 Hamaguchi K, Gaskins HR, Leiter EH. NIT-1, a pancreatic beta-cell line established from a transgenic NOD/Lt mouse. Diabetes 40(7), 842–849 (1991).
    • 28 Mooranian A, Negrulj R, Chen-Tan N et al. Microencapsulation as a novel delivery method for the potential antidiabetic drug, Probucol. Drug Des. Devel. Ther. 8, 1221–1230 (2014).
    • 29 Mooranian A, Negrulj R, Mathavan S et al. An advanced microencapsulated system: a platform for optimized oral delivery of antidiabetic drug-bile acid formulations. Pharm. Dev. Technol. 20(6), 702–709 (2015).
    • 30 Wu M, Neilson A, Swift AL et al. Multiparameter metabolic analysis reveals a close link between attenuated mitochondrial bioenergetic function and enhanced glycolysis dependency in human tumor cells. Am. J. Physiol. Cell. Physiol. 292(1), C125–136 (2007).
    • 31 Gerencser AA, Neilson A, Choi SW et al. Quantitative microplate-based respirometry with correction for oxygen diffusion. Anal. Chem. 81(16), 6868–6878 (2009).
    • 32 Donath MY, Boni-Schnetzler M, Ellingsgaard H, Ehses JA. Islet inflammation impairs the pancreatic beta-cell in Type 2 diabetes. Physiology (Bethesda, MD, USA) 24, 325–331 (2009).
    • 33 Barshes NR, Wyllie S, Goss JA. Inflammation-mediated dysfunction and apoptosis in pancreatic islet transplantation: implications for intrahepatic grafts. J. Leukoc. Biol. 77(5), 587–597 (2005).
    • 34 Naldini A, Carraro F, Silvestri S, Bocci V. Hypoxia affects cytokine production and proliferative responses by human peripheral mononuclear cells. J Cell Physiol. 173(3), 335–342 (1997).
    • 35 Donath MY, Böni-Schnetzler M, Ellingsgaard H, Halban PA, Ehses JA. Cytokine production by islets in health and diabetes: cellular origin, regulation and function. Trends Endocrinol. Metab. 21(5), 261–267 (2010).
    • 36 Alexandraki K, Piperi C, Kalofoutis C, Singh J, Alaveras A, Kalofoutis A. Inflammatory process in Type 2 diabetes: the role of cytokines. Ann. NY Acad. Sci. 1084, 89–117 (2006).
    • 37 Takka S, Cali AG. Bile salt-reinforced alginate–chitosan beads. Pharm. Dev. Technol. 17(1), 23–29 (2012).