Design and Development of Nanoparticles Containing α-Mangostin for Wound Application

Authors

  • Peerapat Chidchai Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
  • Kanokwan Singpanna Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
  • Kamonchai Ketduang Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
  • Aristarn Uamjan Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
  • Prasopchai Patrojanasophon Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
  • Chaiyakarn Pornpitchanarong Pharmaceutical Development of Green Innovations Group (PDGIG), Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.

DOI:

https://doi.org/10.31584/jhsmr.20231005

Keywords:

α-mangostin, Eudragit® S 100, nanoparticles, polycaprolactone, wound healing

Abstract

Objective: The objective of this research was to design and develop nanoparticles containing α-mangostin (α-MG) for wound applications.
Material and Methods: The nanoparticles were composed of polycaprolactone (PCL) and Eudragit® S 100 (EDG), with 10% wt of α-MG; wherein, the optimal compositions of the nanoparticles were studied using a mixture-typed simplex lattice design. The amount of PCL (5-20 milligram/milliliter (mg/mL)) and EDG (5-20 mg/mL) were varied, and the effects of the components toward particle size, size distribution, zeta potential; drug content, and drug release were examined. The physicochemical properties of the nanoparticles were analyzed using a zetasizer. The content of α-MG was quantified using High Pressure Liquid Chromatography.
Results: It was found that the nanoparticles having different mixtures of PCL and EDG did not affect the physicochemical properties nor the drug content. However, the release of α-MG can be tuned by varying the nanoparticle composition. Formulations with higher EDG showed greater drug release at pH 7.4, because of the polymer dissolution at a specified pH. The composition of the optimized formulation composed of 16.5 mg/mL of EDG and 8.5 mg/mL of PCL. The optimized nanoparticle showed a controlled release profile of up to 12 h, which was superior to the α-MG solution.
Conclusion: The developed nanoparticles of PCL and EDG can be considered as a promising platform to deliver α-MG for wound applications.

References

Venus M, Waterman J, McNab I. Basic physiology of the skin. Surgery (Oxford) 2010;28:469-72.

Nichols RL. Surgical wound infection. Am J Med 1991;91:S54-64.

Lipsky BA, Hoey C. Topical antimicrobial therapy for treating chronic wounds. Clin Infect Dis 2009;49:1541-9.

Negut I, Grumezescu V, Grumezescu AM. Treatment strategies for infected wounds. Molecules 2018;23: 2392.

Pedraza-Chaverri J, Cárdenas-Rodríguez N, Orozco-Ibarra M, Pérez-Rojas JM. Medicinal properties of mangosteen (Garcinia mangostana). Food Chem Toxicol 2008;46:3227-39.

Gutierrez-Orozco F, Chitchumroonchokchai C, Lesinski GB, Suksamrarn S, Failla ML. alpha-Mangostin: anti-inflammatory activity and metabolism by human cells. J Agric Food Chem 2013;61:3891-900.

Obolskiy D, Pischel I, Siriwatanametanon N, Heinrich M. Garcinia mangostana L.: a phytochemical and pharmacological review. Phytother Res 2009;23:1047-65.

Koh J-J, Qiu S, Zou H, Lakshminarayanan R, Li J, Zhou X, et al. Rapid bactericidal action of alpha-mangostin against MRSA as an outcome of membrane targeting. Biochim Biophys Acta 2013;1828:834-44.

Sultan OS, Kantilal HK, Khoo SP, Davamani AF, Eusufzai SZ, Rashid F, et al. The potential of alpha-mangostin from Garcinia mangostana as an effective antimicrobial agent-a systematic review and meta-analysis. Antibiotics (Basel) 2022;11:717.

Iinuma M, Tosa H, Tanaka T, Asai F, Kobayashi Y, Shimano R, et al. Antibacterial activity of xanthones from guttiferaeous plants against methicillin-resistant Staphylococcus aureus. J Pharm Pharmacol 1996;48:861-5.

Duangsrisai S, Choowongkomon K, Bessa LJ, Costa PM, Amat N, Kijjoa A. Antibacterial and EGFR-tyrosine kinase inhibitory activities of polyhydroxylated xanthones from Garcinia succifolia. Molecules 2014;19:19923-34.

Chomnawang MT, Surassmo S, Nukoolkarn VS, Gritsanapan W. Antimicrobial effects of Thai medicinal plants against acneinducing bacteria. J Ethnopharmacol 2005;101:330-3.

Lebedeva AA, Zakharchenko NS, Trubnikova EV, Medvedeva OA, Kuznetsova TV, Masgutova GA, et al. Bactericide, immunomodulating, and wound healing properties of transgenic kalanchoe pinnata synergize with antimicrobial peptide cecropin p1 in vivo. J Immunol Res 2017;2017:4645701.

Yoshikawa M, Harada E, Miki A, Tsukamoto K, Liang SQ, Yamahara J, et al. Antioxidant constituents from the fruit hulls of mangosteen (Garcinia mangostana L.) originating in Vietnam. J Pharm Soc Jpn 1994;114:129-33.

Chomnawang MT, Surassmo S, Nukoolkarn VS, Gritsanapan W. Effect of Garcinia mangostana on inflammation caused by Propionibacterium acnes. Fitoterapia 2007;78:401-8.

Narasimhan S, Maheshwaran S, Abu-Yousef IA, Majdalawieh AF, Rethavathi J, Das PE, et al. Anti-bacterial and anti-fungal activity of xanthones obtained via semi-synthetic modification of α-Mangostin from garcinia mangostana. Molecules 2017;22:275.

Wathoni N, Sari DP, Suharyani I, Motoyama K, Mohammed AFA, Cahyanto A, et al. Enhancement of α-Mangostin wound healing ability by complexation with 2-hydroxypropyl-β-cyclodextrin in hydrogel formulation. Pharmaceuticals (Basel) 2020;13290.

Kotsuchibashi Y, Nakagawa Y, Ebara M. Biomaterials Nanoarchitectonics. Bristol: William Andrew; 2016.

Sandhiya S, Dkhar SA, Surendiran A. Emerging trends of nanomedicine--an overview. Fundam Clin Pharmacol 2009;23:263-9.

Suri SS, Fenniri H, Singh B. Nanotechnology-based drug delivery systems. J Occup Med Toxicol 2007;2:16.

Losi P, Briganti E, Magera A, Spiller D, Ristori C, Battolla B, et al. Tissue response to poly(ether)urethane-polydimethylsiloxanefibrin composite scaffolds for controlled delivery of proangiogenic growth factors. Biomaterials 2010;31:5336-44.

Wang W, Lu KJ, Yu CH, Huang QL, Du YZ. Nano-drug delivery systems in wound treatment and skin regeneration. J Nanobiotechnology 2019;17:82.

Sriboonyong P, Poommarin P, Sittiya J, Opanasopit P, Ngawhirunpat T, Patrojanasophon P, et al. The utilization of mangosteen pericarp extract for anticoccidial drug replacement in broiler feed. Int J Vet Sci Med 2022;10:90-9.

Masoumi HR, Basri M, Samiun WS, Izadiyan Z, Lim CJ. Enhancement of encapsulation efficiency of nanoemulsioncontaining aripiprazole for the treatment of schizophrenia using mixture experimental design. Int J Nanomedicine 2015;10:6469-76.

Downloads

Published

2023-11-20

How to Cite

1.
Chidchai P, Singpanna K, Ketduang K, Uamjan A, Patrojanasophon P, Pornpitchanarong C. Design and Development of Nanoparticles Containing α-Mangostin for Wound Application. J Health Sci Med Res [Internet]. 2023 Nov. 20 [cited 2024 Jul. 18];42(1):e20231005. Available from: https://he01.tci-thaijo.org/index.php/jhsmr/article/view/267198

Issue

Section

Original Article