scholarly journals Evaluation of physicochemical characteristics of hydrophobically modified glycol chitosan nanoparticles and their biocompatibility in murine osteosarcoma and osteoblast-like cells

2019 ◽  
2006 ◽  
Vol 111 (1-2) ◽  
pp. 228-234 ◽  
Author(s):  
Jong-Ho Kim ◽  
Yoo-Shin Kim ◽  
Sungwon Kim ◽  
Jae Hyung Park ◽  
Kwangmeyung Kim ◽  
...  

2020 ◽  
Vol 18 ◽  
pp. 228080002096262 ◽  
Author(s):  
Zhongqing Wu ◽  
Kanna Xu ◽  
Jikang Min ◽  
Minchang Chen ◽  
Liping Shen ◽  
...  

Background: Targeted delivery to the Rheumatoid arthritis (RA) which is characterized by destruction and degeneration of bones due to chronic inflammation is of great need. RA being a chronic autoimmune disorder might result in severe disability and morbidity. A targeted delivery system is designed to deliver methotrexate (MTX) for RA. Methods: Here, we synthesized folic acid (FA) conjugated hydrophobically modified glycol chitosan (GC) self-assembled nanoparticles (FA-GC-SA) for the targeted delivery of MTX to RA. The FA conjugation and hydrophobic modification of GC by stearic acid (SA) was confirmed by Fourier-transform infrared spectroscopy (FTIR). The FA-GC-SA was exploited for developing targeted nanoparticles encapsulating MTX by the ionic gelation method. The particles were characterized and evaluated for their targeting potential in in vitro cell culture studies. Further their in vivo efficacy in arthritis induced rats using collagen was also evaluated. Results: FTIR confirms the successful modification of GC-SA and FA-GC-SA. The FA-GC-SA-MTX of size 153 ± 9 nm were prepared with high encapsulation efficiency of MTX. The FA-GC-SA-MTX size was further confirmed by transmission electron microscopy (TEM). In vitro cell studies revealed the superior efficacy of FA-GC-SA-MTX in cell cytotoxicity. Also, significantly higher cellular uptake of FA functionalized FA-GC-SA-MTX was observed in comparison to non-functionalized GC-SA-MTX attributed to folate receptors (FRs) mediated endocytosis. In vivo results confirms the potential of FA-GC-SA-MTX which reduces reduces the pro-inflammatory cytokines, paw thickness, and arthritis score in collagen induced rats. Conclusion: The results shows that FRs targeted FA-GC-SA-MTX has superior efficacy in the treatment of RA.


2021 ◽  
Author(s):  
Wai Mun Chong ◽  
VUANGHAO LIM ◽  
Erazuliana Abd Kadir

A novel palmitoylated glycol chitosan polymer grafted with PEG (PGC-PEG) was successfully developed to form amphiphilic micelles in aqueous solution. The incorporation of hydrophobic itraconazole (ITZ) with PGC-PEG polymer produced...


2009 ◽  
Vol 135 (3) ◽  
pp. 259-267 ◽  
Author(s):  
Hae Yun Nam ◽  
Seok Min Kwon ◽  
Hyunjin Chung ◽  
Seung-Young Lee ◽  
Seung-Hae Kwon ◽  
...  

2016 ◽  
Vol 11 (4) ◽  
pp. 04B301 ◽  
Author(s):  
Wei Yin ◽  
Weiyi Li ◽  
David A. Rubenstein ◽  
Yizhi Meng

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Chang Seong Kim ◽  
Ansuja Pulickal Mathew ◽  
Arathy Vasukutty ◽  
Saji Uthaman ◽  
Soo Yeon Joo ◽  
...  

Abstract Background Recently, we developed hydrophobically modified glycol chitosan (HGC) nanomicelles loaded with tacrolimus (TAC) (HGC-TAC) for the targeted renal delivery of TAC. Herein, we determined whether the administration of the HGC-TAC nanomicelles decreases kidney injury in a model of lupus nephritis. Lupus-prone female MRL/lpr mice were randomly assigned into three groups that received intravenous administration of either vehicle control, an equivalent dose of TAC, or HGC-TAC (0.5 mg/kg TAC) weekly for 8 weeks. Age-matched MRL/MpJ mice without Faslpr mutation were also treated with HGC vehicle and used as healthy controls. Results Weekly intravenous treatment with HGC-TAC significantly reduced genetically attributable lupus activity in lupus nephritis-positive mice. In addition, HGC-TAC treatment mitigated renal dysfunction, proteinuria, and histological injury, including glomerular proliferative lesions and tubulointerstitial infiltration. Furthermore, HGC-TAC treatment reduced renal inflammation and inflammatory gene expression and ameliorated increased apoptosis and glomerular fibrosis. Moreover, HGC-TAC administration regulated renal injury via the TGF-β1/MAPK/NF-κB signaling pathway. These renoprotective effects of HGC-TAC treatment were more potent in lupus mice compared to those of TAC treatment alone. Conclusion Our study indicates that weekly treatment with the HGC-TAC nanomicelles reduces kidney injury resulting from lupus nephritis by preventing inflammation, fibrosis, and apoptosis. This advantage of a new therapeutic modality using kidney-targeted HGC-TAC nanocarriers may improve drug adherence and provide treatment efficacy in lupus nephritis mice.


Sign in / Sign up

Export Citation Format

Share Document