Cargando…

Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes

BACKGROUND: Keloids represent benign fibroproliferative tumors which result from elevated expression of inflammation. Paclitaxel (PTX) was an effective chemotherapeutic agent and has been reported to have anti-fibrotic effects, but the strong hydrophobicity brings a challenge for its clinical applic...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Mengjiao, Chen, Liqing, Huang, Wei, Jin, Mingji, Wang, Qiming, Gao, Zhonggao, Jin, Zhehu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove Medical Press 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6390862/
https://www.ncbi.nlm.nih.gov/pubmed/30863067
_version_ 1783398217546727424
author Wang, Mengjiao
Chen, Liqing
Huang, Wei
Jin, Mingji
Wang, Qiming
Gao, Zhonggao
Jin, Zhehu
author_facet Wang, Mengjiao
Chen, Liqing
Huang, Wei
Jin, Mingji
Wang, Qiming
Gao, Zhonggao
Jin, Zhehu
author_sort Wang, Mengjiao
collection PubMed
description BACKGROUND: Keloids represent benign fibroproliferative tumors which result from elevated expression of inflammation. Paclitaxel (PTX) was an effective chemotherapeutic agent and has been reported to have anti-fibrotic effects, but the strong hydrophobicity brings a challenge for its clinical application. PURPOSE: The objective of this study was to improve the water solubility of PTX and investigate its anti-keloid effects. METHODS: We prepared a PTX-cholesterol-loaded liposomes (PTXL) by thin film evaporation fashion and characterized their physicochemical properties. We also investigated the effects of PTX on proliferation, invasion and fibrosis of keloid fibroblasts in vitro and in vivo. RESULTS: The prepared PTXL have a spherical appearance, a particle size of 101.43 nm and a zeta potential of −41.63 mV. PTXL possessed a high drug entrapment efficiency of 95.63% and exhibited a good stability within 30 days. The drugs in PTXL were released in a slow and sustained mode. The PTXL could be effectively uptaken into human keloids fibroblast (HKFs) in a time-dependent manner. In vitro, PTXL showed better ability on inhibiting cell proliferation, migration and invasion, and effectively on promoting apoptosis and arresting cell cycle in G(2)/M phase compared to PTX. Meanwhile, in vivo studies indicated that the PTXL had better performance on inhibiting the keloids growth compared to the PTX in keloid-bearing BALB/c nude mice model. Finally, we found PTX treatment suppressed the production of tumor necrosis factor alpah (TNF-α), interleukin 6 (IL-6) and transforming growth factor beta (TGF-β) and inhibited the expression of alpha smooth muscle actin (α-SMA) and collagen I in HKFs. The activation of protein kinase B (AKT)/glycogen synthase kinase 3 beta (GSK3β) signaling pathway also blocked by PTX in cultured HKFs and keloid tissues. LY294002, a PI3K (phosphatidylinositol 3-kinase)/AKT inhibitor, also suppressed the expression of TNF-α, IL-6 and TGF-β, and simultaneously, reduced the production of α-SMA and collagen I in HKFs. The inhibition of AKT/GSK3β signaling pathway contribute to inhibit the generation of fibrogenic cytokines by PTXL on ameliorating fibrosis progress in keloids. CONCLUSION: Our results suggested that the developed PTXL would become a promising therapeutic agent in the field of anti-keloid therapy.
format Online
Article
Text
id pubmed-6390862
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Dove Medical Press
record_format MEDLINE/PubMed
spelling pubmed-63908622019-03-12 Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes Wang, Mengjiao Chen, Liqing Huang, Wei Jin, Mingji Wang, Qiming Gao, Zhonggao Jin, Zhehu Int J Nanomedicine Original Research BACKGROUND: Keloids represent benign fibroproliferative tumors which result from elevated expression of inflammation. Paclitaxel (PTX) was an effective chemotherapeutic agent and has been reported to have anti-fibrotic effects, but the strong hydrophobicity brings a challenge for its clinical application. PURPOSE: The objective of this study was to improve the water solubility of PTX and investigate its anti-keloid effects. METHODS: We prepared a PTX-cholesterol-loaded liposomes (PTXL) by thin film evaporation fashion and characterized their physicochemical properties. We also investigated the effects of PTX on proliferation, invasion and fibrosis of keloid fibroblasts in vitro and in vivo. RESULTS: The prepared PTXL have a spherical appearance, a particle size of 101.43 nm and a zeta potential of −41.63 mV. PTXL possessed a high drug entrapment efficiency of 95.63% and exhibited a good stability within 30 days. The drugs in PTXL were released in a slow and sustained mode. The PTXL could be effectively uptaken into human keloids fibroblast (HKFs) in a time-dependent manner. In vitro, PTXL showed better ability on inhibiting cell proliferation, migration and invasion, and effectively on promoting apoptosis and arresting cell cycle in G(2)/M phase compared to PTX. Meanwhile, in vivo studies indicated that the PTXL had better performance on inhibiting the keloids growth compared to the PTX in keloid-bearing BALB/c nude mice model. Finally, we found PTX treatment suppressed the production of tumor necrosis factor alpah (TNF-α), interleukin 6 (IL-6) and transforming growth factor beta (TGF-β) and inhibited the expression of alpha smooth muscle actin (α-SMA) and collagen I in HKFs. The activation of protein kinase B (AKT)/glycogen synthase kinase 3 beta (GSK3β) signaling pathway also blocked by PTX in cultured HKFs and keloid tissues. LY294002, a PI3K (phosphatidylinositol 3-kinase)/AKT inhibitor, also suppressed the expression of TNF-α, IL-6 and TGF-β, and simultaneously, reduced the production of α-SMA and collagen I in HKFs. The inhibition of AKT/GSK3β signaling pathway contribute to inhibit the generation of fibrogenic cytokines by PTXL on ameliorating fibrosis progress in keloids. CONCLUSION: Our results suggested that the developed PTXL would become a promising therapeutic agent in the field of anti-keloid therapy. Dove Medical Press 2019-02-21 /pmc/articles/PMC6390862/ /pubmed/30863067 Text en © 2019 Wang et al. This work is published and licensed by Dove Medical Press Limited The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed.
spellingShingle Original Research
Wang, Mengjiao
Chen, Liqing
Huang, Wei
Jin, Mingji
Wang, Qiming
Gao, Zhonggao
Jin, Zhehu
Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title_full Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title_fullStr Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title_full_unstemmed Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title_short Improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
title_sort improving the anti-keloid outcomes through liposomes loading paclitaxel–cholesterol complexes
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6390862/
https://www.ncbi.nlm.nih.gov/pubmed/30863067
work_keys_str_mv AT wangmengjiao improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT chenliqing improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT huangwei improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT jinmingji improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT wangqiming improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT gaozhonggao improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes
AT jinzhehu improvingtheantikeloidoutcomesthroughliposomesloadingpaclitaxelcholesterolcomplexes