Volume 38 Issue 10
Oct.  2022
Turn off MathJax
Article Contents
Zhu M,Chen YZ,Ou JZ,et al.Effects and mechanism of water-soluble chitosan hydrogel on infected full-thickness skin defect wounds in diabetic mice[J].Chin J Burns Wounds,2022,38(10):923-931.DOI: 10.3760/cma.j.cn501225-20220507-00175.
Citation: Zhu M,Chen YZ,Ou JZ,et al.Effects and mechanism of water-soluble chitosan hydrogel on infected full-thickness skin defect wounds in diabetic mice[J].Chin J Burns Wounds,2022,38(10):923-931.DOI: 10.3760/cma.j.cn501225-20220507-00175.

Effects and mechanism of water-soluble chitosan hydrogel on infected full-thickness skin defect wounds in diabetic mice

doi: 10.3760/cma.j.cn501225-20220507-00175
Funds:

General Program of National Natural Science Foundation of China 52073293

Innovation Special Foundation for High Level Research Institutions in Zhongshan 2019AG003

More Information
  •   Objective  To explore the effects and mechanism of water-soluble chitosan hydrogel on infected full-thickness skin defect wounds in diabetic mice.  Methods  The experimental research method was adopted. The control hydrogel composed of polyvinyl alcohol and gelatin, and the water-soluble chitosan hydrogel composed of the aforementioned two materials and water-soluble chitosan were prepared by the cyclic freeze-thaw method. The fluidity of the two dressings in test tube before and after the first freeze-thawing was generally observed, and the difference in appearance of the final state of two dressings in 12-well plates were compared. According to random number table (the same grouping method below), the cell strains of L929 and HaCaT were both divided into water-soluble chitosan hydrogel group and control hydrogel group, respectively. After adding corresponding dressings and culturing for 24 h, the cell proliferation activity was measured using cell counting kit 8. Rabbit blood erythrocyte suspensions were divided into normal saline group, polyethylene glycol octyl phenyl ether (Triton X-100) group, water-soluble chitosan hydrogel group, and control hydrogel group, which were treated accordingly and incubated for 1 hour, and then the hemolysis degree of erythrocyte was detected by a microplate reader. Twenty-four female db/db mice aged 11-14 weeks were selected, and full-thickness skin defect wounds on their backs were inflicted and inoculated with the methicillin-resistant Staphylococcus aureus (MRSA), 72 h later, the mice were divided into blank control group, sulfadiazine silver hydrogel group, control hydrogel group, and water-soluble chitosan hydrogel group, which were treated accordingly. On post injury day (PID) 0 (immediately), 7, 14, and 21, the healing of the wound was observed. On PID 14 and 21, the wound healing rate was calculated. On PID 14, MRSA concentration in wounds was determined. On PID 21, the wounds were histologically analyzed by hematoxylin and eosin staining; the expression of CD31 in the wounds was detected by immunofluorescence method, and its positive percentage was calculated. Raw264.7 cells were taken and divided into interleukin-4 (IL-4) group, blank control group, control hydrogel group, and water-soluble chitosan hydrogel group, which were treated accordingly. At 48 h of culture, the percentages of CD206 positive cells were detected by flow cytometry. The number of samples was all 3. Data were statistically analyzed with independent sample t test, one-way analysis of variance, analysis of variance for repeated measurement, least significant difference test, and Dunnett T3 test.  Results  Two dressings in test tube had certain fluidity before freeze-thawing and formed semi-solid gels after freeze-thawing for once. The final forms of two dressings in 12-well plates were basically stable and translucent sheets, with little difference in transparency. At 24 h of culture, the cell proliferation activities of L929 and HaCaT in water-soluble chitosan hydrogel group were significantly higher than those in control hydrogel group (with t values of 6.37 and 7.50, respectively, P<0.01). At 1 h of incubation, the hemolysis degree of erythrocyte in water-soluble chitosan hydrogel group was significantly lower than that in Triton X-100 group (P<0.01), but similar to that in normal saline group and control hydrogel group (P>0.05). On PID 0, the traumatic conditions of mice in the 4 groups were similar. On PID 7, more yellowish exudates were observed inside the wound in blank control group and control hydrogel group, while a small amount of exudates were observed in the wound in sulfadiazine silver hydrogel group and water-soluble chitosan hydrogel group. On PID 14, the wounds in blank control group and control hydrogel group were dry and crusted without obvious epithelial coverage; in sulfadiazine silver hydrogel group, the scabs fell off and purulent exudate was visible on the wound; in water-soluble chitosan hydrogel group, the base of wound was light red and obvious epithelial coverage could be observed on the wound. On PID 14, the wound healing rate in water-soluble chitosan hydrogel group was significantly higher than that in the other 3 groups (all P<0.01). On PID 21, the wound in water-soluble chitosan hydrogel group was completely closed, while the wounds in the other 3 groups were not completely healed; the wound healing rate in water-soluble chitosan hydrogel group was significantly higher than that in the other 3 groups (all P<0.01). On PID 14, the concentration of MRSA in the wound in water-soluble chitosan hydrogel group was significantly lower than that in blank control group (P<0.01), but similar to that in control hydrogel group and sulfadiazine silver hydrogel group (P>0.05). On PID 21, the new epidermis was severely damaged in blank control group; the epidermis on the wound in control hydrogel group also had a large area of defect; complete new epidermis had not yet being formed on the wound in sulfadiazine silver hydrogel group; the wound in water-soluble chitosan hydrogel group was not only completely covered by the new epidermis, the basal cells of the new epidermis were also regularly aligned. On PID 21, the percentage of CD31 positivity in the wound in water-soluble chitosan hydrogel group was (2.19±0.35)%, which was significantly higher than (0.18±0.05)% in blank control group, (0.23±0.06)% in control hydrogel group, and (0.62±0.25)% in sulfadiazine silver hydrogel group, all P<0.01. At 48 h of culture, the percentage of CD206 positive Raw264.7 cells in water-soluble chitosan hydrogel group was lower than that in IL-4 group (P>0.01) but significantly higher than that in blank control group and control hydrogel group (P<0.05 or P<0.01).  Conclusions  The water-soluble chitosan hydrogel has good biosafety and can induce higher level of macrophage M2 polarization than control hydrogel without water-soluble chitosan, so it can enhance the repair effect of MRSA-infected full-thickness skin defect wounds in diabetic mice and promote rapid wound healing.

     

  • loading
  • [1]
    KrzyszczykP, SchlossR, PalmerA, et al. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes[J]. Front Physiol, 2018,9:419. DOI: 10.3389/fphys.2018.00419.
    [2]
    PlikusMV, Guerrero-JuarezCF, ItoM, et al. Regeneration of fat cells from myofibroblasts during wound healing[J]. Science, 2017,355(6326):748-752. DOI: 10.1126/science.aai8792.
    [3]
    MalikVS, WillettWC, HuFB. Global obesity: trends, risk factors and policy implications[J]. Nat Rev Endocrinol, 2013,9(1):13-27. DOI: 10.1038/nrendo.2012.199.
    [4]
    ZimmetP, AlbertiKG, MaglianoDJ, et al. Diabetes mellitus statistics on prevalence and mortality: facts and fallacies[J]. Nat Rev Endocrinol, 2016,12(10):616-622. DOI: 10.1038/nrendo.2016.105.
    [5]
    EmingSA, MartinP, Tomic-CanicM. Wound repair and regeneration: mechanisms, signaling, and translation[J]. Sci Transl Med, 2014,6(265):265sr6. DOI: 10.1126/scitranslmed.3009337.
    [6]
    WolfSJ, MelvinWJ, GallagherK. Macrophage-mediated inflammation in diabetic wound repair[J]. Semin Cell Dev Biol, 2021,119:111-118. DOI: 10.1016/j.semcdb.2021.06.013.
    [7]
    VasconcelosDP, CostaM, AmaralIF, et al. Modulation of the inflammatory response to chitosan through M2 macrophage polarization using pro-resolution mediators[J]. Biomaterials, 2015,37:116-123. DOI: 10.1016/j.biomaterials.2014.10.035.
    [8]
    VasconcelosDP, de Torre-MinguelaC, GomezAI, et al. 3D chitosan scaffolds impair NLRP3 inflammasome response in macrophages[J]. Acta Biomater, 2019,91:123-134. DOI: 10.1016/j.actbio.2019.04.035.
    [9]
    CheungRC, NgTB, WongJH, et al. Chitosan: an update on potential biomedical and pharmaceutical applications[J]. Mar Drugs, 2015,13(8):5156-5186. DOI: 10.3390/md13085156.
    [10]
    DashM, ChielliniF, OttenbriteRM. Chitosan-a versatile semi-synthetic polymer in biomedical applications[J]. Prog Polym Sci, 2011, 36(8):981-1014. DOI: 10.1016/j.progpolymsci.2011.02.001.
    [11]
    JeongYI, KimDG, JangMK, et al. Preparation and spectroscopic characterization of methoxy poly(ethylene glycol)-grafted water-soluble chitosan[J]. Carbohydr Res, 2008,343(2):282-289. DOI: 10.1016/j.carres.2007.10.025.
    [12]
    YangC, GaoS, Dagnæs-HansenF, et al. Impact of PEG chain length on the physical properties and bioactivity of PEGylated chitosan/siRNA nanoparticles in vitro and in vivo[J]. ACS Appl Mater Interfaces, 2017,9(14):12203-12216. DOI: 10.1021/acsami.6b16556.
    [13]
    ZhuM, LiuP, ShiH, et al. Balancing antimicrobial activity with biological safety: bifunctional chitosan derivative for the repair of wounds with Gram-positive bacterial infections[J]. J Mater Chem B, 2018,6(23):3884-3893. DOI: 10.1039/c8tb00620b.
    [14]
    LuH, YuanL, YuX, et al. Recent advances of on-demand dissolution of hydrogel dressings[J/OL]. Burns Trauma, 2018,6:35[2022-05-07]. https://pubmed.ncbi.nlm.nih.gov/30619904/. DOI: 10.1186/s41038-018-0138-8.
    [15]
    ThuHE, ZulfakarMH, NgSF. Alginate based bilayer hydrocolloid films as potential slow-release modern wound dressing[J]. Int J Pharm, 2012,434(1/2):375-383. DOI: 10.1016/j.ijpharm.2012.05.044.
    [16]
    FakhariA, BerklandC. Applications and emerging trends of hyaluronic acid in tissue engineering, as a dermal filler and in osteoarthritis treatment[J]. Acta Biomater, 2013,9(7):7081-7092. DOI: 10.1016/j.actbio.2013.03.005.
    [17]
    LiC, YeR, BouckaertJ, et al. Flexible nanoholey patches for antibiotic-free treatments of skin infections[J]. ACS Appl Mater Interfaces, 2017,9(42):36665-36674. DOI: 10.1021/acsami.7b12949.
    [18]
    GurtnerGC, WernerS, BarrandonY, et al. Wound repair and regeneration[J]. Nature, 2008,453(7193):314-321. DOI: 10.1038/nature07039.
    [19]
    PowersJG, HighamC, BroussardK, et al. Wound healing and treating wounds: chronic wound care and management[J]. J Am Acad Dermatol, 2016,74(4):607-625; quiz 625-626. DOI: 10.1016/j.jaad.2015.08.070.
    [20]
    MohseniM, ShamlooA, AghababaeiZ, et al. Antimicrobial wound dressing containing silver sulfadiazine with high biocompatibility: in vitro study[J]. Artif Organs, 2016,40(8):765-773. DOI: 10.1111/aor.12682.
    [21]
    MinJH, PatelM, KohWG. Incorporation of conductive materials into hydrogels for tissue engineering applications[J]. Polymers (Basel), 2018,10(10):1078. DOI: 10.3390/polym10101078.
    [22]
    HomaeigoharS, BoccacciniAR. Antibacterial biohybrid nanofibers for wound dressings[J]. Acta Biomater, 2020,107:25-49. DOI: 10.1016/j.actbio.2020.02.022.
    [23]
    HuT, CuiX, ZhuM, et al. 3D-printable supramolecular hydrogels with shear-thinning property: fabricating strength tunable bioink via dual crosslinking[J]. Bioact Mater, 2020,5(4):808-818. DOI: 10.1016/j.bioactmat.2020.06.001.
    [24]
    沈括, 王许杰, 刘开拓, 等. 人脂肪间充质干细胞外泌体对小鼠RAW264.7细胞的炎症反应和小鼠全层皮肤缺损创面愈合的影响[J].中华烧伤与创面修复杂志,2022,38(3):215-226.DOI: 10.3760/cma.j.cn501120-20201116-00477.
    [25]
    KharazihaM, BaidyaA, AnnabiN. Rational design of immunomodulatory hydrogels for chronic wound healing[J]. Adv Mater, 2021,33(39):e2100176. DOI: 10.1002/adma.202100176.
    [26]
    PeppasNA, StaufferSR. Reinforced uncrosslinked poly (vinyl alcohol) gels produced by cyclic freezing-thawing processes-a short review[J].J Control Release, 1991, 16(3): 305-310. DOI: 10.1016/0168-3659(91)90007-z.
    [27]
    KitaM, OguraY, HondaY, et al. Evaluation of polyvinyl alcohol hydrogel as a soft contact lens material[J]. Graefes Arch Clin Exp Ophthalmol, 1990,228(6):533-537. DOI: 10.1007/BF00918486.
    [28]
    XuN, YuanY, DingL, et al. Multifunctional chitosan/gelatin@tannic acid cryogels decorated with in situ reduced silver nanoparticles for wound healing[J/OL]. Burns Trauma, 2022,10:tkac019[2022-05-09]. https://pubmed.ncbi.nlm.nih.gov/35910193/. DOI: 10.1093/burnst/tkac019.
    [29]
    ShamlooA, AghababaieZ, AfjoulH, et al. Fabrication and evaluation of chitosan/gelatin/PVA hydrogel incorporating honey for wound healing applications: an in vitro, in vivo study[J]. Int J Pharm, 2021,592:120068. DOI: 10.1016/j.ijpharm.2020.120068.
    [30]
    BoniakowskiAE, KimballAS, JacobsBN, et al. Macrophage-mediated inflammation in normal and diabetic wound healing[J]. J Immunol, 2017,199(1):17-24. DOI: 10.4049/jimmunol.1700223.
    [31]
    SebastianW, Sanin DavidE, AlexanderJ, et al. Mitochondrial metabolism coordinates stage-specific repair processes in macrophages during wound healing[J]. Cell Metabolism, 2021,33(12):2398-2414.e9. DOI: 10.1016/j.cmet.2021.10.004.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(5)  / Tables(1)

    Article Metrics

    Article views (3890) PDF downloads(60) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return