Volume 37 Issue 11
Nov.  2021
Turn off MathJax
Article Contents
Zhang C,Li Z,Song W,et al.Preliminary investigation on the wound healing effect of three-dimensional bioprinting ink containing human adipose-derived protein complexes[J].Chin J Burns,2021,37(11):1011-1023.DOI: 10.3760/cma.j.cn501120-20210813-00282.
Citation: Zhang C,Li Z,Song W,et al.Preliminary investigation on the wound healing effect of three-dimensional bioprinting ink containing human adipose-derived protein complexes[J].Chin J Burns,2021,37(11):1011-1023.DOI: 10.3760/cma.j.cn501120-20210813-00282.

Preliminary investigation on the wound healing effect of three-dimensional bioprinting ink containing human adipose-derived protein complexes

doi: 10.3760/cma.j.cn501120-20210813-00282
Funds:

Science Fund for Creative Research Groups of National Natural Science Foundation of China 81721092

Key Program of National Natural Science Foundation of China 81830064

Youth Science Foundation Project of National Natural Science Foundation of China 32000969, 82002056

Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences 2019-I2M-5-059

Military Medical Innovation Research Project of PLA General Hospital CX19026

Wang Zhengguo Foundation for Traumatic Medicine Growth Factor Rejuvenation Plan SZYZ-TR-03

More Information
  • Corresponding author: Huang Sha, Email: stellarahuang@sina.com
  • Received Date: 2021-08-13
  •     Objective   To investigate the effects of human adipose-derived protein complex (ADPC) on the proliferation and migration ability of human skin fibroblasts (HSFs) and human umbilical vein endothelial cells (HUVECs), and the repairing effects of ADPC-containing three-dimensional (3D) bioprinting ink (Bioink) in full-thickness skin defect wounds of nude mice.    Methods   The experimental research method was used. Discarded subcutaneous adipose tissue from 3 female patients with chronic wounds (aged 29-34 years) admitted to PLA General Hospital for abdominal flap transfer from October 2020 to March 2021 and discarded liposuction adipose tissue from 3 healthy female (aged 24-36 years) for abdominal liposuction during the same period were collected to prepare normal ADPC (nADPC) and liposuction-derived ADPC (lADPC), respectively. The protein concentration of the two kinds of ADPC was measured by bicinchoninic acid method, and the extraction efficiency of them was calculated. The sample numbers were 3. HSFs and HUVECs in logarithmic growth phase were taken for the subsequent experiments. HSFs and HUVECs were divided into phosphate buffered saline (PBS) control group, 4 μg/mL nADPC group, 20 μg/mL nADPC group, 100 μg/mL nADPC group, and 200 μg/mL nADPC group according to the random number table (the same grouping method below), with 5 wells in each group. Cells in PBS control group were cultured with PBS, and the cells in the 4 remaining groups were cultured with the corresponding final mass concentration of nADPC. After 24 h of conventional culture, the cell proliferation viability was detected by cell counting kit 8 method. HSFs and HUVECs were taken and divided into PBS control group, nADPC alone group, lADPC alone group, tumor necrosis factor-α (TNF-α) alone group, TNF-α+nADPC group, and TNF-α+lADPC group. Cells in PBS control group and TNF-α alone group were added with PBS. nADPC or lADPC was added to the cells in nADPC alone group, lADPC alone group, TNF-α+nADPC group, and TNF-α+lADPC group with a final mass concentration of 100 μg/mL, respectively. TNF-α with a final mass concentration of 20 ng/mL was added to the cells in TNF-α alone group, TNF-α+nADPC group, and TNF-α+lADPC group. The cell migration rate was calculated after the scratch test at 24 h after scratching (n=3), and the cell proliferation viability was detected after 24 h of culture as above (n=5). Gelatin-alginate composite Bioink (Bioink AG) was taken. Bioink AG containing 100 μg/mL lADPC (lADPC-Bioink AG) was prepared. The morphology of the two at room temperature and after condensation was observed. The morphology after 3D bioprinting and cross-linking was observed. The low-temperature gel formation time was recorded when detecting rheological properties using rheometer (n=3). Twenty BALB/c-NU female nude mice of 8-10 weeks old were taken to establish the full-thickness skin defect wounds on the back, and then they were divided into routine dressing change group, lADPC alone group, Bioink AG alone group, and lADPC-Bioink AG group, with 5 nude mice in each group. The wounds of nude mice in routine dressing change group were covered with hydrocolloid dressings and performed with routine dressing changes only, while the wounds of nude mice in the remaining 3 groups were treated with lADPC, Bioink AG, and lADPC-Bioink AG accordingly in addition. General observation was performed from treatment day (TD) 0, and the wound healing rate was calculated on TD 2, 6, and 10. On TD 10, histopathological observation of wounds was performed with hematoxylin eosin staining. Data were statistically analyzed with independent samples t test, one-way analysis of variance, analysis of variance for repeated measurement, Student-Newman-Keuls q test, and least significant difference t test.    Results   The protein concentration and extraction efficiency of lADPC were respectively (1.306±0.011) mg/mL and (11.1±1.5)%, which were significantly lower than (2.039±0.042) mg/mL and (22.2±2.0)% of nADPC (t=23.83, 6.38, P<0.05 or P<0.01). After 24 h of culture, compared with those in PBS control group, the proliferation viabilities of HSFs (q=6.943, 6.375, P<0.01) and HUVECs (q=6.301, 6.496, P<0.01) were significantly decreased in 100 μg/mL nADPC group and 200 μg/mL nADPC group; compared with those in 100 μg/mL nADPC group, the proliferation viabilities of HSFs and HUVECs in 200 μg/mL nADPC group did not change significantly (P>0.05). At 24 h after scratching, compared with those in PBS control group, the HSF and HUVEC migration rates were significantly lower in nADPC alone group, lADPC alone group, and TNF-α alone group (q=5.642, 6.645, 11.480, 4.772, 6.298, 10.420, P<0.05 or P<0.01); compared with those in nADPC alone group, there were no significant changes in the HSF and HUVEC migration rates in lADPC alone group (P>0.05); compared with those in TNF-α alone group, there were no significant changes in the HSF migration rates in TNF-α+nADPC group or TNF-α+lADPC group (P>0.05), the HUVEC migration rates were significantly higher in TNF-α+nADPC group and TNF-α+lADPC group (q=8.585, 7.253, P<0.01); compared with those in TNF-α+nADPC group, there were no significant changes in the HSF and HUVEC migration rates in TNF-α+lADPC group (P>0.05). After 24 h of culture, compared with those in PBS control group, the HSF and HUVEC proliferation viabilities were significantly lower in nADPC alone group, lADPC alone group, and TNF-α alone group (q=5.803, 5.371, 9.136, 11.580, 9.493, 13.510, P<0.05 or P<0.01); compared with those in nADPC alone group, the HSF and HUVEC proliferation viabilities in lADPC alone group did not change significantly (P>0.05); compared with those in TNF-α alone group, the HSF (q=14.990, 10.850, P<0.01) and HUVEC (q=7.066, 8.942, P<0.01) proliferation viabilities were significantly higher in TNF-α+nADPC group and TNF-α+lADPC group; compared with those in TNF-α+nADPC group, the HSF and HUVEC proliferation viabilities in TNF-α+lADPC group did not change significantly (P>0.05). At room temperature and in the condensed state, lADPC-Bioink AG had a more slightly turbid appearance than Bioink AG. lADPC-Bioink AG had a similar morphology to Bioink AG after 3D bioprinting and cross-linking. At 10 ℃, the coagulation time of lADPC-Bioink AG was (76.6±0.4) s, which was significantly slower than (74.4±0.6) s of Bioink AG (t=4.64, P<0.01). On TD 2, the nude mice in routine dressing change group had more wound exudation, while the nude mice in the remaining 3 groups had no significant exudation. On TD 8, the nude mice in lADPC-Bioink AG group had the smallest residual wound area and obvious epithelial coverage. On TD 2, the wound healing rate of nude mice in lADPC-Bioink AG group was significantly higher than that in lADPC alone group (t=3.59, P<0.05) and similar to the rates in routine dressing change group and Bioink AG alone group (P>0.05). On TD 6, the wound healing rate of nude mice in lADPC-Bioink AG group was significantly higher than the rates in routine dressing change group, lADPC alone group, and Bioink AG alone group (t=18.70, 15.70, 3.15, P<0.05 or P<0.01). On TD 10, the wound healing rate of nude mice in lADPC-Bioink AG group was significantly higher than the rates in routine dressing change group and lADPC alone group (t=12.51, 4.84, P<0.01) but similar to that in Bioink AG alone group (P>0.05). On TD 10, the wounds of nude mice in lADPC-Bioink AG group had moderate vascularization of the traumatic tissue, adequate epithelialization, and the best healing effect.    Conclusions   Liposuction-related operations have little effect on the characterization of ADPC protein concentration and extraction efficiency. LADPC and nADPC have the same biological effects, which can inhibit the proliferation and migration ability of HSFs and HUVECs in non-inflammatory environment and improve the proliferation viabilities of HSFs and HUVECs in inflammatory environment, while improving the migration ability of HUVECs. Adding lADPC to Bioink AG does not significantly affect the physical properties or printing performance of Bioink AG, but can enhance the wound repair effect of full-thickness skin defect wounds in nude mice.

     

  • loading
  • [1]
    MascharakS, desJardins-ParkHE, DavittMF, et al. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring[J].Science,2021,372(6540):eaba2374. DOI: 10.1126/science.aba2374.
    [2]
    ZhangJ, ZhengYJ, LeeJ, et al. A pulsatile release platform based on photo-induced imine-crosslinking hydrogel promotes scarless wound healing[J]. Nat Commun,2021,12(1):1670. DOI: 10.1038/s41467-021-21964-0.
    [3]
    GriffinDR, ArchangMM, KuanCH, et al. Activating an adaptive immune response from a hydrogel scaffold imparts regenerative wound healing[J]. Nat Mater,2021,20(4):560-569. DOI: 10.1038/s41563-020-00844-w.
    [4]
    GriffinDR,WeaverWM,ScumpiaPO, et al. Accelerated wound healing by injectable microporous gel scaffolds assembled from annealed building blocks[J].Nat Mater,2015,14(7):737-744. DOI: 10.1038/nmat4294.
    [5]
    NgWL, WangS, YeongWY, et al. Skin bioprinting: impending reality or fantasy?[J]. Trends Biotechnol,2016,34(9):689-699. DOI: 10.1016/j.tibtech.2016.04.006.
    [6]
    WonJY,LeeMH,KimMJ, et al. A potential dermal substitute using decellularized dermis extracellular matrix derived bio-ink[J].Artif Cells Nanomed Biotechnol,2019,47(1):644-649. DOI: 10.1080/21691401.2019.1575842.
    [7]
    ChimeneD, KaunasR, GaharwarAK. Hydrogel bioink reinforcement for additive manufacturing: a focused review of emerging strategies[J]. Adv Mater,2020,32(1):e1902026. DOI: 10.1002/adma.201902026.
    [8]
    ValotL,MartinezJ,MehdiA, et al. Chemical insights into bioinks for 3D printing[J].Chem Soc Rev,2019,48(15):4049-4086. DOI: 10.1039/c7cs00718c.
    [9]
    SomasekharanLT,RajuR,KumarS, et al. Biofabrication of skin tissue constructs using alginate, gelatin and diethylaminoethyl cellulose bioink[J].Int J Biol Macromol,2021,189:398-409. DOI: 10.1016/j.ijbiomac.2021.08.114.
    [10]
    ChenXF,YueZL,WinbergPC, et al. 3D bioprinting dermal-like structures using species-specific ulvan[J].Biomater Sci,2021,9(7):2424-2438. DOI: 10.1039/d0bm01784a.
    [11]
    ChoudhuryD, TunHW, WangTY, et al. Organ-derived decellularized extracellular matrix: a game changer for bioink manufacturing?[J]. Trends Biotechnol,2018,36(8):787-805. DOI: 10.1016/j.tibtech.2018.03.003.
    [12]
    ShookB, Rivera GonzalezG, EbmeierS, et al. The role of adipocytes in tissue regeneration and stem cell niches[J]. Annu Rev Cell Dev Biol,2016,32:609-631. DOI: 10.1146/annurev-cellbio-111315-125426.
    [13]
    Guerrero-JuarezCF,PlikusMV.Emerging nonmetabolic functions of skin fat[J].Nat Rev Endocrinol,2018,14(3):163-173. DOI: 10.1038/nrendo.2017.162.
    [14]
    ZwickRK, Guerrero-JuarezCF, HorsleyV, et al. Anatomical, physiological, and functional diversity of adipose tissue[J]. Cell Metab,2018,27(1):68-83. DOI: 10.1016/j.cmet.2017.12.002.
    [15]
    ZhangZZ,ShaoML,HeplerC, et al. Dermal adipose tissue has high plasticity and undergoes reversible dedifferentiation in mice[J].J Clin Invest,2019,129(12):5327-5342. DOI: 10.1172/JCI130239.
    [16]
    SarkanenJR,RuusuvuoriP,KuokkanenH, et al. Bioactive acellular implant induces angiogenesis and adipogenesis and sustained soft tissue restoration in vivo[J].Tissue Eng Part A,2012,18(23/24):2568-2580. DOI: 10.1089/ten.TEA.2011.0724.
    [17]
    HeYF,LinMH,WangXC, et al. Optimized adipose tissue engineering strategy based on a neo-mechanical processing method[J].Wound Repair Regen,2018,26(2):163-171. DOI: 10.1111/wrr.12640.
    [18]
    SarkanenJR,KailaV,MannerströmB, et al. Human adipose tissue extract induces angiogenesis and adipogenesis in vitro[J].Tissue Eng Part A,2012,18(1/2):17-25. DOI: 10.1089/ten.TEA.2010.0712.
    [19]
    LiZ, HuangS, LiuYF, et al. Tuning alginate-gelatin bioink properties by varying solvent and their impact on stem cell behavior[J]. Sci Rep,2018,8(1):8020. DOI: 10.1038/s41598-018-26407-3.
    [20]
    LiuYF,LiJJ,YaoB, et al. The stiffness of hydrogel-based bioink impacts mesenchymal stem cells differentiation toward sweat glands in 3D-bioprinted matrix[J].Mater Sci Eng C Mater Biol Appl,2021,118:111387. DOI: 10.1016/j.msec.2020.111387.
    [21]
    WeiLC,LiZ,LiJJ, et al. An approach for mechanical property optimization of cell-laden alginate-gelatin composite bioink with bioactive glass nanoparticles[J].J Mater Sci Mater Med,2020,31(11):103. DOI: 10.1007/s10856-020-06440-3.
    [22]
    ChangM, NguyenTT. Strategy for treatment of infected fiabetic foot ulcers[J]. Acc Chem Res,2021,54(5):1080-1093. DOI: 10.1021/acs.accounts.0c00864.
    [23]
    NinanN,ThomasS,GrohensY.Wound healing in urology[J].Adv Drug Deliv Rev,2015(82/83):93-105. DOI: 10.1016/j.addr.2014.12.002.
    [24]
    WalkerJT,McLeodK,KimS, et al. Periostin as a multifunctional modulator of the wound healing response[J].Cell Tissue Res,2016,365(3):453-465. DOI: 10.1007/s00441-016-2426-6.
    [25]
    ChouhanD,DeyN,BhardwajN, et al. Emerging and innovative approaches for wound healing and skin regeneration: current status and advances[J].Biomaterials,2019,216:119267. DOI: 10.1016/j.biomaterials.2019.119267.
    [26]
    RodriguesM, KosaricN, BonhamCA, et al. Wound healing: a cellular perspective[J]. Physiol Rev,2019,99(1):665-706. DOI: 10.1152/physrev.00067.2017.
    [27]
    DongMW,LiM,ChenJ, et al. Activation of α7nAChR promotes diabetic wound healing by suppressing AGE-induced TNF-α production[J].Inflammation,2016,39(2):687-699. DOI: 10.1007/s10753-015-0295-x.
    [28]
    YenYH,PuCM,LiuCW, et al. Curcumin accelerates cutaneous wound healing via multiple biological actions: the involvement of TNF-α, MMP-9, α-SMA, and collagen[J].Int Wound J,2018,15(4):605-617. DOI: 10.1111/iwj.12904.
    [29]
    ZhouDJ, LiuTF, WangS, et al. Effects of IL-1β and TNF-α on the expression of P311 in vascular endothelial cells and wound healing in mice[J]. Front Physiol,2020,11:545008. DOI: 10.3389/fphys.2020.545008.
    [30]
    GurtnerGC,WernerS,BarrandonY, et al. Wound repair and regeneration[J].Nature,2008,453(7193):314-321. DOI: 10.1038/nature07039.
    [31]
    SpiekmanM,PrzybytE,PlantingaJA, et al. Adipose tissue- derived stromal cells inhibit TGF-β1-induced differentiation of human dermal fibroblasts and keloid scar-derived fibroblasts in a paracrine fashion[J].Plast Reconstr Surg,2014,134(4):699-712. DOI: 10.1097/PRS.0000000000000504.
    [32]
    KruglikovIL, SchererPE. Dermal adipocytes: from irrelevance to metabolic targets?[J]. Trends Endocrinol Metab,2016,27(1):1-10. DOI: 10.1016/j.tem.2015.11.002.
    [33]
    SpiekmanM,van DongenJA,WillemsenJC, et al. The power of fat and its adipose-derived stromal cells: emerging concepts for fibrotic scar treatment[J].J Tissue Eng Regen Med,2017,11(11):3220-3235. DOI: 10.1002/term.2213.
    [34]
    KoleskyDB,TrubyRL,GladmanAS, et al. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs[J].Adv Mater,2014,26(19):3124-3130. DOI: 10.1002/adma.201305506.
    [35]
    NingLQ,GilCJ,HwangB, et al. Biomechanical factors in three-dimensional tissue bioprinting[J].Appl Phys Rev,2020,7(4):041319. DOI: 10.1063/5.0023206.
    [36]
    KimBS,KwonYW,KongJS, et al. 3D cell printing of in vitro stabilized skin model and in vivo pre-vascularized skin patch using tissue-specific extracellular matrix bioink: a step towards advanced skin tissue engineering[J].Biomaterials,2018,168:38-53. DOI: 10.1016/j.biomaterials.2018.03.040.
    [37]
    YaoB,WangR,WangYH, et al. Biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation for functional sweat gland regeneration[J].Sci Adv,2020,6(10):eaaz1094. DOI: 10.1126/sciadv.aaz1094.
    [38]
    WangSF, WangXH, NeufurthM, et al. Biomimetic alginate/gelatin cross-linked hydrogels supplemented with polyphosphate for wound healing applications[J]. Molecules, 2020, 25(21):5210. DOI: 10.3390/molecules25215210.
    [39]
    KaravasiliC,TsongasK,AndreadisII, et al. Physico-mechanical and finite element analysis evaluation of 3D printable alginate- methylcellulose inks for wound healing applications[J].Carbohydr Polym,2020,247:116666. DOI: 10.1016/j.carbpol.2020.116666.
    [40]
    TurnerPR,MurrayE,McAdamCJ, et al. Peptide chitosan/dextran core/shell vascularized 3D constructs for wound healing[J].ACS Appl Mater Interfaces,2020,12(29):32328-32339. DOI: 10.1021/acsami.0c07212.
  • 张超视频解读~1.mp4
  • 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(4)

    Article Metrics

    Article views (2552) PDF downloads(43) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return