Volume 38 Issue 7
Jul.  2022
Turn off MathJax
Article Contents
Sun LX,Wu S,Zhang XW,et al.Investigation on the growth factor regulatory network of dermal fibroblasts in mouse full-thickness skin defect wounds based on single-cell RNA sequencing[J].Chin J Burns Wounds,2022,38(7):629-639.DOI: 10.3760/cma.j.cn501225-20220215-00029.
Citation: Sun LX,Wu S,Zhang XW,et al.Investigation on the growth factor regulatory network of dermal fibroblasts in mouse full-thickness skin defect wounds based on single-cell RNA sequencing[J].Chin J Burns Wounds,2022,38(7):629-639.DOI: 10.3760/cma.j.cn501225-20220215-00029.

Investigation on the growth factor regulatory network of dermal fibroblasts in mouse full-thickness skin defect wounds based on single-cell RNA sequencing

doi: 10.3760/cma.j.cn501225-20220215-00029
Funds:

National Key Research and Development Program of China 2020YFA0112901

General Program of National Natural Science Foundation of China 81971551

Youth Science Fund Project of National Natural Science Foundation of China 82103702

China Postdoctoral Science Foundation 2020M682095

More Information
  •   Objective  To explore the heterogeneity and growth factor regulatory network of dermal fibroblasts (dFbs) in mouse full-thickness skin defect wounds based on single-cell RNA sequencing.  Methods  The experimental research methods were adopted. The normal skin tissue from 5 healthy 8-week-old male C57BL/6 mice (the same mouse age, sex, and strain below) was harvested, and the wound tissue of another 5 mice with full-thickness skin defect on the back was harvested on post injury day (PID) 7. The cell suspension was obtained by digesting the tissue with collagenase D and DNase Ⅰ, sequencing library was constructed using 10x Genomics platform, and single-cell RNA sequencing was performed by Illumina Novaseq6000 sequencer. The gene expression matrices of cells in the two kinds of tissue were obtained by analysis of Seurat 3.0 program of software R4.1.1, and two-dimensional tSNE plots classified by cell group, cell source, and gene labeling of major cells in skin were used for visual display. According to the existing literature and the CellMarker database searching, the expression of marker genes in the gene expression matrices of cells in the two kinds of tissue was analyzed, and each cell group was numbered and defined. The gene expression matrices and cell clustering information were introduced into CellChat 1.1.3 program of software R4.1.1 to analyze the intercellular communication in the two kinds of tissue and the intercellular communication involving vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), epidermal growth factor (EGF), and fibroblast growth factor (FGF) signal pathways in the wound tissue, the relative contribution of each pair of FGF subtypes and FGF receptor (FGFR) subtypes (hereinafter referred to as FGF ligand receptor pairs) to FGF signal network in the two kinds of tissue, and the intercellular communication in the signal pathway of FGF ligand receptor pairs with the top 2 relative contributions in the two kinds of tissue. The normal skin tissue from one healthy mouse was harvested, and the wound tissue of one mouse with full-thickness skin defect on the back was harvested on PID 7. The multiple immunofluorescence staining was performed to detect the expression and distribution of FGF7 protein and its co-localized expression with dipeptidyl peptidase 4 (DPP4), stem cell antigen 1 (SCA1), smooth muscle actin (SMA), and PDGF receptor α (PDGFRα) protein.  Results  Both the normal skin tissue of healthy mice and the wound tissue of full-thickness skin defected mice on PID 7 contained 25 cell groups, but the numbers of cells in each cell group between the two kinds of tissue were different. Genes PDGFRα, platelet endothelial cell adhesion molecule 1, lymphatic endothelial hyaluronic acid receptor 1, receptor protein tyrosine phosphatase C, keratin 10, and keratin 79 all had distinct distributions on two-dimensional tSNE plots, indicating specific cell groups respectively. The 25 cell groups were numbered by C0-C24 and divided into 9 dFb subgroups and 16 non-dFb groups. dFb subgroups included C0 as interstitial progenitor cells, C5 as adipose precursor cells, and C13 as contractile muscle cells related fibroblasts, etc. Non-dFb group included C3 as neutrophils, C8 as T cells, and C18 as erythrocytes, etc. Compared with that of the normal skin tissue of healthy mice, the intercellular communication in the wound tissue of full-thickness skin defected mice on PID 7 was more and denser, and the top 3 cell groups in intercellular communication intensity were dFb subgroups C0, C1, and C2, of which all communicated with other cell groups in the wound tissue. In the wound tissue of full-thickness skin defected mice on PID 7, VEGF signals were mainly sent by the dFb subgroup C0 and received by vascular related cell groups C19 and C21, PDGF signals were mainly sent by peripheral cells C14 and received by multiple dFb subgroups, EGF signals were mainly sent by keratinocyte subgroups C9 and C11 and received by the dFb subgroup C0, and the main sender and receiver of FGF signals were the dFb subgroup C6. In the relative contribution rank of FGF ligand receptor pairs to FGF signal network in the normal skin tissue of healthy mice and the wound tissue of full-thickness skin defected mice on PID 7, FGF7-FGFR1 was the top 1, and FGF7-FGFR2 or FGF10-FGFR1 was in the second place, respectively; compared with those in the normal skin tissue, there was more intercellular communication in FGF7-FGFR1 signal pathway, while the intercellular communication in FGF7-FGFR2 and FGF10-FGFR1 signal pathways decreased slightly or did not change significantly in the wound tissue; the intercellular communication in FGF7-FGFR1 signal pathway in the wound tissue was stronger than that in FGF7-FGFR2 or FGF10-FGFR1 signal pathway; in the two kinds of tissue, FGF7 signal was mainly sent by dFb subgroups C0, C1, and C2, and received by dFb subgroups C6 and C7. Compared with that in the normal skin tissue of healthy mouse, the expression of FGF7 protein was higher in the wound tissue of full-thickness skin defected mouse on PID 7; in the normal skin tissue, FGF7 protein was mainly expressed in the skin interstitium and also expressed in the white adipose tissue near the dermis layer; in the two kinds of tissue, FGF7 protein was co-localized with DPP4 and SCA1 proteins and expressed in the skin interstitium, co-localized with PDGFRα protein and expressed in dFbs, but was not co-localized with SMA protein, with more co-localized expression of FGF7 in the wound tissue than that in the normal skin tissue.  Conclusions  In the process of wound healing of mouse full-thickness skin defect wound, dFbs are highly heterogeneous, act as potential major secretory or receiving cell populations of a variety of growth factors, and have a close and complex relationship with the growth factor signal pathways. FGF7-FGFR1 signal pathway is the main FGF signal pathway in the process of wound healing, which targets and regulates multiple dFb subgroups.

     

  • loading
  • [1]
    GalloRL,HooperLV.Epithelial antimicrobial defence of the skin and intestine[J].Nat Rev Immunol,2012,12(7):503-516.DOI: 10.1038/nri3228.
    [2]
    Cañedo-DorantesL,Cañedo-AyalaM.Skin acute wound healing: a comprehensive review[J].Int J Inflam,2019,2019:3706315.DOI: 10.1155/2019/3706315.
    [3]
    SorgH,TilkornDJ,HagerS,et al.Skin wound healing: an update on the current knowledge and concepts[J].Eur Surg Res,2017,58(1/2):81-94.DOI: 10.1159/000454919.
    [4]
    GrennanD.Diabetic foot ulcers[J].JAMA,2019,321(1):114.DOI: 10.1001/jama.2018.18323.
    [5]
    HanG,CeilleyR.Chronic wound healing: a review of current management and treatments[J].Adv Ther,2017,34(3):599-610.DOI: 10.1007/s12325-017-0478-y.
    [6]
    AlsterTS,TanziEL.Hypertrophic scars and keloids: etiology and management[J].Am J Clin Dermatol,2003,4(4):235-243.DOI: 10.2165/00128071-200304040-00003.
    [7]
    KischerCW,ThiesAC,ChvapilM.Perivascular myofibroblasts and microvascular occlusion in hypertrophic scars and keloids[J].Hum Pathol,1982,13(9):819-824.DOI: 10.1016/s0046-8177(82)80078-6.
    [8]
    BarrientosS,StojadinovicO,GolinkoMS,et al.Growth factors and cytokines in wound healing[J].Wound Repair Regen,2008,16(5):585-601.DOI: 10.1111/j.1524-475X.2008.00410.x.
    [9]
    AmiriN,GolinAP,JaliliRB,et al.Roles of cutaneous cell-cell communication in wound healing outcome: an emphasis on keratinocyte-fibroblast crosstalk[J].Exp Dermatol,2022,31(4):475-484.DOI: 10.1111/exd.14516.
    [10]
    HanCM,ChengB,WuP.Clinical guideline on topical growth factors for skin wounds[J/OL].Burns Trauma,2020,8:tkaa035[2022-02-15].https://pubmed.ncbi.nlm.nih.gov/33015207/.DOI: 10.1093/burnst/tkaa035.
    [11]
    ZubairM,AhmadJ.Role of growth factors and cytokines in diabetic foot ulcer healing: a detailed review[J].Rev Endocr Metab Disord,2019,20(2):207-217.DOI: 10.1007/s11154-019-09492-1.
    [12]
    WeiY,LiJ,HuangY,et al.The clinical effectiveness and safety of using epidermal growth factor, fibroblast growth factor and granulocyte-macrophage colony stimulating factor as therapeutics in acute skin wound healing: a systematic review and meta-analysis[J/OL].Burns Trauma,2022,10:tkac002[2022-02-15]. https://pubmed.ncbi.nlm.nih.gov/35265723/.DOI: 10.1093/burnst/tkac002.
    [13]
    ChenK,RaoZ,DongS,et al.Roles of the fibroblast growth factor signal transduction system in tissue injury repair[J/OL].Burns Trauma,2022,10:tkac005[2022-02-15]. https://pubmed.ncbi.nlm.nih.gov/35350443/.DOI: 10.1093/burnst/tkac005.
    [14]
    LiuY,LiuY,DengJ,et al.Fibroblast growth factor in diabetic foot ulcer: progress and therapeutic prospects[J].Front Endocrinol (Lausanne),2021,12:744868.DOI: 10.3389/fendo.2021.744868.
    [15]
    HuiQ,JinZ,LiX,et al.FGF family: from drug development to clinical application[J].Int J Mol Sci,2018,19(7):1875.DOI: 10.3390/ijms19071875.
    [16]
    KrookMA,ReeserJW,ErnstG,et al.Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance[J].Br J Cancer,2021,124(5):880-892.DOI: 10.1038/s41416-020-01157-0.
    [17]
    TracyLE,MinasianRA,CatersonEJ.Extracellular matrix and dermal fibroblast function in the healing wound[J].Adv Wound Care (New Rochelle),2016,5(3):119-136.DOI: 10.1089/wound.2014.0561.
    [18]
    desJardins-ParkHE,FosterDS,LongakerMT.Fibroblasts and wound healing: an update[J].Regen Med,2018,13(5):491-495.DOI: 10.2217/rme-2018-0073.
    [19]
    DriskellRR,LichtenbergerBM,HosteE,et al.Distinct fibroblast lineages determine dermal architecture in skin development and repair[J].Nature,2013,504(7479):277-281.DOI: 10.1038/nature12783.
    [20]
    TabibT,MorseC,WangT,et al.SFRP2/DPP4 and FMO1/LSP1 define major fibroblast populations in human skin[J].J Invest Dermatol,2018,138(4):802-810.DOI: 10.1016/j.jid.2017.09.045.
    [21]
    ZhangZ,ShaoM,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.
    [22]
    HaenselD,JinS,SunP,et al.Defining epidermal basal cell states during skin homeostasis and wound healing using single-cell transcriptomics[J].Cell Rep,2020,30(11):3932-3947.e6.DOI: 10.1016/j.celrep.2020.02.091.
    [23]
    Guerrero-JuarezCF,DedhiaPH,JinS,et al.Single-cell analysis reveals fibroblast heterogeneity and myeloid-derived adipocyte progenitors in murine skin wounds[J].Nat Commun,2019,10(1):650.DOI: 10.1038/s41467-018-08247-x.
    [24]
    ZhangLJ,ChenSX,Guerrero-JuarezCF,et al.Age-related loss of innate immune antimicrobial function of dermal fat is mediated by transforming growth factor beta[J].Immunity,2019,50(1):121-136.e5.DOI: 10.1016/j.immuni.2018.11.003.
    [25]
    ZhangX,LanY,XuJ,et al.CellMarker: a manually curated resource of cell markers in human and mouse[J].Nucleic Acids Res,2019,47(D1):D721-D728.DOI: 10.1093/nar/gky900.
    [26]
    JinS,Guerrero-JuarezCF,ZhangL,et al.Inference and analysis of cell-cell communication using CellChat[J].Nat Commun,2021,12(1):1088.DOI: 10.1038/s41467-021-21246-9.
    [27]
    MerrickD,SakersA,IrgebayZ,et al.Identification of a mesenchymal progenitor cell hierarchy in adipose tissue[J].Science,2019,364(6438):eaav2501.DOI: 10.1126/science.aav2501.
    [28]
    Berry-KilgourC,CabralJ,WiseL.Advancements in the delivery of growth factors and cytokines for the treatment of cutaneous wound indications[J].Adv Wound Care (New Rochelle),2021,10(11):596-622.DOI: 10.1089/wound.2020.1183.
    [29]
    YamakawaS,HayashidaK.Advances in surgical applications of growth factors for wound healing[J/OL].Burns Trauma,2019,7:10[2022-02-15].https://pubmed.ncbi.nlm.nih.gov/30993143/.DOI: 10.1186/s41038-019-0148-1.
    [30]
    MaddalunoL,UrwylerC,WernerS.Fibroblast growth factors: key players in regeneration and tissue repair[J].Development,2017,144(22):4047-4060.DOI: 10.1242/dev.152587.
    [31]
    XieY,SuN,YangJ,et al.FGF/FGFR signaling in health and disease[J].Signal Transduct Target Ther,2020,5(1):181.DOI: 10.1038/s41392-020-00222-7.
    [32]
    LindnerV,MajackRA,ReidyMA.Basic fibroblast growth factor stimulates endothelial regrowth and proliferation in denuded arteries[J].J Clin Invest,1990,85(6):2004-2008.DOI: 10.1172/JCI114665.
    [33]
    TaiAL,ShamJS,XieD,et al.Co-overexpression of fibroblast growth factor 3 and epidermal growth factor receptor is correlated with the development of nonsmall cell lung carcinoma[J].Cancer,2006,106(1):146-155.DOI: 10.1002/cncr.21581.
    [34]
    HuL,ShamJS,XieD,et al.Up-regulation of fibroblast growth factor 3 is associated with tumor metastasis and recurrence in human hepatocellular carcinoma[J].Cancer Lett,2007,252(1):36-42.DOI: 10.1016/j.canlet.2006.12.003.
    [35]
    YenTT,ThaoDT,ThuocTL.An overview on keratinocyte growth factor: from the molecular properties to clinical applications[J].Protein Pept Lett,2014,21(3):306-317.DOI: 10.2174/09298665113206660115.
    [36]
    WernerS,KriegT,SmolaH.Keratinocyte-fibroblast interactions in wound healing[J].J Invest Dermatol,2007,127(5):998-1008.DOI: 10.1038/sj.jid.5700786.
    [37]
    ZinkleA,MohammadiM.Structural bology of the FGF7 subfamily[J].Front Genet,2019,10:102.DOI: 10.3389/fgene.2019.00102.
  • 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(10)

    Article Metrics

    Article views (3177) PDF downloads(55) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return