Skip to main content

Advertisement

Log in

Genome-wide expression analysis of wounded skin reveals novel genes involved in angiogenesis

  • Original Paper
  • Published:
Angiogenesis Aims and scope Submit manuscript

Abstract

Wound healing is a multistage process involving collaborative efforts of different cell types and distinct cellular functions. Among others, the high metabolic activity at the wound site requires the formation and sprouting of new blood vessels (angiogenesis) to ensure an adequate supply of oxygen and nutrients for a successful healing process. Thus, a cutaneous wound healing model was established to identify new factors that are involved in vascular formation and remodeling in human skin after embryonic development. By analyzing global gene expression of skin biopsies obtained from wounded and unwounded skin, we identified a small set of genes that were highly significant differentially regulated in the course of wound healing. To initially investigate whether these genes might be involved in angiogenesis, we performed siRNA experiments and analyzed the knockdown phenotypes using a scratch wound assay which mimics cell migration and proliferation in vitro. The results revealed that a subset of these genes influence cell migration and proliferation in primary human endothelial cells (EC). Furthermore, histological analyses of skin biopsies showed that two of these genes, ALBIM2 and TMEM121, are colocalized with CD31, a well known EC marker. Taken together, we identified new genes involved in endothelial cell biology, which might be relevant to develop therapeutics not only for impaired wound healing but also for chronic inflammatory disorders and/or cardiovascular diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

References

  1. Adams RH, Alitalo K (2007) Molecular regulation of angiogenesis and lymphangiogenesis. Nat Rev Mol Cell Biol 8(6):464–478

    Article  CAS  PubMed  Google Scholar 

  2. Herbert SP, Stainier DY (2011) Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat Rev Mol Cell Biol 12(9):551–564

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  3. Carmeliet P, Jain RK (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature 473(7347):298–307

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Phng LK, Gerhardt H (2009) Angiogenesis: a team effort coordinated by notch. Dev Cell 16(2):196–208

    Article  CAS  PubMed  Google Scholar 

  5. Gerhardt H (2008) VEGF and endothelial guidance in angiogenic sprouting. Organogenesis 4(4):241–246

    Article  PubMed Central  PubMed  Google Scholar 

  6. Jakobsson L et al (2010) Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat Cell Biol 12(10):943–953

    Article  CAS  PubMed  Google Scholar 

  7. Inai T et al (2004) Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol 165(1):35–52

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Bergers G, Song S (2005) The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol 7(4):452–464

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Armulik A, Abramsson A, Betsholtz C (2005) Endothelial/pericyte interactions. Circ Res 97(6):512–523

    Article  CAS  PubMed  Google Scholar 

  10. Gurtner GC et al (2008) Wound repair and regeneration. Nature 453(7193):314–321

    Article  CAS  PubMed  Google Scholar 

  11. Singer AJ, Clark RA (1999) Cutaneous wound healing. N Engl J Med 341(10):738–746

    Article  CAS  PubMed  Google Scholar 

  12. Freinkel RK (2001) The biology of the skin. Parthenon Publishing Group, New York

  13. Mendonca RJ (2012) Angiogenesis in wound healing. Tissue regeneration—from basic biology to clinical application

  14. Eming SA et al (2007) Regulation of angiogenesis: wound healing as a model. Prog Histochem Cytochem 42(3):115–170

    Article  CAS  PubMed  Google Scholar 

  15. DiPietro LA (2013) Angiogenesis and scar formation in healing wounds. Curr Opin Rheumatol 25(1):87–91

  16. Gronniger E et al (2010) Aging and chronic sun exposure cause distinct epigenetic changes in human skin. PLoS Genet 6(5):e1000971

  17. Schmittgen TD, Livak KJ (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 3(6):1101–1108

    Article  CAS  PubMed  Google Scholar 

  18. Bronneke S et al (2012) DNA methylation regulates lineage-specifying genes in primary lymphatic and blood endothelial cells. Angiogenesis 15(2):317–329

  19. Kiistala U (1968) Suction blister device for separation of viable epidermis from dermis. J Invest Dermatol 50(2):129–137

    CAS  PubMed  Google Scholar 

  20. Kool J et al (2007) Suction blister fluid as potential body fluid for biomarker proteins. Proteomics 7(20):3638–3650

    Article  CAS  PubMed  Google Scholar 

  21. Kanitakis J (2002) Anatomy, histology and immunohistochemistry of normal human skin. Eur J Dermatol 12(4):390–399; quiz 400–401

  22. Fritsch P (2004) Aufbau und Funktionen der Haut. In: Dermatologie und Venerologie. Springer, Berlin. 2. Auflage

  23. Braverman IM (1997) The cutaneous microcirculation: ultrastructure and microanatomical organization. Microcirculation 4(3):329–340

    Article  CAS  PubMed  Google Scholar 

  24. Werner S, Grose R (2003) Regulation of wound healing by growth factors and cytokines. Physiol Rev 83(3):835–870

    CAS  PubMed  Google Scholar 

  25. Nissen NN et al (1998) Vascular endothelial growth factor mediates angiogenic activity during the proliferative phase of wound healing. Am J Pathol 152(6):1445–1452

    CAS  PubMed Central  PubMed  Google Scholar 

  26. Brown LF et al (1992) Expression of vascular permeability factor (vascular endothelial growth factor) by epidermal keratinocytes during wound healing. J Exp Med 176(5):1375–1379

    Article  CAS  PubMed  Google Scholar 

  27. Liang CC, Park AY, Guan JL (2007) In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc 2(2):329–333

    Article  CAS  PubMed  Google Scholar 

  28. Vitorino P, Meyer T (2008) Modular control of endothelial sheet migration. Genes Dev 22(23):3268–3281

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Brown NJ et al (2002) Angiogenesis induction and regression in human surgical wounds. Wound Repair Regen 10(4):245–251

    Article  PubMed  Google Scholar 

  30. Roy S et al (2007) Transcriptome-wide analysis of blood vessels laser captured from human skin and chronic wound-edge tissue. Proc Natl Acad Sci USA 104(36):14472–14477

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  31. Cooper L et al (2005) Wound healing and inflammation genes revealed by array analysis of `macrophageless’ PU.1 null mice. Genome Biol 6(1):R5

    Article  PubMed Central  PubMed  Google Scholar 

  32. Chen L et al (2010) Positional differences in the wound transcriptome of skin and oral mucosa. BMC Genomics 11:471

  33. Roy S et al (2008) Characterization of the acute temporal changes in excisional murine cutaneous wound inflammation by screening of the wound-edge transcriptome. Physiol Genomics 34(2):162–184

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Sullivan TP et al (2001) The pig as a model for human wound healing. Wound Repair Regen 9(2):66–76

    Article  CAS  PubMed  Google Scholar 

  35. Rodero MP, Khosrotehrani K (2010) Skin wound healing modulation by macrophages. Int J Clin Exp Pathol 3(7):643–653

    CAS  PubMed Central  PubMed  Google Scholar 

  36. Morris MR et al (2010) Genome-wide methylation analysis identifies epigenetically inactivated candidate tumour suppressor genes in renal cell carcinoma. Oncogene 30(12):1390–1401

    Article  PubMed  Google Scholar 

  37. Zhou J et al (2005) A novel six-transmembrane protein hhole functions as a suppressor in MAPK signaling pathways. Biochem Biophys Res Commun 333(2):344–352

    Article  CAS  PubMed  Google Scholar 

  38. Barrientos T et al (2007) Two novel members of the ABLIM protein family, ABLIM-2 and -3, associate with STARS and directly bind F-actin. J Biol Chem 282(11):8393–8403

    Article  CAS  PubMed  Google Scholar 

  39. Carmeliet P, Tessier-Lavigne M (2005) Common mechanisms of nerve and blood vessel wiring. Nature 436(7048):193–200

    Article  CAS  PubMed  Google Scholar 

  40. Presta M et al (2005) Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev 16(2):159–178

    Article  CAS  PubMed  Google Scholar 

  41. Boilly B et al (2000) FGF signals for cell proliferation and migration through different pathways. Cytokine Growth Factor Rev 11(4):295–302

    Article  CAS  PubMed  Google Scholar 

  42. Peterson SM et al (2010) Human Sulfatase 2 inhibits in vivo tumor growth of MDA-MB-231 human breast cancer xenografts. BMC Cancer 10:427

    Article  PubMed Central  PubMed  Google Scholar 

  43. Baluk P, Hashizume H, McDonald DM (2005) Cellular abnormalities of blood vessels as targets in cancer. Curr Opin Genet Dev 15(1):102–111

    Article  CAS  PubMed  Google Scholar 

  44. Ozawa MG et al (2008) Beyond receptor expression levels: the relevance of target accessibility in ligand-directed pharmacodelivery systems. Trends Cardiovasc Med 18(4):126–132

    Article  CAS  PubMed  Google Scholar 

  45. Ai X et al (2007) SULF1 and SULF2 regulate heparan sulfate-mediated GDNF signaling for esophageal innervation. Development 134(18):3327–3338

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank Sonja Wessel, Boris Kristof, and Thomas Lange for helpful discussions and support concerning capillary microscopy and analyses.

Conflict of interest

All authors were employees at Beiersdorf at the time point of the study. B. Brückner received consultation fees from Beiersdorf.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marc Winnefeld.

Additional information

Elke Grönniger and Marc Winnefeld have contributed equally.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10456_2015_9472_MOESM1_ESM.pptx

Supplementary material 1 S1 Preprocessing of the microarray data. Flowchart showing the different steps in the microarray experiments and the preprocessing of the microarray data. (PPTX 57 kb)

10456_2015_9472_MOESM2_ESM.pptx

Supplementary material 2 S2 FGFR-1 knockdown reduced endothelial wound closure in vitro. a Expression of FGFR-1 after siRNA-induced knockdown was analyzed 24 h after transfection using qRT-PCR. b The Relative Wound Density in % of scrambled siRNA-transfected blood endothelial cells was compared with siFGFR-1-transfected cells, 30 h after cell scraping. The Relative Wound Density of control-transfected cells was set as 100% and is shown as the dotted line. The horizontal black lines denote medians and whiskers the 2.5th and 97.5th percentiles. Significant differences are indicated by asterisks. * = p ≤ 0.05; ** = p ≤ 0.01; *** = p ≤ 0.001 (unpaired t test). N (scrambled) = 10, n(siFGFR-1) = 6. c Primary endothelial cell viability was measured using Cell-Titer-Blue-Cell Viability Assay (Promega). The data were normalized to scrambled siRNA which is marked as the dotted line. As a negative control, we used cells which were treated with cis-diammineplatinum(II)dichloride (10 µg/mL) for 24 h. n = 12–24. (PPTX 55 kb)

10456_2015_9472_MOESM3_ESM.pptx

Supplementary material 3 S3 Efficiency of siRNA-induced gene knockdown. Primary endothelial cells were transfected with 3 different siRNAs per gene, indicated by the number 1, 2, or 3 (only the two most potent ones are depicted). Expression of siRNA-induced knockdown of a ABLIM2, b GGCT, c SULF2, d TMEM121, and e ZSCAN18 were analyzed 24 h after transfection using qRT-PCR. Knockdown of the corresponding genes was compared to scrambled siRNA-transfected endothelial cells, which were set to 100 %. The horizontal black lines denote medians and whiskers the 2.5th and 97.5th percentiles. Significant differences are indicated by asterisks. * = p ≤ 0.05; ** = p ≤ 0.01; *** = p ≤ 0.001 (unpaired t test). n = 6. (PPTX 76 kb)

10456_2015_9472_MOESM4_ESM.pptx

Supplementary material 4 S4 Cell viability and apoptotic activity after siRNA knockdown. Primary endothelial cells were transfected with 3 different siRNAs per gene, indicated by the number 1, 2, or 3 (only the two most potent ones are depicted). a Cell viability was analyzed 48 h after knockdown of the following genes: ABLIM2, GGCT, SULF2, TMEM121, ZSCAN18, and FGFR-1. Scrambled siRNA-transfected endothelial cells served as controls and were set to 100 %. b Apoptotic activity was determined 48 h after knockdown. For a positive control, cell populations were cultivated for 24 h in the presence of staurosporine. (PPTX 147 kb)

10456_2015_9472_MOESM5_ESM.pptx

Supplementary material 5 S5 Proliferative effects in in vitro wound closure. To analyze the effects of proliferation, cell nuclei were counted via propidium iodide staining. The impact of gene knockdown of a ABLIM2, b GGCT, c SULF2, d TMEM121, and e ZSCAN18 was studied. The left panel shows the results from the first, and the right panel the results from the second screen. The average value of the scrambled control is shown as the dotted line. The horizontal black lines denote medians and whiskers the 2.5th and 97.5th percentiles. Comparison between scrambled siRNA- and siFGFR-1-transfected cells was significant with p ≤ 0,001 (asterisks not shown). Significant differences are indicated by asterisks. * = p ≤ 0.05; ** = p ≤ 0.01; *** = p ≤ 0.001 (unpaired t test). n (per single siRNA) = 6. (PPTX 75 kb)

10456_2015_9472_MOESM6_ESM.pptx

Supplementary material 6 S6 Histological analysis of ABLIM2 and TMEM121. Images captured by immunofluorescence microscopy are shown for one representative volunteer. Skin samples were taken 2 weeks after suction blistering and embedded in paraffin. For DNA staining, Hoechst 33342 was used. a) (Co)localization of CD31, ABLIM2. Bar 50 µm. b) (Co)localization of CD31, ABLIM2 in a higher resolution. Bar: 20 µm. c) (Co)localization of CD31, TMEM121. Bar 50 µm. d) (Co)localization of CD31, TMEM121 in a higher resolution. Bar: 20 µm. (PPTX 1425 kb)

10456_2015_9472_MOESM7_ESM.pptx

Supplementary material 7 S7 Expression of ABLIM2, GGCT, SULF2, TMEM121, and ZSCAN18 in different tissues and cell types. The heat map shows the expression intensities (in percent) of the selected genes normalized to GAPDH intensities (Gene Intensity / GAPDH Intensity * 100). The mean values of 5 or 3 independent microarray experiments have been determined. Color code: blue = highly expressed; red = slightly expressed. (PPTX 43 kb)

Supplementary material 8 Table S1: Summary of cleaned set of 90 samples. sb: suction blister; co: control. (XLSX 18 kb)

10456_2015_9472_MOESM9_ESM.xlsx

Supplementary material 9 Table S2: Differentially expressed genes between wounded and unwounded skin that are enriched in the “Function Angiogenesis.” The analysis has been conducted using the Ingenuity software. (XLSX 15 kb)

10456_2015_9472_MOESM10_ESM.xlsx

Supplementary material 10 Table S3: List of the 48 genes, which are differentially expressed between unwounded (co) and wounded (sb) skin as well as expressed in primary human dermal endothelial cells. Three different siRNAs (Qiagen) were tested per gene. Target sequences are listed, respectively. (XLSX 24 kb)

10456_2015_9472_MOESM11_ESM.xlsx

Supplementary material 11 Table S4: RNA expression differences (FC) of the 48 genes between unwounded skin and skin 2 weeks after wounding. (XLSX 17 kb)

10456_2015_9472_MOESM12_ESM.xlsx

Supplementary material 12 Table S5: RNA expression differences (FC) of the 5 genes between unwounded and wounded skin are listed over time of the wound healing process. Positive data reflect repression, whereas negative data reflect increased expression of the associated gene in sb vs. co. (XLSX 15 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brönneke, S., Brückner, B., Söhle, J. et al. Genome-wide expression analysis of wounded skin reveals novel genes involved in angiogenesis. Angiogenesis 18, 361–371 (2015). https://doi.org/10.1007/s10456-015-9472-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10456-015-9472-7

Keywords

Navigation