Skip to main content

Advertisement

Log in

Adipose-derived Mesenchymal Stem Cells are Ideal for the Cell-based Treatment of Refractory Wounds: Strong Potential for Angiogenesis

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

Although Mesenchymal Stem Cells (MSCs)-based therapy has been proposed as a promising strategy for the treatment of chronic lower-extremity ulcers, their optimal sources, amounts, and delivery methods are urgently needed to be determined. In this study, we compared the heterogeneity of the human MSCs derived from bone marrow (BMSCs), umbilical cord (UCMSCs), and adipose tissue (ADSCs) in accelerating wound healing and promoting angiogenesis and explored the underlying mechanism. Briefly, a diabetic rat model with a full-thickness cutaneous wound on the dorsal foot was developed. The wound was topically administered with three types of MSCs. Additionally, we carried out in vitro and in vivo analysis of the angiogenic properties of the MSCs. Moreover, the molecular mechanism of the heterogeneity of the MSCs derived from the three tissues was explored by transcriptome sequencing. When compared with the BMSCs- and UCMSCs-treated groups, the ADSCs-treated group exhibited markedly accelerated healing efficiency, characterized by increased wound closure rates, enhanced angiogenesis, and collagen deposition at the wound site. The three types of MSCs formed three-dimensional capillary-like structures and promoted angiogenesis in vitro and in vivo, with ADSCs exhibiting the highest capacity for tube formation and pro-angiogenesis. Furthermore, transcriptome sequencing revealed that ADSCs had higher expression levels of angiogenesis-associated genes. Our findings indicate that MSCs-based therapy accelerates the healing of ischemia- and diabetes-induced lower-extremity ulcers and that adipose tissue-derived MSCs might be ideal for therapeutic angiogenesis and treatment of chronic ischemic wounds.

Graphical Abstract

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
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data Availability

The data and materials used to support the findings of this study are available from the corresponding author upon request.

Code Availability

Not applicable.

Abbreviations

ACVRL1:

Activin A receptor like type 1

ADSCs:

Adipose-derived mesenchymal stem cells

BMSCs:

Bone marrow mesenchymal stem cells

CCK-8:

Cell counting kit-8

CD29:

Cluster of differentiation 29

CD31:

Cluster of differentiation 31

CD34:

Cluster of differentiation 34

CD44:

Cluster of differentiation 44

CD45:

Cluster of differentiation 45

CD90:

Cluster of differentiation 90

CTSH:

Cathepsin H

DEGs:

Differentially expressed genes

ECs:

Endothelial cells

EMT:

Epithelial to mesenchymal transition

FPKM:

Fragments per kilobase of transcript per millions mapped reads

GO:

Gene Ontology

GSEA:

Gene set enrichment analysis

H&E:

Hematoxylin and eosin

IHC:

Immunohistochemistry

JAK1:

Janus Kinase 1

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LG-DMEM:

Low glucose Dulbecco’s modified Eagle’s medium

MSCs:

Mesenchymal stem cells

OD:

Optical density

OLGs:

Overlapping genes

PAS:

Periodic acid-Schiff

PCA:

Principal components analysis

PFA:

Paraformaldehyde

RUNX1:

Runt-related transcription factor-1

SFRP2:

Secreted frizzled related-protein 2

STZ:

Streptozotocin

UCMSCs:

Umbilical cord mesenchymal stem cells

References

  1. Mishra, S. C. (2017). Diabetic foot Bmj, 359: j5064.

    PubMed  Google Scholar 

  2. Singh, N., Armstrong, D. G., & Lipsky, B. A. (2005). Preventing foot ulcers in patients with Diabetes. Jama, 293(2), 217–228.

    Article  CAS  PubMed  Google Scholar 

  3. O’Loughlin, A., et al. (2013). Topical administration of allogeneic mesenchymal stromal cells seeded in a collagen scaffold augments wound healing and increases angiogenesis in the diabetic rabbit Ulcer. Diabetes, 62(7), 2588–2594.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kosaric, N., Kiwanuka, H., & Gurtner, G. C. (2019). Stem cell therapies for wound healing. Expert Opinion on Biological Therapy, 19(6), 575–585.

    Article  CAS  PubMed  Google Scholar 

  5. Kolluru, G. K., Bir, S. C., & Kevil, C. G. (2012). Endothelial Dysfunction and Diabetes: Effects on Angiogenesis, Vascular Remodeling, and Wound Healing International Journal of Vascular Medicine, 2012: p. 1–30.

  6. Maamoun, H., et al. (2019). Crosstalk between oxidative stress and endoplasmic reticulum (ER) stress in endothelial dysfunction and aberrant angiogenesis Associated with Diabetes: A focus on the protective roles of Heme Oxygenase (HO)-1. Frontiers in Physiology, 10, 70.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Hu, Y., et al. (2021). Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J Nanobiotechnology, 19(1), 150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen, Y., et al. (2014). Timing of transplantation of autologous bone marrow derived mesenchymal stem cells for treating Myocardial Infarction. Sci China Life Sci, 57(2), 195–200.

    Article  CAS  PubMed  Google Scholar 

  9. Kanji, S., & Das, H. (2017). Advances of Stem Cell Therapeutics in Cutaneous Wound Healing and Regeneration Mediators Inflamm, 2017: p. 5217967.

  10. Mazini, L. (2020). Hopes and limits of adipose-derived stem cells (ADSCs) and mesenchymal stem cells (MSCs) in Wound Healing. International Journal of Molecular Sciences, 21(4).

  11. Nilforoushzadeh, M. A. (2022). Promotion of cutaneous diabetic wound healing by subcutaneous administration of Wharton’s jelly mesenchymal stem cells derived from umbilical cord. Archives of Dermatological Research.

  12. Shi, R., et al. (2016). Localization of human adipose-derived stem cells and their effect in repair of diabetic foot ulcers in rats. Stem Cell Research & Therapy, 7(1), 155.

    Article  Google Scholar 

  13. Wu, Y., et al. (2007). Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells, 25(10), 2648–2659.

    Article  CAS  PubMed  Google Scholar 

  14. Nammian, P., et al. (2021). Comparative analysis of mouse bone marrow and adipose tissue mesenchymal stem cells for critical limb ischemia cell therapy. Stem Cell Research & Therapy, 12(1), 58.

    Article  CAS  Google Scholar 

  15. Amable, P. R., et al. (2014). Protein synthesis and secretion in human mesenchymal cells derived from bone marrow, adipose tissue and Wharton’s jelly. Stem Cell Research & Therapy, 5(2), 53.

    Article  Google Scholar 

  16. Li, C. Y., et al. (2015). Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Research & Therapy, 6, 55.

    Article  Google Scholar 

  17. Wang, Z. G., et al. (2020). Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Research & Therapy, 11(1), 511.

    Article  CAS  Google Scholar 

  18. Kern, S., et al. (2006). Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells, 24(5), 1294–1301.

    Article  CAS  PubMed  Google Scholar 

  19. Yan, J., et al. (2021). Efficacy of topical and systemic transplantation of mesenchymal stem cells in a rat model of diabetic ischemic wounds. Stem Cell Research & Therapy, 12(1), 220.

    Article  CAS  Google Scholar 

  20. Tang, S., et al. (2021). TMSB4 overexpression enhances the potency of Marrow Mesenchymal stromal cells for myocardial repair. Front Cell Dev Biol, 9, 670913.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Gui, L., et al. (2022). ROS-responsive nanoparticle-mediated delivery of CYP2J2 gene for therapeutic angiogenesis in severe hindlimb ischemia. Mater Today Bio, 13, 100192.

    Article  CAS  PubMed  Google Scholar 

  22. Martinotti, S., & Ranzato, E. (2020). Scratch Wound Healing Assay. Methods in Molecular Biology, 2109, 225–229.

    Article  CAS  PubMed  Google Scholar 

  23. Ponce, M. L. (2009). Tube formation: An in vitro matrigel angiogenesis assay. Methods in Molecular Biology, 467, 183–188.

    Article  CAS  PubMed  Google Scholar 

  24. Malinda, K. M. (2009). In vivo matrigel migration and angiogenesis assay. Methods in Molecular Biology, 467, 287–294.

    Article  CAS  PubMed  Google Scholar 

  25. Lin, J., et al. (2018). Long non-coding RNA UBE2CP3 enhances HCC cell secretion of VEGFA and promotes angiogenesis by activating ERK1/2/HIF-1alpha/VEGFA signalling in hepatocellular carcinoma. Journal of Experimental & Clinical Cancer Research : Cr, 37(1), 113.

    Article  PubMed Central  Google Scholar 

  26. Cheng, H. W., et al. (2017). Cancer cells increase endothelial cell tube formation and survival by activating the PI3K/Akt signalling pathway. Journal of Experimental & Clinical Cancer Research : Cr, 36(1), 27.

    Article  PubMed Central  Google Scholar 

  27. Annex, B. H. (2013). Therapeutic angiogenesis for critical limb ischaemia. Nature Reviews. Cardiology, 10(7), 387–396.

    Article  CAS  PubMed  Google Scholar 

  28. Fitzsimmons, R. E. B. (2018). Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering Stem Cells Int, 2018: p. 8031718.

  29. Cao, Y. (2017). Mesenchymal Stem Cells Improve Healing of Diabetic Foot Ulcer J Diabetes Res, 2017: p. 9328347.

  30. Kim, Y., et al. (2007). Direct comparison of human mesenchymal stem cells derived from adipose tissues and bone marrow in mediating neovascularization in response to vascular ischemia. Cellular Physiology and Biochemistry, 20(6), 867–876.

    Article  CAS  PubMed  Google Scholar 

  31. Brennan, M. A., et al. (2017). Inferior in vivo Osteogenesis and Superior Angiogenesis of Human adipose-derived stem cells compared with bone marrow-derived stem cells cultured in Xeno-Free conditions. Stem Cells Transl Med, 6(12), 2160–2172.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Xu, H., et al. (2020). Exosomes derived from adipose tissue, bone marrow, and umbilical cord blood for cardioprotection after Myocardial Infarction. Journal of Cellular Biochemistry, 121(3), 2089–2102.

    Article  CAS  PubMed  Google Scholar 

  33. Veith, A. P., et al. (2019). Therapeutic strategies for enhancing angiogenesis in wound healing. Advanced Drug Delivery Reviews, 146, 97–125.

    Article  CAS  PubMed  Google Scholar 

  34. Shrestha, C. (2013). Enhanced healing of diabetic wounds by subcutaneous administration of human umbilical cord derived stem cells and their conditioned media Int J Endocrinol, 2013: p. 592454.

  35. Rousselle, P., Montmasson, M., & Garnier, C. (2019). Extracellular matrix contribution to skin wound re-epithelialization. Matrix Biology, 75–76: p. 12–26.

  36. Arenas Gómez, C. M., Sabin, K. Z., & Echeverri, K. (2020). Wound healing across the animal kingdom: Crosstalk between the immune system and the extracellular matrix. Developmental Dynamics, 249(7), 834–846.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Tonnesen, M. G., Feng, X., & Clark, R. A. (2000). Angiogenesis in wound healing. The Journal of Investigative Dermatology. Symposium Proceedings / the Society for Investigative Dermatology, Inc. [And] European Society for Dermatological Research, 5(1), 40–46.

    Article  CAS  Google Scholar 

  38. Landen, N. X., Li, D., & Stahle, M. (2016). Transition from inflammation to proliferation: A critical step during wound healing. Cellular and Molecular Life Sciences, 73(20), 3861–3885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Marconi, G. D. (2021). Epithelial-mesenchymal transition (EMT): The Type-2 EMT in Wound Healing, tissue regeneration and Organ Fibrosis. Cells, 10(7).

  40. Haensel, D., & Dai, X. (2018). Epithelial-to-mesenchymal transition in cutaneous wound healing: Where we are and where we are heading. Developmental Dynamics, 247(3), 473–480.

    Article  PubMed  Google Scholar 

  41. Seki, T., Yun, J., & Oh, S. P. (2003). Arterial endothelium-specific activin receptor-like kinase 1 expression suggests its role in arterialization and vascular remodeling. Circ Res, 93(7), 682–689.

    Article  CAS  PubMed  Google Scholar 

  42. Alfaro, M. P., et al. (2010). sFRP2 suppression of bone morphogenic protein (BMP) and wnt signaling mediates mesenchymal stem cell (MSC) self-renewal promoting engraftment and myocardial repair. Journal of Biological Chemistry, 285(46), 35645–35653.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Alfaro, M. P., et al. (2008). The wnt modulator sFRP2 enhances mesenchymal stem cell engraftment, granulation tissue formation and myocardial repair. Proc Natl Acad Sci U S A, 105(47), 18366–18371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lam, J. D., et al. (2017). Identification of RUNX1 as a mediator of aberrant retinal angiogenesis. Diabetes, 66(7), 1950–1956.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Abbasi, S., et al. (2020). Distinct Regulatory Programs Control the Latent Regenerative Potential of Dermal fibroblasts during Wound Healing. Cell Stem Cell, 27(3), 396–412e6.

    Article  CAS  PubMed  Google Scholar 

  46. Ren, S., et al. (2021). The whole profiling and competing endogenous RNA network analyses of noncoding RNAs in adipose-derived stem cells from diabetic, old, and young patients. Stem Cell Research & Therapy, 12(1), 313.

    Article  CAS  Google Scholar 

  47. Skiles, M. L., et al. (2020). Comparison of umbilical cord tissue-derived mesenchymal stromal cells isolated from cryopreserved material and extracted by explantation and digestion methods utilizing a split manufacturing model. Cytotherapy, 22(10), 581–591.

    Article  CAS  PubMed  Google Scholar 

  48. Xu, R., et al. (2018). MicroRNA-31a-5p from aging BMSCs links bone formation and resorption in the aged bone marrow microenvironment. Aging Cell, 17(4), e12794.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Li, H., et al. (2016). Isolation and characterization of primary bone marrow mesenchymal stromal cells. Annals of the New York Academy of Sciences, 1370(1), 109–118.

    Article  CAS  PubMed  Google Scholar 

  50. Wang, Q., et al. (2019). Comparative evaluation of the osteogenic capacity of human mesenchymal stem cells from bone marrow and umbilical cord tissue. Exp Ther Med, 17(1), 764–772.

    CAS  PubMed  Google Scholar 

  51. Gadelkarim, M., et al. (2018). Adipose-derived stem cells: Effectiveness and advances in delivery in diabetic wound healing. Biomedicine & Pharmacotherapy, 107, 625–633.

    Article  CAS  Google Scholar 

  52. Kilinc, M. O., et al. (2018). The ratio of ADSCs to HSC-progenitors in adipose tissue derived SVF may provide the key to predict the outcome of stem-cell therapy. Clin Transl Med, 7(1), 5.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Choudhery, M. S., et al. (2015). Subcutaneous adipose tissue-derived stem cell utility is Independent of Anatomical Harvest Site. Biores Open Access, 4(1), 131–145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Eto, H., et al. (2009). Characterization of structure and cellular components of aspirated and excised adipose tissue. Plastic and Reconstructive Surgery, 124(4), 1087–1097.

    Article  CAS  PubMed  Google Scholar 

  55. Huang, Y. Z., et al. (2020). Mesenchymal stem cells for Chronic Wound Healing: Current status of preclinical and clinical studies. Tissue Eng Part B Rev, 26(6), 555–570.

    Article  CAS  PubMed  Google Scholar 

  56. Li, T., et al. (2015). Human umbilical cord mesenchymal stem cells: An overview of their potential in cell-based therapy. Expert Opinion on Biological Therapy, 15(9), 1293–1306.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by National Nature and Science Foundation of P.R. China (No.81871563, 82102345), Medical Research Foundation of Guangdong Province (A2021165), Fundamental Research Funds for the Central Universities (21619350), Guangdong Basic and Applied Basic Research Foundation (2019A1515110161, 2022A1515110940), Funding by Science and Technology Projects in Guangzhou (202201020002, 202201020004, 202201020468), Clinical Frontier Technology Program of the First Affiliated Hospital of Jinan University, China (No. JNU1AF-CFTP-2022-a01231, No. JNU1AF-CFTP-2022-a01208), Guangdong Provincial Key Areas R&D Programs (2022B1111080007), Special fund of Foshan Summit plan (No.2019D008), Medical Research Fund of Guangdong Province (No. A2020322), Research Grant of Key Laboratory of Regenerative Medicine, Ministry of Education, Jinan University (No: ZSYXM202209), China Postdoctoral Science Foundation (2022M721348).

Author information

Authors and Affiliations

Authors

Contributions

YXC: Provision of study material or patients, collection and assembly of data, data analysis and interpretation, manuscript writing, final approval of manuscript; JXY: Collection and assembly of data, data analysis and interpretation, manuscript writing, final approval of manuscript; ZQD: Data analysis and interpretation, manuscript writing, final approval of manuscript; JRW, XJ: Financial support, final approval of manuscript; TXC: Conception and design, data analysis and interpretation, final approval of manuscript; YSH: Financial support, final approval of manuscript, final approval of manuscript; HWL: Conception and design, financial support, administrative support, manuscript writing, final approval of manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Taixing Cui, Yuesheng Huang or Hongwei Liu.

Ethics declarations

Ethics Approval

The study was performed in accordance with the Declaration of Helsinki principles and was approved by the Medical Ethics Committee of the First Affiliated Hospital of Jinan University. The animal experiment protocol was approved by the Laboratory Animal Ethics Committee of Jinan University and performed following institutional guidelines.

Consent to Participate

Informed consent was obtained from all individual participants included in the study.

Consent for Publication

Not applicable.

Conflict of Interest

The authors declare that they have no potential conflicts of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

12015_2023_10641_MOESM1_ESM.png

Supplementary Material 1: Figure S1. MSC migration and proliferation capacity. (A) Scratch assay to detect the migration of BMSCs, UCMSCs, and ADSCs. (B) Quantitative analysis of wound closure rate. (C) CCK-8 assay at 1, 2, 3, 4, and 5 days of culture showing the proliferation of BMSCs, UCMSCs, and ADSCs. Data are presented as the mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001

12015_2023_10641_MOESM2_ESM.png

Supplementary Material 2: Figure S2. The top 20 enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway terms of positive regulation of angiogenesis-associated differentially expressed genes (DEGs)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cao, Y., Yan, J., Dong, Z. et al. Adipose-derived Mesenchymal Stem Cells are Ideal for the Cell-based Treatment of Refractory Wounds: Strong Potential for Angiogenesis. Stem Cell Rev and Rep 20, 313–328 (2024). https://doi.org/10.1007/s12015-023-10641-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-023-10641-y

Keywords

Navigation