We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

LncRNA MALAT1 from human adipose-derived stem cell exosomes accelerates wound healing via miR-378a/FGF2 axis

    Li Pi

    Department of Burn & Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, P.R. China

    ,
    Li Yang

    Department of Burn & Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, P.R. China

    ,
    Bai-Rong Fang

    Department of Burn & Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, P.R. China

    ,
    Xian-Xi Meng

    Department of Burn & Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, P.R. China

    &
    Li Qian

    *Author for correspondence: Tel.: +86 158 7485 9977;

    E-mail Address: 616067028@csu.edu.cn

    Department of Burn & Plastic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, P.R. China

    Published Online:https://doi.org/10.2217/rme-2021-0170

    Aim: The effects of MALAT1 from human adipose-derived stem cell (ADSC) exosomes in skin wound healing were investigated. Material & methods: The viability, apoptosis and migration ability of human skin fibroblasts (HSFs) were evaluated by Cell Counting Kit-8 assay, flow cytometry and scratch assay, respectively. A mouse model was established to evaluate the role of exosomal MALAT1 in skin wound healing in vivo. Results: Human ADSC exosomes promoted the proliferation and migration of HSFs and increased MALAT1 expression. MALAT1 silencing in human ADSCs inhibited HSF viability and migration, promoted HSF apoptosis and inhibited angiogenesis by upregulating miR-378a. Overexpression of miR-378a inhibited the migration and proliferation of HSFs by downregulating FGF2 expression. ADSC exosomes promoted skin wound healing by mediating MALAT1 in vivo. Conclusion: Exosomal MALAT1 accelerated skin wound healing by regulating the miR-378a/FGF2 axis, suggesting that MALAT1 might be used as a potential target for cutaneous wound treatment.

    Graphical abstract

    Plain language summary

    Skin wound healing is a process of synergistic action of multiple factors. Adipose-derived stem cells (ADSCs), a group of stem cells, are recruited into damaged tissues and secret several cytokines, which promote nascent tissue formation. ADSC-derived exosomes play crucial roles in wound healing as a paracrine vehicle for delivering chemokines, growth factors and RNAs to host cells. LncRNAs are involved in multiple physiological processes, including tissue repair. Furthermore, lncRNA MALAT1 is associated with endothelial cell migration and angiogenesis in different types of diseases. This study demonstrated that hADSC exosomes promoted the proliferation and migration of human skin fibroblasts and increased MALAT1 expression. MALAT1 silencing in human ADSCs inhibited human skin fibroblast viability and migration, promoted apoptosis and suppressed angiogenesis by upregulating miR-378a. miR-378a overexpression inhibited the phenotypic characteristics of human skin fibroblasts by downregulating FGF2. Exosomal MALAT1 appeared to accelerate skin wound healing by mediating the miR-378a/FGF2 axis.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Rúben F Pereira1 CCBPLGPJB. Advanced biofabrication strategies for skin regeneration and repair. Nanomedicine 8(4), 603–621 (2013).
    • 2. Gonzalez AC, Costa TF, Andrade ZA, Medrado AR. Wound healing – a literature review. An. Bras. Dermatol. 91(5), 614–620 (2016).
    • 3. Bainbridge P. Wound healing and the role of fibroblasts. J. Wound Care 22(8), 407–408 410–412 (2013). • Fibroblasts play a key role in wound healing.
    • 4. Addis R, Cruciani S, Santaniello S et al. Fibroblast proliferation and migration in wound healing by phytochemicals: evidence for a novel synergic outcome. Int. J. Med. Sci. 17(8), 1030–1042 (2020).
    • 5. Bajada S, Mazakova I, Richardson JB, Ashammakhi N. Updates on stem cells and their applications in regenerative medicine. J. Tissue Eng. Regen. Med. 2(4), 169–183 (2008).
    • 6. Curley GF, Hayes M, Ansari B et al. Mesenchymal stem cells enhance recovery and repair following ventilator-induced lung injury in the rat. Thorax 67(6), 496–501 (2012).
    • 7. Gnecchi M, Danieli P, Malpasso G, Ciuffreda MC. Paracrine mechanisms of mesenchymal stem cells in tissue repair. Methods Mol. Biol. 1416, 123–146 (2016).
    • 8. Hu L, Wang J, Zhou X et al. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 6, 32993 (2016).
    • 9. Zhang J, Guan J, Niu X et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 13, 49 (2015). • Evidence supports that exosomes secreted by stem cells promote skin wound healing.
    • 10. Zhao W, Zheng XL, Zhao SP. Exosome and its roles in cardiovascular diseases. Heart Fail. Rev. 20(3), 337–348 (2015).
    • 11. Cabili MN, Trapnell C, Goff L et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 25(18), 1915–1927 (2011).
    • 12. De Lima DS, Cardozo LE, Maracaja-Coutinho V et al. Long noncoding RNAs are involved in multiple immunological pathways in response to vaccination. Proc. Natl Acad. Sci. USA 116(34), 17121–17126 (2019).
    • 13. Jiang Y, Li Y, Fang S et al. The role of MALAT1 correlates with HPV in cervical cancer. Oncol. Lett. 7(6), 2135–2141 (2014).
    • 14. Fahs F, Bi X, Yu FS, Zhou L, Mi QS. New insights into microRNAs in skin wound healing. IUBMB Life 67(12), 889–896 (2015).
    • 15. Li D, Wang A, Liu X et al. MicroRNA-132 enhances transition from inflammation to proliferation during wound healing. J. Clin. Investig. 125(8), 3008–3026 (2015).
    • 16. Aunin E, Broadley D, Ahmed MI, Mardaryev AN, Botchkareva NV. Exploring a Role for Regulatory miRNAs In Wound Healing during Ageing: Involvement of miR-200c in wound repair. Sci. Rep. 7(1), 3257 (2017).
    • 17. Wang T, Feng Y, Sun H et al. miR-21 regulates skin wound healing by targeting multiple aspects of the healing process. Am. J. Pathol. 181(6), 1911–1920 (2012). •• The construction of a wound model.
    • 18. Wang JM, Tao J, Chen DD et al. MicroRNA miR-27b rescues bone marrow-derived angiogenic cell function and accelerates wound healing in type 2 diabetes mellitus. Arterioscler. Thromb. Vasc. Biol. 34(1), 99–109 (2014).
    • 19. Li P, He Q, Luo C, Qian L. Differentially expressed miRNAs in acute wound healing of the skin: a pilot study. Medicine 94(7), e458 (2015).
    • 20. Liang L, Stone RC, Stojadinovic O et al. Integrative analysis of miRNA and mRNA paired expression profiling of primary fibroblast derived from diabetic foot ulcers reveals multiple impaired cellular functions. Wound Repair Regen. 24(6), 943–953 (2016).
    • 21. Hu ZC, Chen D, Guo D et al. Randomized clinical trial of autologous skin cell suspension combined with skin grafting for chronic wounds. Br. J. Surg. 102(2), e117–123 (2015).
    • 22. Kanapathy M, Hachach-Haram N, Bystrzonowski N, Harding K, Mosahebi A, Richards T. Epidermal grafting versus split-thickness skin grafting for wound healing (EPIGRAAFT): study protocol for a randomised controlled trial. Trials 17(1), 245 (2016).
    • 23. Loreti EH, Pascoal VL, Nogueira BV, Silva IV, Pedrosa DF. Use of laser therapy in the healing process: a literature review. Photomed. Laser Surg. 33(2), 104–116 (2015).
    • 24. Lorenz P, Leavitt T, Hu M, Marshall C, Barnes L, Longaker M. Stem cells and chronic wound healing: state of the art. Chronic Wound Care Manag. Res. doi: 10.2147/cwcmr.s84369 7 (2016).
    • 25. Tsai HW, Wang PH, Tsui KH. Mesenchymal stem cell in wound healing and regeneration. JCMA 81(3), 223–224 (2018).
    • 26. Kølle SF, Fischer-Nielsen A, Mathiasen AB et al. Enrichment of autologous fat grafts with ex-vivo expanded adipose tissue-derived stem cells for graft survival: a randomised placebo-controlled trial. Lancet (London) 382(9898), 1113–1120 (2013).
    • 27. Fang S, Xu C, Zhang Y et al. Umbilical cord-derived mesenchymal stem cell-derived exosomal microRNAs suppress myofibroblast differentiation by inhibiting the transforming growth factor-beta/SMAD2 pathway during wound healing. Stem Cells Transl. Med. 5(10), 1425–1439 (2016).
    • 28. Januszyk K, Lima CD. The eukaryotic RNA exosome. Curr. Opin. Struct. Biol. 24, 132–140 (2014).
    • 29. Cui Y, Fu S, Sun D, Xing J, Hou T, Wu X. EPC-derived exosomes promote osteoclastogenesis through LncRNA-MALAT1. J. Cell Mol. Med. 23(6), 3843–3854 (2019).
    • 30. Li B, Luan S, Chen J et al. The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via microRNA-152-3p. Mol. Ther. Nucleic Acids 19, 814–826 (2020).
    • 31. Li D, Kular L, Vij M et al. Human skin long noncoding RNA WAKMAR1 regulates wound healing by enhancing keratinocyte migration. Proc. Natl Acad. Sci. USA 116(19), 9443–9452 (2019).
    • 32. Liu XQ, Duan LS, Chen YQ et al. lncRNA MALAT1 accelerates wound healing of diabetic mice transfused with modified autologous blood via the HIF-1alpha signaling pathway. Mol. Ther. Nucleic Acids 17, 504–515 (2019). • MALAT1 participates in the skin wound healing process.
    • 33. Cooper DR, Wang C, Patel R et al. Human adipose-derived stem cell conditioned media and exosomes containing MALAT1 promote human dermal fibroblast migration and ischemic wound healing. Adv. Wound Care 7(9), 299–308 (2018).
    • 34. Yu F, Zheng J, Mao Y et al. Long non-coding RNA growth arrest-specific transcript 5 (GAS5) inhibits liver fibrogenesis through a mechanism of competing endogenous RNA. J. Biol. Chem. 290(47), 28286–28298 (2015).
    • 35. Li H, Chang L, Du WW et al. Anti-microRNA-378a enhances wound healing process by upregulating integrin beta-3 and vimentin. Mol. Ther. 22(10), 1839–1850 (2014).
    • 36. Wu M, Huang Y, Chen T et al. LncRNA MEG3 inhibits the progression of prostate cancer by modulating miR-9-5p/QKI-5 axis. J. Cell Mol. Med. 23(1), 29–38 (2019).
    • 37. Peng C, Chen B, Kao HK, Murphy G, Orgill DP, Guo L. Lack of FGF-7 further delays cutaneous wound healing in diabetic mice. Plast. Reconstr. Surg. 128(6), 673e–684e (2011).
    • 38. Subramaniam N, Petrik JJ, Vickaryous MK. VEGF, FGF-2 and TGFbeta expression in the normal and regenerating epidermis of geckos: implications for epidermal homeostasis and wound healing in reptiles. J. Anat. 232(5), 768–782 (2018).
    • 39. Sideek MA, Teia A, Kopecki Z, Cowin AJ, Gibson MA. Co-localization of LTBP-2 with FGF-2 in fibrotic human keloid and hypertrophic scar. J. Mol. Histol. 47(1), 35–45 (2016).
    • 40. Wang X, Li M, Wang Z et al. Silencing of long noncoding RNA MALAT1 by miR-101 and miR-217 inhibits proliferation, migration, and invasion of esophageal squamous cell carcinoma cells. J. Biol. Chem. 290(7), 3925–3935 (2015).
    • 41. Noishiki C, Yuge S, Ando K et al. Live imaging of angiogenesis during cutaneous wound healing in adult zebrafish. Angiogenesis 22(2), 341–354 (2019).
    • 42. Dipietro LA. Angiogenesis and wound repair: when enough is enough. J. Leukoc. Biol. 100(5), 979–984 (2016).
    • 43. Gushiken LFS, Beserra FP, Bastos JK, Jackson CJ, Pellizzon CH. Cutaneous wound healing: an update from physiopathology to current therapies. Life (Basel) 11(7), 665 (2021).