Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Research Article

Preliminary Study on Human Adipose Stem Cells Promoting Skin Wound Healing through Notch Signaling Pathway

Author(s): Yi Wang, Mengjie Dong, Yang Zheng, Chao Wang, Xu Ding, Heming Wu, Yunong Wu, Wei Zhang* and Xiaomeng Song*

Volume 18, Issue 5, 2023

Published on: 13 January, 2023

Page: [699 - 711] Pages: 13

DOI: 10.2174/1574888X18666221216123259

Price: $65

Abstract

Background: Mesenchymal stem cells (MSCs) have been documented as possible candidates for wound healing treatment because their use could reinforce the regenerative capacity of many tissues. Human adipose stem cells (hADSCs) have the advantages of easy access, large quantity and easy operation. They can be fully applied in the treatment of skin wounds.

Objective: In this study, we aim to explore the roles and potential mechanisms of hADSCs in cutaneous wound healing.

Methods: hADSCs were obtained from human subcutaneous fat. Adipocytes and osteocytes differentiated from hADSCs were determined by staining with Oil Red O and alkaline phosphatase (ALP), respectively. We assessed the effects of hADSCs and hADSC conditional medium (CM) on wound healing in an injury model of mice. Then, we investigated the biological effects of hADSCs on human keratinocytes HaCAT cells in vitro.

Results: The results showed that hADSCs could be successfully differentiated into osteogenic and lipogenic cells. hADSCs and hADSCs-CM significantly promote skin wound healing in vivo. hADSCs significantly promoted HaCAT cell proliferation and migration by activating the Notch signaling pathway and activated the AKT signaling pathway by Rps6kb1 kinase in HaCAT cells. In addition, we found that hADSCs-mediated activation of Rps6kb1/AKT signaling was dependent on the Notch signaling pathway.

Conclusion: We demonstrated that hADSCs can promote skin cell-HaCAT cell proliferation and migration via the Notch pathway, suggesting that hADSCs may provide an alternative therapeutic approach for the treatment of skin injury.

Keywords: Adipose derived stem cells, HaCAT cells, Jagged1, Notch, skin wound healing, Rps6kb1/AKT.

Graphical Abstract
[1]
Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound healing: A cellular perspective. Physiol Rev 2019; 99(1): 665-706.
[http://dx.doi.org/10.1152/physrev.00067.2017] [PMID: 30475656]
[2]
Shafei S, Khanmohammadi M, Heidari R, et al. Exosome loaded alginate hydrogel promotes tissue regeneration in full‐thickness skin wounds: An in vivo study. J Biomed Mater Res A 2020; 108(3): 545-56.
[http://dx.doi.org/10.1002/jbm.a.36835] [PMID: 31702867]
[3]
He L, Zhu C, Jia J, et al. ADSC-Exos containing MALAT1 promotes wound healing by targeting miR-124 through activating Wnt/β-catenin pathway. Biosci Rep 2020; 40(5): BSR20192549.
[http://dx.doi.org/10.1042/BSR20192549] [PMID: 32342982]
[4]
Rousselle P, Montmasson M, Garnier C. Extracellular matrix contribution to skin wound re-epithelialization. Matrix Biol 2019; 75-76: 12-26.
[http://dx.doi.org/10.1016/j.matbio.2018.01.002] [PMID: 29330022]
[5]
Wang X, Jiao Y, Pan Y, et al. Fetal dermal mesenchymal stem cell-derived exosomes accelerate cutaneous wound healing by activating notch signaling. Stem Cells Int 2019; 2019: 1-11.
[http://dx.doi.org/10.1155/2019/2402916] [PMID: 31281370]
[6]
Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10(1): 111.
[http://dx.doi.org/10.1186/s13287-019-1212-2] [PMID: 30922387]
[7]
Hassan WU, Greiser U, Wang W. Role of adipose-derived stem cells in wound healing. Wound Repair Regen 2014; 22(3): 313-25.
[http://dx.doi.org/10.1111/wrr.12173] [PMID: 24844331]
[8]
Jeon YK, Jang YH, Yoo DR, Kim SN, Lee SK, Nam MJ. Mesenchymal stem cells’ interaction with skin: Wound-healing effect on fibroblast cells and skin tissue. Wound Repair Regen 2010; 18(6): 655-61.
[http://dx.doi.org/10.1111/j.1524-475X.2010.00636.x] [PMID: 20955344]
[9]
Luo G, Cheng W, He W, et al. Promotion of cutaneous wound healing by local application of mesenchymal stem cells derived from human umbilical cord blood. Wound Repair Regen 2010; 18(5): 506-13.
[http://dx.doi.org/10.1111/j.1524-475X.2010.00616.x] [PMID: 20840520]
[10]
Qiu X, Liu J, Zheng C, et al. Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif 2020; 53(8): e12830.
[http://dx.doi.org/10.1111/cpr.12830] [PMID: 32608556]
[11]
Gu J, Liu N, Yang X, Feng Z, Qi F. Adiposed-derived stem cells seeded on PLCL/P123 eletrospun nanofibrous scaffold enhance wound healing. Biomed Mater 2014; 9(3): 035012.
[http://dx.doi.org/10.1088/1748-6041/9/3/035012] [PMID: 24846986]
[12]
Ha DH, Kim H, Lee J, et al. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells 2020; 9(5): 1157.
[http://dx.doi.org/10.3390/cells9051157] [PMID: 32392899]
[13]
Smith AN, Willis E, Chan VT, et al. Mesenchymal stem cells induce dermal fibroblast responses to injury. Exp Cell Res 2010; 316(1): 48-54.
[http://dx.doi.org/10.1016/j.yexcr.2009.08.001] [PMID: 19666021]
[14]
Ma T, Fu B, Yang X, Xiao Y, Pan M. Adipose mesenchymal stem cell‐derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β‐catenin signaling in cutaneous wound healing. J Cell Biochem 2019; 120(6): 10847-54.
[http://dx.doi.org/10.1002/jcb.28376] [PMID: 30681184]
[15]
Puissant B, Barreau C, Bourin P, et al. Immunomodulatory effect of human adipose tissue-derived adult stem cells: Comparison with bone marrow mesenchymal stem cells. Br J Haematol 2005; 129(1): 118-29.
[http://dx.doi.org/10.1111/j.1365-2141.2005.05409.x] [PMID: 15801964]
[16]
McIntosh KR. Evaluation of cellular and humoral immune responses to allogeneic adipose-derived stem/stromal cells. Methods Mol Biol 2011; 702: 133-50.
[http://dx.doi.org/10.1007/978-1-61737-960-4_11] [PMID: 21082400]
[17]
Charles-de-Sá L, Gontijo-de-Amorim NF, Maeda Takiya C, et al. Antiaging treatment of the facial skin by fat graft and adipose-derived stem cells. Plast Reconstr Surg 2015; 135(4): 999-1009.
[http://dx.doi.org/10.1097/PRS.0000000000001123] [PMID: 25811565]
[18]
Hong SJ, Jia SX, Xie P, et al. Topically delivered adipose derived stem cells show an activated-fibroblast phenotype and enhance granulation tissue formation in skin wounds. PLoS One 2013; 8(1): e55640.
[http://dx.doi.org/10.1371/journal.pone.0055640] [PMID: 23383253]
[19]
Kilroy GE, Foster SJ, Wu X, et al. Cytokine profile of human adipose-derived stem cells: Expression of angiogenic, hematopoietic, and pro-inflammatory factors. J Cell Physiol 2007; 212(3): 702-9.
[http://dx.doi.org/10.1002/jcp.21068] [PMID: 17477371]
[20]
Cho JW, Kang MC, Lee KS. TGF-β1-treated ADSCs-CM promotes expression of type I collagen and MMP-1, migration of human skin fibroblasts, and wound healing in vitro and in vivo. Int J Mol Med 2010; 26(6): 901-6.
[PMID: 21042785]
[21]
Heirani-Tabasi A, Toosi S, Mirahmadi M, et al. Chemokine receptors expression in MSCs: Comparative analysis in different sources and passages. Tissue Eng Regen Med 2017; 14(5): 605-15.
[http://dx.doi.org/10.1007/s13770-017-0069-7] [PMID: 30603514]
[22]
Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: Cell fate control and signal integration in development. Science 1999; 284(5415): 770-6.
[http://dx.doi.org/10.1126/science.284.5415.770] [PMID: 10221902]
[23]
Radtke F, Fasnacht N, MacDonald HR. Notch signaling in the immune system. Immunity 2010; 32(1): 14-27.
[http://dx.doi.org/10.1016/j.immuni.2010.01.004] [PMID: 20152168]
[24]
Yang RH, Qi SH, Shu B, et al. Epidermal stem cells (ESCs) accelerate diabetic wound healing via the notch signalling pathway. Biosci Rep 2016; 36(4): e00364.
[http://dx.doi.org/10.1042/BSR20160034] [PMID: 27129289]
[25]
Shi Y, Shu B, Yang R, et al. Wnt and Notch signaling pathway involved in wound healing by targeting c-Myc and Hes1 separately. Stem Cell Res Ther 2015; 6(1): 120.
[http://dx.doi.org/10.1186/s13287-015-0103-4] [PMID: 26076648]
[26]
Pförringer D, Aitzetmüller MM, Brett EA, et al. Single-cell gene expression analysis and evaluation of the therapeutic function of murine Adipose-Derived Stromal Cells (ASCs) from the subcutaneous and visceral compartment. Stem Cells Int 2018; 2018: 2183736.
[http://dx.doi.org/10.1155/2018/2183736] [PMID: 30651733]
[27]
Dai MY, Fang F, Zou Y, et al. Downregulation of Notch1 induces apoptosis and inhibits cell proliferation and metastasis in laryngeal squamous cell carcinoma. Oncol Rep 2015; 34(6): 3111-9.
[http://dx.doi.org/10.3892/or.2015.4274] [PMID: 26398771]
[28]
Cai Y, Li J, Jia C, He Y, Deng C. Therapeutic applications of adipose cell-free derivatives: A review. Stem Cell Res Ther 2020; 11(1): 312.
[http://dx.doi.org/10.1186/s13287-020-01831-3] [PMID: 32698868]
[29]
Mazini L, Rochette L, Admou B, Amal S, Malka G. Hopes and limits of adipose-derived stem cells (ADSCs) and mesenchymal stem cells (MSCs) in wound healing. Int J Mol Sci 2020; 21(4): 1306.
[http://dx.doi.org/10.3390/ijms21041306] [PMID: 32075181]
[30]
Stojanović S, Najman S. The effect of conditioned media of stem cells derived from lipoma and adipose tissue on macrophages’ response and wound healing in indirect co-culture system in vitro. Int J Mol Sci 2019; 20(7): 1671.
[http://dx.doi.org/10.3390/ijms20071671] [PMID: 30987193]
[31]
Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells. Identification of a 5′ enhancer. Circ Res 1995; 77(3): 638-43.
[http://dx.doi.org/10.1161/01.RES.77.3.638] [PMID: 7641334]
[32]
Liu SC, Bamodu OA, Kuo KT, et al. Adipose-derived stem cell induced-tissue repair or wound healing is mediated by the concomitant upregulation of miR-21 and miR-29b expression and activation of the AKT signaling pathway. Arch Biochem Biophys 2021; 705: 108895.
[http://dx.doi.org/10.1016/j.abb.2021.108895] [PMID: 33933426]
[33]
Wang X, Ma Y, Gao Z, Yang J. Human adipose-derived stem cells inhibit bioactivity of keloid fibroblasts. Stem Cell Res Ther 2018; 9(1): 40.
[http://dx.doi.org/10.1186/s13287-018-0786-4] [PMID: 29467010]
[34]
Wong VW, Sorkin M, Glotzbach JP, Longaker MT, Gurtner GC. Surgical approaches to create murine models of human wound healing. J Biomed Biotechnol 2011; 2011: 969618.
[http://dx.doi.org/10.1155/2011/969618] [PMID: 21151647]
[35]
Gerber PA, Buhren BA, Schrumpf H, Homey B, Zlotnik A, Hevezi P. The top skin-associated genes: A comparative analysis of human and mouse skin transcriptomes. Biol Chem 2014; 395(6): 577-91.
[http://dx.doi.org/10.1515/hsz-2013-0279] [PMID: 24497224]
[36]
Cibelli J, Emborg ME, Prockop DJ, et al. Strategies for improving animal models for regenerative medicine. Cell Stem Cell 2013; 12(3): 271-4.
[http://dx.doi.org/10.1016/j.stem.2013.01.004] [PMID: 23472868]
[37]
Dutta S, Sengupta P. Men and mice: Relating their ages. Life Sci 2016; 152: 244-8.
[http://dx.doi.org/10.1016/j.lfs.2015.10.025] [PMID: 26596563]
[38]
Abdullahi A, Amini-Nik S, Jeschke MG. Animal models in burn research. Cell Mol Life Sci 2014; 71(17): 3241-55.
[http://dx.doi.org/10.1007/s00018-014-1612-5] [PMID: 24714880]
[39]
Zomer HD, Trentin AG. Skin wound healing in humans and mice: Challenges in translational research. J Dermatol Sci 2018; 90(1): 3-12.
[http://dx.doi.org/10.1016/j.jdermsci.2017.12.009] [PMID: 29289417]
[40]
Fang RC, Mustoe TA. Animal models of wound healing: Uility in transgenic mice. J Biomater Sci Polym Ed 2008; 19(8): 989-1005.
[http://dx.doi.org/10.1163/156856208784909327] [PMID: 18644226]
[41]
Wang X, Ge J, Tredget EE, Wu Y. The mouse excisional wound splinting model, including applications for stem cell transplantation. Nat Protoc 2013; 8(2): 302-9.
[http://dx.doi.org/10.1038/nprot.2013.002] [PMID: 23329003]
[42]
Galiano RD, Michaels JV, Dobryansky M, Levine JP, Gurtner GC. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen 2004; 12(4): 485-92.
[http://dx.doi.org/10.1111/j.1067-1927.2004.12404.x] [PMID: 15260814]
[43]
Hsu YH, Lin YF, Chen CH, Chiu YJ, Chiu HW. Far infrared promotes wound healing through activation of Notch1 signaling. J Mol Med 2017; 95(11): 1203-13.
[http://dx.doi.org/10.1007/s00109-017-1580-y] [PMID: 28831511]
[44]
Zhang C, Zhu Y, Lu S, Zhong W, Wang Y, Chai Y. Platelet-rich plasma with endothelial progenitor cells accelerates diabetic wound healing in rats by upregulating the notch1 signaling pathway. J Diabetes Res 2019; 2019: 5920676.
[http://dx.doi.org/10.1155/2019/5920676] [PMID: 31559315]
[45]
Nickoloff BJ, Qin J-Z, Chaturvedi V, Denning MF, Bonish B, Miele L. Jagged-1 mediated activation of notch signaling induces complete maturation of human keratinocytes through NF-κB and PPARγ. Cell Death Differ 2002; 9(8): 842-55.
[http://dx.doi.org/10.1038/sj.cdd.4401036] [PMID: 12107827]
[46]
Wang Y, Tu W, Lou Y, et al. Mesenchymal stem cells regulate the proliferation and differentiation of neural stem cells through Notch signaling. Cell Biol Int 2009; 33(11): 1173-9.
[http://dx.doi.org/10.1016/j.cellbi.2009.08.004] [PMID: 19706332]
[47]
Shen Q, Goderie SK, Jin L, et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 2004; 304(5675): 1338-40.
[http://dx.doi.org/10.1126/science.1095505] [PMID: 15060285]
[48]
Ou J, Guan D, Yang Y. Non-contact co-culture with human vascular endothelial cells promotes epithelial-to-mesenchymal transition of cervical cancer SiHa cells by activating the NOTCH1/LOX/SNAIL pathway. Cell Mol Biol Lett 2019; 24(1): 39.
[http://dx.doi.org/10.1186/s11658-019-0163-z] [PMID: 31205475]
[49]
Guo D, Ye J, Li L, Dai J, Ma D, Ji C. Down-regulation of Notch-1 increases co-cultured Jurkat cell sensitivity to chemotherapy. Leuk Lymphoma 2009; 50(2): 270-8.
[http://dx.doi.org/10.1080/10428190802553257] [PMID: 19160126]
[50]
Hemmings BA. Akt signaling: linking membrane events to life and death decisions. Science 1997; 275(5300): 628-30.
[http://dx.doi.org/10.1126/science.275.5300.628] [PMID: 9019819]
[51]
Liu W, Yu M, Xie D, et al. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther 2020; 11(1): 259.
[http://dx.doi.org/10.1186/s13287-020-01756-x] [PMID: 32600435]
[52]
Yu M, Liu W, Li J, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther 2020; 11(1): 350.
[http://dx.doi.org/10.1186/s13287-020-01824-2] [PMID: 32787917]
[53]
Jun E, Zhang Q, Yoon B, et al. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-β/SMAD2 and PI3K/Akt pathways. Int J Mol Sci 2014; 15(1): 605-28.
[http://dx.doi.org/10.3390/ijms15010605] [PMID: 24398984]
[54]
Li JY, Ren KK, Zhang WJ, et al. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res Ther 2019; 10(1): 247.
[http://dx.doi.org/10.1186/s13287-019-1366-y] [PMID: 31399039]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy