Abstract

BackgroundIt is well established that adipose-derived stem cells (ADSCs) produce and secrete cytokines/growth factors that antagonize UV-induced photoaging of skin. However, the exact molecular basis underlying the anti-photoaging effects exerted by ADSCs is not well understood, and whether ADSCs cooperate with fractional carbon dioxide (CO2) laser to facilitate photoaging skin healing process has not been explored. Here, we investigated the impacts of ADSCs on photoaging in a photoaging animal model, its associated mechanisms, and its functional cooperation with fractional CO2 laser in treatment of photoaging skin.ResultsWe showed that ADSCs improved dermal thickness and activated the proliferation of dermal fibroblast. We further demonstrated that the combined treatment of ADSCs and fractional CO2 laser, the latter which is often used to resurface skin and treat wrinkles, had more beneficial effects on the photoaging skin compared with each individual treatment. In our prepared HDF photoaging model, flow cytometry showed that, after adipose derived stem cells conditioned medium (ADSC-CM) co-cultured HDF photoaging model, the cell proliferation rate is higher than UVB irradiation induced HDF modeling (p < 0.05). Additionally, the expressions of β-catenin and Wnt3a, which were up-regulated after the transplantation of ADSCs alone or in combination with fractional CO2 laser treatment. And the expression of wnt3a and β-catenin has the positive correlation with photoaging related protein TGF-β2 and COLI. We also verified these protein expressions in tissue level. In addition, after injected SFRP2 into ADSC-CM co-cultured HDF photoaging model, wnt3a inhibitor, compared with un-intervened group, wnt3a, β-catenin protein level significantly decreased.ConclusionBoth ADSCs and fractional CO2 laser improved photoaging skin at least partially via targeting dermal fibroblast activity which was increased in photoaging skin. The combinatorial use of ADSCs and fractional CO2 laser synergistically improved the healing process of photoaging skin. Thus, we provide a strong rationale for a combined use of ADSCs and fractional CO2 laser in treatment of photoaging skin in clinic in the future. Moreover, we provided evidence that the Wnt/β-catenin signaling pathway may contribute to the activation of dermal fibroblast by the transplantation of ADSCs in both vitro and vivo experiment.

Highlights

  • It is well established that adipose-derived stem cells (ADSCs) produce and secrete cytokines/growth factors that antagonize UV-induced photoaging of skin

  • One primary mechanism by which UV causes photoaging is the suppression of synthesis of type I procollagen (COLI) [2,3], a major structural protein in the skin connective tissue which is mainly produced by the fibroblasts located within dermis

  • We investigated the impacts of ADSCs alone or in combination with fractional CO2 laser on photoaging skin caused by UV irradiation in the animal model, and report here that ADSCs improved photoaging skin recovery at least partially via restoring the expression of β-catenin and Wnt3a, and the combined use of ADSCs transplantation and fractional CO2 laser synergistically benefited the UV-irradiation induced skin damage compared with either individual treatment

Read more

Summary

Introduction

It is well established that adipose-derived stem cells (ADSCs) produce and secrete cytokines/growth factors that antagonize UV-induced photoaging of skin. One primary mechanism by which UV causes photoaging is the suppression of synthesis of type I procollagen (COLI) [2,3], a major structural protein in the skin connective tissue which is mainly produced by the fibroblasts located within dermis. The TGF-β/Smad pathway is the major regulator of synthesis of several components of the extracellular matrix, including type I and type III collagen by skin fibroblasts. It is TGF-β stimulates fibroblast proliferation in the dermis to enhance collagen synthesis [4,5,6,7]. Among known Wnt protein family members, Wnt1 [12,13], Wnt3a [14] and Wnt8 [15] are able to activate classical Wnt-β-catenin-LEF/TCF pathway, and Wnt3a upregulates TGF-β in a β-catenin dependent manner through Smad, and inducts the differentiation of myofibroblast [16]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call