You are on page 1of 3

Accepted: 12 September 2017

DOI: 10.1111/exd.13451

LETTERS TO THE EDITOR

Exosomes from human adipose-­derived stem cells promote


proliferation and migration of skin fibroblasts
Abstract 2 | QUESTIONS ADDRESSED
This study was undertaken to evaluate whether exosomes from
human adipose-­derived stem cells (ASC-­exo) can stimulate the re- We asked whether ASC-­derived exosomes can stimulate the skin der-
generation of human dermal fibroblasts (HDFs). Immunoblotting mal fibroblasts’ proliferation and migration, and by what mechanism
and FACS analyses showed that ASC-­exo was positive for exosome these exosomes play such functions.
markers. Fluorescence tracking revealed that the contents of ASC-­
exo were transferred into the HDFs. ASC-­exo treatment also stimu-
lated the proliferation and migration of HDFs in a dose-­dependent 3 | EXPERIMENTAL DESIGN
manner. Similarly, the expression levels of genes involved in skin
cell proliferation were increased by ASC-­
exo. Microarray analy- See supplementary information.
sis showed an enrichment of microRNAs that have regenerative
function. We suggest that the ASC-­
exo can stimulate skin cell
proliferation. 4 | RESULTS

Transmission electron microscopy (TEM) and nanoparticle tracking


1 | BACKGRO UND analysis (NTA) showed that ASC-­exo isolated from the supernatants

It has been well reported that adipose tissue-­d erived stem cell
(ASC) secretes various soluble factors, that is growth factors,
cytokines, exosomes that can rescue damaged cells. [1] Exosome,
a 30-­ to 150-­n m-­s ized extracellular vesicle that is secreted from
most cell types, is formed within endosomal compartments and
released into the extracellular milieu. [2] It has been demon-
strated that exosomes possess similar functional properties of
mesenchymal stem cells (MSCs) from which they are derived, [3]
and due to such functions, exosomes are regarded now as a
communicator between tissues. [4] Functionally, exosomes can
reduce the immune recognition, so that the integrity of cell
membrane can be maintained. [5] A study of stem cell transplan-
tation therapy demonstrated that the role of MSCs in cell-­t o-­
cell communication was exerted through a paracrine mechanism
and that exosomes play a major role in this process. [6] Other
findings also described that the conditioned media of ASC
(ASC-­C M) [7] or exosomes [8] were able to promote the migration
of dermal fibroblasts during the process of wound healing. [7] F I G U R E   1   Proliferation and migration effect of human ASC-­exos
on HDFs. (A) Gene expression profiling of collagen type 1, elastin,
Although the mechanism of the exosomes’ role on proliferation
KGF, CD34 and VEGF in HDFs treated with vehicle and those treated
and migration of fibroblasts was demonstrated, [8] no study has either 5 or 10 μg/mL of ASC-­exo (*P < .05). (B) Assessment of the
been conducted to identify the expressional changes of microR- migratory capacity of HDFs after being incubated with 5 or 10 μg/mL
NAs, which play major role in various cellular responses, includ- of ASC-­exo. Assays were repeated three times. (C) The effect of ASC-­
ing proliferation. exo treatment on HDF cell cycle (*P < .05) (N = 3)

This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any
medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
© 2017 The Authors. Experimental Dermatology Published by John Wiley & Sons Ltd

1170  |  wileyonlinelibrary.com/journal/exd Experimental Dermatology. 2018;27:1170–1175.


LETTERS TO THE EDITOR |
      1171

for 24 hours, of PKH26-­labelled ASC-­exos with HDFs, it was found


that red fluorescence of PKH26 was localized within the cytoplasm
of HDFs (Fig. S1D), showing that ASC-­exo can be internalized into
HDFs. Also, quantitative real-­time PCR showed that the expression
of genes related with skin regeneration, such as CD34, collagen type
1, elastin and keratinocyte growth factor (KGF), was increased after
being treated with ASC-­exos in a dose-­dependent manner (Figure 1A).
We also found that ASC-­exo stimulated the proliferation of HDFs
and that such effect was significantly higher in 10 μg/mL compared
with those treated with 5 μg/mL (Fig. S2). In line with this finding, we
found that the migration of HDFs was increased in a similar manner
(Figure 1B). Similarly, cell cycle analysis by FACS demonstrated that
ASC-­exo treatment significantly increased the population of those
within S-­phase (Figure 1C).
We next conducted a comprehensive analysis of microRNA ex-
pression between ASC and ASC-­exo by microarray. We selected the
microRNAs expressed differentially (>2-­folds, Figure 2A) and found
that 292 among 333 microRNAs were differentially expressed (199
and 93 upregulated and downregulated, respectively, in ASC-­
exo)
(Figure 2B). These differentially expressed microRNAs were grouped
based on function, as referenced with miRTarBase (http://http://mir-
tarbase.mbc.nctu.edu.tw/). The 718 validated gene targets of top 10
upregulated miRNAs were grouped according to the gene ontology
(GOTERM_BP_FAT) with an enrichment EASE score < 0.05 and 10
more than number of target genes (Figure 2C). The role of ASC-­exo
microRNAs with their corresponding validated target genes was sche-
matically represented in Figure 2D. From this results, it can be con-
cluded that exosomes from ASC contain microRNAs that can inhibit
F I G U R E   2   (A) Scatter plot showing the microRNAs that
genes including NPM1, PDCD4, CCL5 and NUP62, thereby contribut-
are differentially expressed between ASC and ASC-­exo. The
ing to the regeneration of skin fibroblasts by stimulating the prolifera-
x-­axis and y-­axis represent the normalized expression level of
ASC ASC-­exo, respectively. Each miRNA, having a fold change tion of dermal fibroblasts.
more than two, is represented by a dot. (B) Heatmap analysis
of microarray results showing hierarchical clustering of 292
differentially expressed miRNAs between ASC and ASC-­exo. 5 | CONCLUSION
Red (high) or green (low) colours indicate upregulated or
downregulated microRNA intensity. Mean signal values were
background-­corrected and 0. Genes showing a minimum of 2-­fold Recent studies have demonstrated that exosomes are involved in the
changes with P value <.05 at the 95% confidence level were paracrine activity of stem cells.[9] This study demonstrates that the
considered as significant. (C) Biological processes abundantly exosomes derived from ASCs play a prosurvival role in human dermal
expressed by microRNAs. The 718 validated target genes fibroblasts by stimulating its cell cycle and that microRNAs that are
of the top 10 upregulated miRNAs in ASC-­exo are grouped
known to have regenerative potentials were enriched in ASC-­exo. Our
according to the gene ontology (GOTERM_BP_FAT) with an
finding may be of useful for developing novel therapeutic methods for
enrichment EASE score < 0.05 and more than 10 target genes.
(D) Schematic representation of miRNAs with their validated the enhancing wound healing.
targets contributing to reduced cellular senescence and, ageing-­
associated genes, deregulated cell growth, ageing-­related disease.
NPM1, nucleophosmin; PDCD4, programmed cell death 4; CCL5, CO NFL I C T O F I NT ER ES T
C-­C motif chemokine ligand 5; NUP62, nucleoporin 62 kDa
The authors have no conflict of interest to declare.
of the ASC culture showed the typical morphology of exosomes that
are within the range of 70-­150 nm (Fig. S1A). Western blot analysis
AU T HO R CO NT R I B U T I O NS
revealed that ASC-­exo expresses exosome markers HSP70, CD63
and CD9 (Fig. S1B), while these markers were absent in ASC lysate. EW Choi performed the research, designed the study, analysed the
Flow cytometry analysis also showed that ASC-­exos were positive for data and wrote the manuscript. MK Seo, EY Woo, SH Kim and EJ.
CD9, CD63 and CD81 (Fig. S1C). We next asked whether the con- Park designed the study and performed the research. S Kim edited
tents of ASC-­exos could be transmitted to HDFs. After co-­incubation, the manuscript.
|
1172       LETTERS TO THE EDITOR

Keywords [6] S. Raik, A. Kumar, S. Bhattacharyya, Biotechnol. Appl. Biochem. 2017,


extracellular vesicle, microRNA array, skin regeneration, wound https://www.ncbi.nlm.nih.gov/pubmed/28321921. [Epub ahead of
print].
healing
[7] W. S. Kim, B. S. Park, J. H. Sung, J. M. Yang, S. B. Park, S. J. Kwak, J. S.
Park, J. Dermatol. Sci. 2007, 48, 15.
[8] L. Hu, J. Wang, X. Zhou, Z. Xiong, J. Zhao, R. Yu, F. Huang, H. Zhang, L.
O RCI D
Chen, Sci. Rep. 2016, 6, 32993.
Sue Kim  http://orcid.org/0000-0002-1818-0551 [9] G. D. Kusuma, J. Carthew, R. Lim, J. E. Frith, Stem Cells Dev. 2017, 26,
617.
Eun Wook Choi
Min Koo Seo
Eun Young Woo S U P P O RT I NG I NFO R M AT I O N
Suk Hyung Kim Additional Supporting Information may be found online in the
Eun Joo Park ­supporting information tab for this article.
Sue Kim
Prostemics Research Institute, Seongdong-gu, Seoul, South Korea FIGURE S1 Characterization of human ASC-­exos. (A) A representative
Correspondence image of the size-­distribution plot of NTA, and the inlet is an image of
Sue Kim, Stem Cell Center, ASAN Institute for Life Sciences, ASAN TEM analysis. (B) Detection of HSP70, CD63, and CD9 in ASC-­exos by
Medical Center, Songpa-gu, Seoul, South Korea. Western blot analysis. (C) Flow cytometry analysis showing that ASC-­
Email: sue2949@gmail.com exo are positive for CD9, CD63 and CD81. (D) The uptake of internal
contents of ASC-­exo into HDFs. PKH-­26-­labled ASC-­exos were incu-

REFERENCES bated with HDFs for 24 h and the fluorescence was detected
FIGURE S2 Effect of ASCs derived exosomes on the cell growth of
[1] W. S. Kim, B. S. Park, S. H. Park, H. K. Kim, J. H. Sung, J. Dermatol. Sci.
HDFs. HDFs were treated with 5 μg/mL or 10 μg/mL of ASCs derived
2009, 53, 96.
[2] A. Marote, F. G. Teixeira, B. Mendes-Pinheiro, A. J. Salgado, Front exosomes (ASC-­Exo) for 48 h and HDFs proliferation was determined
Pharmacol. 2016, 7, 231. by MTS assay (n = 3). Data are expressed as the means ± SD. Asterisk
[3] R. C. Lai, F. Arslan, M. M. Lee, N. S. Sze, A. Choo, T. S. Chen, M. Salto- indicates a statistically significant difference between two groups of
Tellez, L. Timmers, C. N. Lee, R. M. El Oakley, G. Pasterkamp, D. P. de
cells (P < .05)
Kleijn, S. K. Lim, Stem Cell Res. 2010, 4, 214.
[4] N. Vyas, J. Dhawan, Cell. Mol. Life Sci. 2017, 74, 1567. TABLE S1 Primers used in the real-­time RT-­PCR analysis
[5] P. Vader, E. A. Mol, G. Pasterkamp, R. M. Schiffelers, Adv. Drug Deliv. APPENDIX S1 Experimental design
Rev. 2010, 106, 148. DATA S1 ASC and ASC-exo miRNA array results

Accepted: 26 September 2017

DOI: 10.1111/exd.13453

RNA-­seq analysis of Lgr6+ stem cells and identification of an


Lgr6 isoform
Abstract 1 | INTRODUCTION
We studied Lgr6+ stem cells in experimental UV carcinogenesis in
hairless mice. For further characterization through RNA-­seq, these We investigated the fate of a novel class of Lgr6+ proliferative stem
stem cells were isolated by FACS from transgenic hairless mice bear- cells in UV carcinogenesis in hairless mice, a model closely mimick-
ing an EGFP-Ires-CreERT2 reporter cassette inserted into exon 1 of ing the development of cutaneous squamous cell carcinoma in man.[1]
the Lgr6 gene (purity confirmed by human ERT2 expression). Between Using transgenic mice with an EGFP-Ires-CreERT2 reporter cassette
Lgr6/EGFP+ and Lgr6/EGFP− basal cells (Tg/wt), 682 RNAs were dif- inserted into exon 1 of Lgr6, we found—similar to what was found in
ferentially expressed, indicating stemness and expression of cancer-­ haired mice[2]—that Lgr6/EGFP+ stem cells appeared in large clusters in
related pathways in Lgr6/EGFP+ cells. We discovered that suspected the interfollicular epidermis (IFE) and in very small clusters in the rem-
“Lgr6 null” mice (Tg/Tg) expressed RNA of an Lgr6 isoform (delta-­Lgr6, nants of hair follicles of hairless mice.[3] Contrary to our expectation,
lacking 74 N-­terminal aa) which could be functional and explain the the Lgr6/EGFP+ cells dwindled in numbers in the hyperplastic IFE under
lack of a phenotype. chronic UV exposure and did not become tumor-­initiating cells (TICs).

You might also like