Skip to main content
Journal of Histochemistry and Cytochemistry logoLink to Journal of Histochemistry and Cytochemistry
. 2014 Apr;62(4):276–285. doi: 10.1369/0022155414520710

Immunohistological Localization of Endogenous Unlabeled Stem Cells in Wounded Skin

Song Hong 1,1,, Bhagwat V Alapure 1,1, Yan Lu 1, Haibin Tian 1, Quansheng Wang 1
PMCID: PMC3966289  PMID: 24399040

Abstract

Various types of endogenous stem cells (SCs) participate in wound healing in the skin at different anatomical locations. SCs need to be identified through multiple markers, and this is usually performed using flow cytometry. However, immunohistological identification of endogenous stem cells in the skin at different anatomical locations by co-staining multiple SC markers has been seldom explored. We examined the immunohistological localization of four major types of SCs in wounded skin by co-staining for their multiple markers. Hematopoietic SCs were co-stained for Sca1 and CD45; mesenchymal SCs for Sca1, CD29, and CD106; adipose SCs for CD34, CD90, and CD105; and endothelial progenitor cells and their differentiated counterparts were co-stained for CD34, Tie2, and von Willebrand factor. We found Sca1+CD45+ SCs in the epidermis, dermis and hypodermis of wounded skin. Sca1+CD29+ and Sca1+CD106+ mesenchymal SCs, CD34+CD105+, CD34+CD90+, and CD90+CD105+ adipose SCs, as well as CD34+Tie2+ endothelial progenitor cells were also located in the epidermis, dermis, and hypodermis. This study demonstrates the feasibility of using immunohistological staining to determine the location of SCs in wounded skin and the intracellular distribution of their molecular markers.

Keywords: endogenous stem cell, hair follicle, mesenchymal, adipose, endothelial progenitor, wound healing, immunohistological, confocal microscopy, localization

Introduction

Stem cells (SCs) are so-called because of their unique ability to self-renew and differentiate into a variety of cell lineages (Barrilleaux et al. 2006; He et al. 2009; Phinney and Prockop 2007; Raff 2003; Smith et al. 2007). Wounding recruits and activates SCs to home to wounded skin. SCs produce factors that modulate healing and proliferate and differentiate to replenish lost cells at the site. The typical endogenous SCs that contribute to skin wound healing include mesenchymal SCs (MSCs), adipose SCs (ASCs), endothelial progenitor cells (EpCs), hematopoietic SCs (HSCs), and epidermal SCs (Barrilleaux et al. 2006; Fathke et al. 2004; Hamou et al. 2009; Jeyarajah et al. 1986; Phinney and Prockop 2007; Plikus et al. 2012; Prockop 2009; Raff 2003; Seppanen et al. 2013; Smith et al. 2007). MSCs and ASCs promote healing and angiogenesis mainly by producing paracrine factors that act on neighboring cells; however, they can also differentiate into skin cells, including epithelial cells, endothelial cells, and/or fibroblasts (Badiavas et al. 2003; Brittan et al. 2005; Chen et al. 2008; Fathke et al. 2004; Hocking and Gibran 2010; Hong et al. 2013; Javazon et al. 2007; Jin et al. 2010; Khan et al. 2011; Kwon et al. 2008; Stepanovic et al. 2003; Tian et al. 2011; Wu et al. 2007). EpCs differentiate into endothelial cells during skin repair (Albiero et al. 2011; Nie et al. 2011; Ong and Sugii 2013) whereas HSCs differentiate into leukocytes (Jeyarajah et al. 1986).

One of the major hurdles hindering the elucidation of the precise roles of different SCs in wound healing is a lack of means to identify and locate diversified SCs in hair follicles, dermis, and hypodermis of wounded skin. In principle, each type of SC can be identified by staining for specific markers. SCs are readily identifiable using flow cytometry (Albiero et al. 2011; Bourin et al. 2013; Jensen et al. 2009; Rebelatto et al. 2008; Rostovskaya and Anastassiadis 2012; Sasaki et al. 2008; Tobita et al. 2011), which requires relatively large amounts of tissue to obtain sufficient numbers of cells (105-106 cells dispersed from as much as 1 g of tissue) (Marsee et al. 2010). In contrast, histological analysis can be performed on as few as tens of milligrams of tissue. Additionally, flow cytometry does not provide the location (within tens of micrometers) of SCs in wounded skin, or the cellular morphology or intracellular distribution of molecules within the cells. Western blotting and PCR can also be used to detect SCs in skin, but these techniques are also limited by their inability to localize cells anatomically.

Florescence-tagged SCs, infused or transplanted into animals, will mimic the endogenous untagged SCs and provide an opportunity to study the location of the cells. However, the validity of these results remains a concern, as the tags or tagging process may alter the behavior of the proteins or the SCs themselves (Michaelson and Philips 2006; Skube et al. 2010; Taghizadeh and Sherley 2008). Transgenic reporter mouse strains and lineage tracing experiments are well-established, definitive approaches to track SCs (Snippert and Clevers 2011). These tools are continuously used in wide range of SC research so long as the needed transgenic reporter mouse strains or lineage tracing animals are available. If not, the animals need to be first generated. As an alternative, an immunohistological method combined with confocal microscopy with multiple wavelength excitation and signal acquisition may offer a way to localize endogenous SCs in tissues, using less tissue samples as compared with other methods.

Protein markers expressed by SCs are critical for SC localization in wounded skin. Stem cell antigen 1 (Sca1) and CD45 are expressed by HSCs and are thus commonly used for HSC identification (Granick et al. 2013; Hamou et al. 2009; Meindl et al. 2006). In addition, the GFP-labeled MSC lineage populations have been used to examine epidermis and dermis wound repair processes (Borue et al. 2004; Harris et al. 2004; Tian et al. 2011; Wu et al. 2010). MSCs express high levels of CD29 or CD106 and Sca1, and therefore immunohistological co-staining of CD29 or CD106 and Sca1 can be used to localize MSCs in wounded skin. CD34, CD90, and CD105 are expressed by ASCs, but absent or weakly expressed by MSCs, and are thus considered as good markers for detecting ASCs (De Francesco et al. 2009; Festa et al. 2011; Ong and Sugii 2013; Rostovskaya and Anastassiadis 2012; Tobita et al. 2011; Zimmerlin et al. 2011). EpCs are usually identified by multiple markers, including CD34 and Tie2, using flow cytometry (Chen et al. 2013; Sheridan et al. 2006). Additionally, EpC differentiation into endothelial cells can be traced using CD34 plus identification of the endothelial marker, von Willebrand factor (vWF) (Albiero et al. 2011; Chen et al. 2013; Sheridan et al. 2006). In this report, we devised an immunohistological approach to localize and discriminate the four main types of endogenous stem cells (HSCs, MSCs, ASCs, and EpCs) in wounded skin by staining different combinations of antigens prominently present in the each type of SC. The resulting scheme should facilitate analysis of homing, recruitment flux, and fate of endogenous stem cells during wound healing under various pathophysiological and pharmacological conditions.

Materials & Methods

Animals and Procedures: Full-Thickness Incisional Wounding

Established protocols were used (Ashcroft et al. 1997), and studies were conducted in conformity with the Public Health Service Policy on Humane Care and Use of Laboratory Animals. Animal protocols were approved by the Institutional Animal Care and Use Committee and Institutional Review Board of Louisiana State University Health Sciences Center, New Orleans; and followed the National Institutes of Health guidelines for experimental animal use. For wounding, mice (~19 g body weight, C57BL/6, 10 weeks old) (Jackson Laboratories, Bar Harbor, ME) were anesthetized by intraperitoneal injection of ketamine hydrochloride (0.2 mg/Kg body weight) and xylazine (0.4 mg/Kg body weight). The hair on the dorsal side was removed by Veet depilating cream, and the area was cleaned with 70% ethanol. Two full-thickness incisions (each 20-mm length) on the dorsal side were created symmetrically across the midline between the 5th thoracic vertebrae and the ischium bone. Incisions were closed with two sutures (Ethilon 6-0). The sutures were removed at day 5 post-wounding. Hair was similarly removed from sham-treated controls but incisions were not made.

Collection and Immunohistology of Wounded Skin

At day 10 post-wounding, mice (n=6/group) were perfused via cardiac puncture with PBS−/− (without calcium and magnesium) at a flow rate of 5 ml/min for 5 min under anesthesia, and then sacrificed. Wounds and a 2-mm margin of normal surrounding skin were excised, fixed in 4% paraformaldehyde (4 hr, ~23C), incubated in 30% sucrose (12 hr, 4C), embedded in OCT, and then stored at -80C, as reported previously (Lu et al. 2010; Tian et al. 2010). Unwounded skin from the same position on the dorsal side from sham controls was similarly processed. Serial sagittal cryosections (7-µm-thick) were cut using a Leica CM3050s cryostat microtome (Buffalo Grove, IL) and transferred to Superfrost slides (Fisher Scientific, Pittsburgh, PA). Cryosections were treated sequentially as follows: incubated in methanol/acetone (1/1) (20 min, -20C), washed with 1× phosphate-buffered saline (PBS) for 10 min for the removal of OCT; incubated in citrate buffer (20 min, 90–100C), then in blocking buffer [5% Goat/Donkey serum (MP Biomedical, Solon, OH), and 0.3% trition-X-100 (Fluka, St. Louis, MO)] for 1 hr, ~23C; incubated with two anti-mouse primary antibodies that were raised from two different hosts and diluted as in Table 1 (4C, 12 hr), washed thrice with 1× PBS (10 min, ~23C), incubated with two secondary antibodies (labeled with Alexa Fluor 488 (green) and Alexa Fluor 568 (red), raised from compatible hosts) at 23C for 1 hr, and then washed thrice with 1× PBS (10 min, ~23C). Nuclei were counterstained with 4′-6-diamidino-2-phenylindole (DAPI, 1:2000, 10 min) (Life Technologies, Grand Island, NY). Slides were rinsed with 1× PBS (10 min, ~23°C, 3 times) to remove extra DAPI, mounted using mounting media (Vector Laboratories, Burlingame, CA), and imaged using a Zeiss 510 Meta confocal laser-scanning microscope (Carl Zeiss, Jena, Germany) with a 40× oil immersion objective (numerical aperture: 1.25). The images were analyzed by Zen 2011 software (Carl Zeiss) and annotated for presentation using Adobe Photoshop (Adobe Systems, Inc. San Jose, CA).

Table 1.

List of Antibodies and Dilutions.

Target Host Dilution Supplier Cat No.
CD34 Rat 1:100 eBioscience, San Diego, CA 14-0341
CD34 Goat 1:50 Santa Cruz Biotechnology, San Diego, CA sc-7045
CD45 Rabbit 1:50 Santa Cruz Biotechnology sc-25590
CD29/Integrin β1 Rabbit 1:50 Santa Cruz Biotechnology sc-8978
CD90/Thy1 Rat 1:50 Abcam, Cambridge, MA ab44898
CD105/Endoglin Rabbit 1:50 Santa Cruz Biotechnology sc-20632
CD106/VCAM1 Rabbit 1:50 Santa Cruz Biotechnology sc-8304
Sca1 Rat 1:50 eBioscience 14-5981
Tie2 Rabbit 1:50 Santa Cruz Biotechnology sc-9026
vWF Rabbit 1:50 Santa Cruz Biotechnology sc-14014

Results

Immunohistological Localization of HSCs in Wounded Skin

Localization of typical HSCs was studied by staining cryosections of wounded skin for specific HSCs markers, CD45 and Sca1 (Table 2; Fig. 1). Confocal images revealed Sca1+CD45+ HSCs in the epidermis (Fig. 1A), dermis (Fig. 1A, 1B) and hypodermis (Fig. 1C) of wounded skin. Sca1+CD45+ HSCs were also found in the dermis and hypodermis (Fig. 1E, 1F), but not in epidermis (Fig. 1D) of unwounded skin. These results show that immunofluorescence staining and confocal imaging can uncover the anatomical locations of Sca1+CD45+ in wounded skin.

Table 2.

Specific Marker Expression in Different Types of Stem Cells.

Stem Cells (SCs) Specific SC marker References
Hematopoietic SCs (HSCs) Sca1, CD45 (Fathke et al. 2004; Granick et al. 2013; Hamou et al. 2009; Seppanen et al. 2013)
Adipose SCs (ASCs) CD34 (90%), CD90 (99%), and CD105 (97%) (Baer et al. 2013; Festa et al. 2011; Ong and Sugii 2013; Tobita et al. 2011; Zhang et al. 2013)
Mesenchymal SCs (MSCs) Sca1 (85.3%), CD29 (90.7%), and CD106 (92.1%) (Bourin et al. 2013; Houlihan et al. 2012; Rostovskaya and Anastassiadis 2012; Yang et al. 2013)
Endothelial progenitor cells (EpCs) CD34+Tie2; CD34+vWF (Albiero et al. 2011; Felcht et al. 2012; Neumuller et al. 2006; Untergasser et al. 2006)

Note: The percentage (%) refers to the relative population of the SCs that express the marker.

Figure 1.

Figure 1.

Localization and morphological observation of endogenous hematopoietic stem cells (HSCs) in wounded skin. Sca1+CD45+ cells (white short arrow) and Sca1CD45+ cells (white arrowhead) resided in the epidermis (A), dermis (B) and hypodermis (C) of wounded skin. The Sca1+CD45+ and Sca1CD45+ cells were also observed in the dermis (E) and hypodermis (F) of unwounded skin. (G) Image showing the non-specific binding of the secondary antibodies. Nuclei were counterstained with DAPI (blue). Abbreviations: bu-bulge; ho-hypodermis; d-dermis; e-epidermis; hf-hair follicle; dashed line (—)-border of dermis and epidermis. The large yellow arrow indicates the site of the incision wound, and marks the direction from the epidermis to the hypodermis. Asterisk (*) indicates non-specific binding of secondary antibodies. Bar, 38 µm.

Immunohistological Identification and Localization of Mesenchymal Stem Cells in Wounded Skin

MSCs are usually identified by flow cytometric analysis of multiple markers including Sca1, CD29, and CD106 (Table 2) (Rostovskaya and Anastassiadis 2012; Tobita et al. 2011). However, histological localization of endogenous SCs in situ is usually done by single-marker staining (Hong et al. 2009). To explore the feasibility of histological localization and morphological observation of multiple types of endogenous SCs in wounded skin, we double-stained skin cryosections with combinations of MSC markers Sca1, CD29, and CD106 (Fig. 2) (Rostovskaya and Anastassiadis 2012; Tobita et al. 2011). Sca1+CD29+ and Sca1+CD106+ MSCs were observed in wounded epidermis (Fig. 2A-2C, 2J-2L) and dermis (Fig. 2D-2F, 2M-2O) and in unwounded epidermis and dermis (Fig. 2G-2I & 2P-2R). The increased numbers of positively stained cells in the epidermis and dermis around the wound site suggest that endogenous MSCs may regenerate skin cells to heal the wound.

Figure 2.

Figure 2.

Localization of endogenous MSCs in wounded skin using combinations of typical MSC markers: (A, D, G) CD29 (red; red arrows); (J, M, P) CD106 (red; red arrows), and (B, E, H, N, Q) Sca1 (green; green arrows). (C, F, I, L, O, R) Merged images. White arrowheads show (A-F) CD29+Sca1+ cells and (J-O) CD106+Sca1+ cells in wounded skin epidermis and dermis. Fewer CD106+Sca1+, Sca1+, or CD106+ cells were present in unwounded skin (G-I; P-R) as compared with wounded skin (A-F; J-O). Negative staining (S-U) shows non-specific binding of secondary antibodies. Nuclei were counterstained with DAPI (blue). Abbreviations: e-epidermal; d-dermis; hf-hair follicle; dash line(—)-border of dermis and epidermis. The large yellow arrow indicates the site of the incision wound, and marks the direction from the epidermis to the hypodermis. Asterisk (*) indicates non-specific binding of secondary antibodies. Bars, (C & L) 42 µm; (F, I, O, R, U) 38 µm.

Adipose Stem Cells in Wounded Skin Localized by Immunohistological Co-staining of their Markers

ASCs are often identified by flow cytometric analysis using multiple markers, such as CD34, CD90, and CD105 (Table 2) (Zhang et al. 2013). Based on the flow cytometry method, we double-stained skin cryosections with combinations of ASC markers (Fig. 3) (Chen et al. 2007; Rostovskaya and Anastassiadis 2012; Tobita et al. 2011). CD34+CD90+ (Fig. 3A-3D), CD34+CD105+ (Fig. 3E, 3G, 3H), and CD90+CD105+ (Fig. 3I-3L) ASCs were observed in the dermis and hypodermis of wounded skin. Higher proportions of each ASC subgroup existed in the wounded dermis (Fig. 3A, 3E, 3I) and hypodermis (Fig. 3C, 3G, 3K) as compared with the unwounded dermis (Fig. 3B, 3F, 3J) and hypodermis (Fig. 3D, 3H, 3L). Cells that stained positively for only one marker are unlikely to be ASCs (Fig. 3).

Figure 3.

Figure 3.

Localization of endogenous adipose stem cells (ASCs) in wounded skin using combinations of specific ASC markers: CD34, CD90, and CD105. Each cryosection was co-stained for two of the three ASC markers. (A-D) CD34 (red; red arrows) and CD90 (green; green arrows); (E-H) CD34 (red; red arrows) and CD105 (green; green arrows); (I-L) CD90 (green; green arrows) and CD105 (red; red arrows). Cells in the dermis and hypodermis positive for two markers are regarded as ASCs. Double-positive cells are yellow and indicated by white arrowheads. Negative staining (M) shows non-specific binding of the secondary antibodies. Nuclei were counterstained with DAPI (blue). Abbreviations: d-dermis, ho-hypodermis. The large yellow arrow indicates the site of the incision wound, and marks the direction from the epidermis to the hypodermis. Asterisk (*) indicates non-specific binding of secondary antibodies. Bar, 28 µm.

Localization of Endothelial Progenitor Cells and their Differentiated Counterparts in Wounded Skin

CD34 and Tie2 was used to identify EpCs (Table 2), and CD34 and vWF to identify endothelial cells differentiated from EpCs via flow cytometry (Albiero et al. 2011; Chen et al. 2013; Padfield et al. 2010; Sheridan et al. 2006). vWF is a marker of endothelial cells (Chen et al. 2013; Sheridan et al. 2006). Therefore, we stained the skin sections for CD34 (green) and Tie2 (Fig. 4A, 4B) or vWF (Fig. 4C, 4D) (red). The CD34+Tie2+ cells (Fig. 4A, shown by white arrowheads) are EpCs, and CD34+vWF+ cells (Fig. 4C,D) (white arrowhead) are endothelial cells differentiated from EpCs. These histological images demonstrate that there are more CD34+Tie2+ EpCs and CD34+vWF+ endothelial cells in wounded skin compared with unwounded skin (Fig. 4).

Figure 4.

Figure 4.

Localization of endothelial progenitor cells (EpCs) and differentiated endothelial cells in wounded skin. (A-B) EpCs were localized by co-staining CD34 and Tie2. (C-D) The differentiation of EpCs to endothelial cells was identified by co-staining CD34 and vWF. Negative staining image (E) shows unspecific binding of secondary antibodies. Double-positive cells are yellow and indicated by white arrowheads. Nuclei were counterstained with DAPI (blue). Abbreviations: e-epidermal; d-dermis; hf-hair follicle; bu-bulge; dash line(—)-border between epidermis and dermis. The large yellow arrow indicates the site of the incision wound, and marks the direction from the epidermis to the hypodermis. Asterisk (*) indicates non-specific binding of secondary antibodies. Bar, 38 µm.

Discussion

A variety of endogenous SCs participate in skin wound healing (Daley 2012; Plikus et al. 2012; Prasongchean and Ferretti 2012). However, the anatomical distribution, morphology, and intracellular molecular arrangements of different SCs during skin healing in vivo cannot be readily assessed by flow cytometry and are not well defined. We undertook a histological approach to identify properties of SCs in skin wound healing. We have identified and localized the major endogenous SCs (HSCs, MSCs, ASCs, and EpCs) in wounded skin by immunohistological analysis of sections co-stained for multiple markers.

The anatomical niche for endogenous MSCs in different parts of skin (epidermis, dermis, and hypodermis) is still not well understood. There are reports, however, indicating that MSCs are present in follicular connective sheath tissue and in the papilla of the dermis (Hoogduijn et al. 2006; Kruse et al. 2006; Lako et al. 2002; Richardson et al. 2005). It has also been reported that 4% of transplanted tagged-MSCs engraft into the epidermis (Borue et al. 2004; Harris et al. 2004). Other studies have provided evidence for the localization of dermal MSCs in skin independent of the hair follicles (Chen et al. 2007; Lorenz et al. 2008; Toma et al. 2005). All studies have been performed by flow cytometry in association with complementary immunohistological staining of single MSC markers (Hong et al. 2009). The flow cytometry used in these studies is a limiting factor, and cannot localize MSCs in the skin. Staining of only one marker may lead to a misidentification of SCs because of the potential off-target staining resulting from the expression of a single marker in non-MSCs. We observed that Sca1+CD29+ and Scal+CD106+ cells were co-localized in the epidermis and dermis, but that there were far fewer dual-positive cells than single-positive Scal+, CD29+, or CD106+ cells (Fig. 2). Sca1 is expressed by hematopoietic SCs. CD29 and CD106 are also expressed by epithelial cells (Hong et al. 2009; Kim et al. 2002). Therefore, to avoid mistakenly identifying HSCs, epithelial cells, or other cell types as MSCs, MSCs should be identified and localized in skin sections based on co-staining with the combination of Sca1 and CD29 or Sca1 and CD106. Similarly, cells expressing both Sca1 and CD29 are Sca1+CD29+ HSCs; however, cells expressing only one of Sca1 or CD45 are unlikely to be so.

Sca1 is a member of the Ly6A superfamily of glycophosphatidylinositol (GPI)-anchored membrane proteins (Holmes and Stanford 2007), involved in SC self-renewal, fate decisions, and lipid raft signaling via weak protein-protein interaction in HSCs (Holmes and Stanford 2007; Okada et al. 1992; Spangrude et al. 1988). CD45 is a hematopoietic cell marker present on the surface of HSCs and plays a critical role in survival, expansion, differentiation and migration of HSCs during the process of hematopoiesis (Kiel and Morrison 2008; Raaijmakers and Scadden 2008). In the present study, endogenous Sca1+CD45+ HSCs were identified in the epidermis, dermis, and hypodermis of wounded skin, but not in the epidermis of unwounded skin, implying a role for HSCs in wound healing of the epidermis. Further, Sca CD45+ cells were located in the epidermis, dermis, and hypodermis in unwounded and wounded skin. Our observation of HSCs in skin wounds is consistent with the published research conducted by other groups (Etich et al. 2013; Meindl et al. 2006; Passier and Mummery 2003).

CD34 is a cell-surface glycoprotein thought to mediate the attachment of SCs to the extracellular matrix or directly to stromal cells (Nielsen and McNagny 2008). This implies that CD34 may play a role in the reparative function of CD34-containing SCs. We have localized ASCs to the dermis and hypodermis by co-staining with CD34, CD105, and CD90, which are reported to be maintained at high levels in ASCs (Table 2) (Chen et al. 2010; Folgiero et al. 2010; Tobita et al. 2011) and which do not occur, or occur at very low levels, in MSCs (Zhang et al. 2013). On the other hand, ASCs do not express, or express at very low levels, the MSC markers Sca1, CD29, and CD106 (Chen et al. 2010; Ong and Sugii 2013; Rostovskaya and Anastassiadis 2012; Tobita et al. 2011; Vaculik et al. 2012).

This study was aimed at differentiating among SC populations of ASCs, MSCs, and HSCs, and assessing their involvement in wounded skin qualitatively. To this end, we have explored the localization of the major endogenous stem cells—HSCs, MSCs, ASCs, and EpCs—by co-staining multiple typical markers of each type of SC in sections of wounded murine skin. We demonstrated the feasibility of immunohistological co-staining for localizing endogenous, untagged SCs in wounded skin, and for observing the intracellular distribution of the proteins. Although this report is limited by the small number of SC markers tested, additional SC markers could be assessed to confirm the localization of SCs, and to identify potential new SC phenotypes that may play roles in cutaneous wound healing. Staining of multiple SC markers of skin sections with more than three colors could be developed in combination with advanced confocal microscopic imaging. If successful, such multiplexed staining could offer more definitive localization and morphological understanding of SCs in wounded skin, which could in turn facilitate the development of new diagnostic methods for the treatment of chronic wounds. Our approach is complementary to methodology that uses the transgenic reporter mouse strains or lineage tracing experiments. For example, we can use this relative easy immunohistological approach to make the initial observation or narrow down the studies, then use the transgenic reporter mouse strains and lineage tracing experiments to make a final observation. Alternatively, if this type of immunohistological approach yields potential new SC markers, then the markers can be selected for lineage tracing experiments or for the development of new transgenic reporter mouse strains for definitive discovery. This report could provide a complementary tool that is more accessible, affordable, and realistic to investigators, especially to those in the wound repair community, who may be limited by the availability of transgenic animals for lineage tracing or by the small sample sizes of tissue when studying SCs in wounded skin.

Acknowledgments

Many thanks to Dr. Eric C.B. Milner for excellent editing.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work is supported by NIH grant 5R01DK087800 (S.H.).

References

  1. Albiero M, Menegazzo L, Boscaro E, Agostini C, Avogaro A, Fadini GP. (2011). Defective recruitment, survival and proliferation of bone marrow-derived progenitor cells at sites of delayed diabetic wound healing in mice. Diabetologia 54:945-953 [DOI] [PubMed] [Google Scholar]
  2. Ashcroft GS, Horan MA, Ferguson MW. (1997). Aging is associated with reduced deposition of specific extracellular matrix components, an upregulation of angiogenesis, and an altered inflammatory response in a murine incisional wound healing model. J Invest Dermatol 108:430-437 [DOI] [PubMed] [Google Scholar]
  3. Badiavas EV, Abedi M, Butmarc J, Falanga V, Quesenberry P. (2003). Participation of bone marrow derived cells in cutaneous wound healing. J Cell Physiol 196:245-250 [DOI] [PubMed] [Google Scholar]
  4. Baer PC, Kuci S, Krause M, Kuci Z, Zielen S, Geiger H, Bader P, Schubert R. (2013). Comprehensive phenotypic characterization of human adipose-derived stromal/stem cells and their subsets by a high throughput technology. Stem Cells Dev 22:330-339 [DOI] [PubMed] [Google Scholar]
  5. Barrilleaux B, Phinney DG, Prockop DJ, O’Connor KC. (2006). Review: ex vivo engineering of living tissues with adult stem cells. Tissue Eng 12:3007-3019 [DOI] [PubMed] [Google Scholar]
  6. Borue X, Lee S, Grove J, Herzog EL, Harris R, Diflo T, Glusac E, Hyman K, Theise ND, Krause DS. (2004). Bone marrow-derived cells contribute to epithelial engraftment during wound healing. Am J Pathol 165:1767-1772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, Redl H, Rubin JP, Yoshimura K, Gimble JM. (2013). Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy 15:641-648 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Brittan M, Braun KM, Reynolds LE, Conti FJ, Reynolds AR, Poulsom R, Alison MR, Wright NA, Hodivala-Dilke KM. (2005). Bone marrow cells engraft within the epidermis and proliferate in vivo with no evidence of cell fusion. J Pathol 205:1-13 [DOI] [PubMed] [Google Scholar]
  9. Chen C, Takahashi K, Yoshida A, Takizawa Y, Lee Y, Nakui M, Doi H, Takebayashi Y, Fukumoto M, Yamada T, Katagiri H, Oka Y, Satoh J. (2010). Characterization of a novel murine preadipocyte line, AP-18, isolated from subcutaneous tissue: analysis of adipocyte-related gene expressions. Cell Biol Int 34:293-299 [DOI] [PubMed] [Google Scholar]
  10. Chen FG, Zhang WJ, Bi D, Liu W, Wei X, Chen FF, Zhu L, Cui L, Cao Y. (2007). Clonal analysis of nestin(-) vimentin(+) multipotent fibroblasts isolated from human dermis. J Cell Sci 120:2875-2883 [DOI] [PubMed] [Google Scholar]
  11. Chen L, Tredget EE, Wu PY, Wu Y. (2008). Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 3:e1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Chen X, Chen Q, Wang L, Li G. (2013). Ghrelin induces cell migration through GHSR1a-mediated PI3K/Akt/eNOS/NO signaling pathway in endothelial progenitor cells. Metabolism 62:743-752 [DOI] [PubMed] [Google Scholar]
  13. Daley GQ. (2012). The promise and perils of stem cell therapeutics. Cell Stem Cell 10:740-749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. De Francesco F, Tirino V, Desiderio V, Ferraro G, D’Andrea F, Giuliano M, Libondi G, Pirozzi G, De Rosa A, Papaccio G. (2009). Human CD34/CD90 ASCs are capable of growing as sphere clusters, producing high levels of VEGF and forming capillaries. PLoS One 4:e6537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Etich J, Bergmeier V, Frie C, Kreft S, Bengestrate L, Eming S, Mauch C, Eckes B, Ulus H, Lund FE, Rappl G, Abken H, Paulsson M, Brachvogel B. (2013). PECAM1(+)/Sca1(+)/CD38(+) vascular cells transform into myofibroblast-like cells in skin wound repair. PLoS One 8:e53262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Fathke C, Wilson L, Hutter J, Kapoor V, Smith A, Hocking A, Isik F. (2004). Contribution of bone marrow-derived cells to skin: collagen deposition and wound repair. Stem Cells 22:812-822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Felcht M, Luck R, Schering A, Seidel P, Srivastava K, Hu J, Bartol A, Kienast Y, Vettel C, Loos EK, Kutschera S, Bartels S, Appak S, Besemfelder E, Terhardt D, Chavakis E, Wieland T, Klein C, Thomas M, Uemura A, Goerdt S, Augustin HG. (2012). Angiopoietin-2 differentially regulates angiogenesis through TIE2 and integrin signaling. J Clin Invest 122:1991-2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Festa E, Fretz J, Berry R, Schmidt B, Rodeheffer M, Horowitz M, Horsley V. (2011). Adipocyte lineage cells contribute to the skin stem cell niche to drive hair cycling. Cell 146:761-771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Folgiero V, Migliano E, Tedesco M, Iacovelli S, Bon G, Torre ML, Sacchi A, Marazzi M, Bucher S, Falcioni R. (2010). Purification and characterization of adipose-derived stem cells from patients with lipoaspirate transplant. Cell Transplant 19:1225-1235 [DOI] [PubMed] [Google Scholar]
  20. Granick JL, Falahee PC, Dahmubed D, Borjesson DL, Miller LS, Simon SI. (2013). Staphylococcus aureus recognition by hematopoietic stem and progenitor cells via TLR2/MyD88/PGE2 stimulates granulopoiesis in wounds. Blood 122:1770-1778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Hamou C, Callaghan MJ, Thangarajah H, Chang E, Chang EI, Grogan RH, Paterno J, Vial IN, Jazayeri L, Gurtner GC. (2009). Mesenchymal stem cells can participate in ischemic neovascularization. Plast Reconstr Surg 123:45S-55S [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Harris RG, Herzog EL, Bruscia EM, Grove JE, Van Arnam JS, Krause DS. (2004). Lack of a fusion requirement for development of bone marrow-derived epithelia. Science 305:90-93 [DOI] [PubMed] [Google Scholar]
  23. He S, Nakada D, Morrison SJ. (2009). Mechanisms of stem cell self-renewal. Annu Rev Cell Dev Biol 25:377-406 [DOI] [PubMed] [Google Scholar]
  24. Hocking AM, Gibran NS. (2010). Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res 316:2213-2219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Holmes C, Stanford WL. (2007). Concise review: stem cell antigen-1: expression, function, and enigma. Stem Cells 25:1339-1347 [DOI] [PubMed] [Google Scholar]
  26. Hong HS, Lee J, Lee E, Kwon YS, Lee E, Ahn W, Jiang MH, Kim JC, Son Y. (2009). A new role of substance P as an injury-inducible messenger for mobilization of CD29(+) stromal-like cells. Nat Med 15:425-435 [DOI] [PubMed] [Google Scholar]
  27. Hong SJ, Jia SX, Xie P, Xu W, Leung KP, Mustoe TA, Galiano RD. (2013). Topically delivered adipose derived stem cells show an activated-fibroblast phenotype and enhance granulation tissue formation in skin wounds. PLoS One 8:e55640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Hoogduijn MJ, Gorjup E, Genever PG. (2006). Comparative characterization of hair follicle dermal stem cells and bone marrow mesenchymal stem cells. Stem Cells Dev 15:49-60 [DOI] [PubMed] [Google Scholar]
  29. Houlihan DD, Mabuchi Y, Morikawa S, Niibe K, Araki D, Suzuki S, Okano H, Matsuzaki Y. (2012). Isolation of mouse mesenchymal stem cells on the basis of expression of Sca-1 and PDGFR-alpha. Nat Protoc 7:2103-2111 [DOI] [PubMed] [Google Scholar]
  30. Javazon EH, Keswani SG, Badillo AT, Crombleholme TM, Zoltick PW, Radu AP, Kozin ED, Beggs K, Malik AA, Flake AW. (2007). Enhanced epithelial gap closure and increased angiogenesis in wounds of diabetic mice treated with adult murine bone marrow stromal progenitor cells. Wound Repair Regen 15:350-359 [DOI] [PubMed] [Google Scholar]
  31. Jensen KB, Collins CA, Nascimento E, Tan DW, Frye M, Itami S, Watt FM. (2009). Lrig1 expression defines a distinct multipotent stem cell population in mammalian epidermis. Cell Stem Cell 4:427-439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Jeyarajah R, Jayatissa SK, Senivaratne N, Dharmasena BD. (1986). Intermittent peritoneal dialysis. Review of three years’ experience. Trop Doct 16:13-17 [DOI] [PubMed] [Google Scholar]
  33. Jin P, Zhang X, Wu Y, Li L, Yin Q, Zheng L, Zhang H, Sun C. (2010). Streptozotocin-induced diabetic rat-derived bone marrow mesenchymal stem cells have impaired abilities in proliferation, paracrine, antiapoptosis, and myogenic differentiation. Transplant Proc 42:2745-2752 [DOI] [PubMed] [Google Scholar]
  34. Khan M, Akhtar S, Mohsin S, S NK, Riazuddin S. (2011). Growth factor preconditioning increases the function of diabetes-impaired mesenchymal stem cells. Stem Cells Dev 20:67-75 [DOI] [PubMed] [Google Scholar]
  35. Kiel MJ, Morrison SJ. (2008). Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol 8:290-301 [DOI] [PubMed] [Google Scholar]
  36. Kim JC, Whitaker-Menezes D, Deguchi M, Adair BS, Korngold R, Murphy GF. (2002). Novel expression of vascular cell adhesion molecule-1 (CD106) by squamous epithelium in experimental acute graft-versus-host disease. Am J Pathol 161:763-770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Kruse C, Bodo E, Petschnik AE, Danner S, Tiede S, Paus R. (2006). Towards the development of a pragmatic technique for isolating and differentiating nestin-positive cells from human scalp skin into neuronal and glial cell populations: generating neurons from human skin? Exp Dermatol 15:794-800 [DOI] [PubMed] [Google Scholar]
  38. Kwon DS, Gao X, Liu YB, Dulchavsky DS, Danyluk AL, Bansal M, Chopp M, McIntosh K, Arbab AS, Dulchavsky SA, Gautam SC. (2008). Treatment with bone marrow-derived stromal cells accelerates wound healing in diabetic rats. Int Wound J 5:453-463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Lako M, Armstrong L, Cairns PM, Harris S, Hole N, Jahoda CA. (2002). Hair follicle dermal cells repopulate the mouse haematopoietic system. J Cell Sci 115:3967-3974 [DOI] [PubMed] [Google Scholar]
  40. Lorenz K, Sicker M, Schmelzer E, Rupf T, Salvetter J, Schulz-Siegmund M, Bader A. (2008). Multilineage differentiation potential of human dermal skin-derived fibroblasts. Exp Dermatol 17:925-932 [DOI] [PubMed] [Google Scholar]
  41. Lu Y, Tian H, Hong S. (2010). Novel 14,21-dihydroxy-docosahexaenoic acids: structures, formation pathways, and enhancement of wound healing. J Lipid Res 51:923-932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Marsee DK, Pinkus GS, Yu H. (2010). CD71 (transferrin receptor): an effective marker for erythroid precursors in bone marrow biopsy specimens. Am J Clin Pathol 134:429-435 [DOI] [PubMed] [Google Scholar]
  43. Meindl S, Schmidt U, Vaculik C, Elbe-Burger A. (2006). Characterization, isolation, and differentiation of murine skin cells expressing hematopoietic stem cell markers. J Leukoc Biol 80:816-826 [DOI] [PubMed] [Google Scholar]
  44. Michaelson D, Philips M. (2006). The use of GFP to localize Rho GTPases in living cells. Methods Enzymol 406:296-315 [DOI] [PubMed] [Google Scholar]
  45. Neumuller J, Neumuller-Guber SE, Lipovac M, Mosgoeller W, Vetterlein M, Pavelka M, Huber J. (2006). Immunological and ultrastructural characterization of endothelial cell cultures differentiated from human cord blood derived endothelial progenitor cells. Histochem Cell Biol 126:649-664 [DOI] [PubMed] [Google Scholar]
  46. Nie C, Yang D, Xu J, Si Z, Jin X, Zhang J. (2011). Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant 20:205-216 [DOI] [PubMed] [Google Scholar]
  47. Nielsen JS, McNagny KM. (2008). Novel functions of the CD34 family. J Cell Sci 121:3683-3692 [DOI] [PubMed] [Google Scholar]
  48. Okada S, Nakauchi H, Nagayoshi K, Nishikawa S, Miura Y, Suda T. (1992). In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells. Blood 80:3044-3050 [PubMed] [Google Scholar]
  49. Ong WK, Sugii S. (2013). Adipose-derived stem cells: fatty potentials for therapy. Int J Biochem Cell Biol 45:1083-1086 [DOI] [PubMed] [Google Scholar]
  50. Padfield GJ, Tura O, Haeck ML, Short A, Freyer E, Barclay GR, Newby DE, Mills NL. (2010). Circulating endothelial progenitor cells are not affected by acute systemic inflammation. Am J Physiol Heart Circ Physiol 298:H2054-2061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Passier R, Mummery C. (2003). Origin and use of embryonic and adult stem cells in differentiation and tissue repair. Cardiovasc Res 58:324-335 [DOI] [PubMed] [Google Scholar]
  52. Phinney DG, Prockop DJ. (2007). Concise review: mesenchymal stem/multipotent stromal cells: the state of transdifferentiation and modes of tissue repair–current views. Stem Cells 25:2896-2902 [DOI] [PubMed] [Google Scholar]
  53. Plikus MV, Gay DL, Treffeisen E, Wang A, Supapannachart RJ, Cotsarelis G. (2012). Epithelial stem cells and implications for wound repair. Semin Cell Dev Biol 23:946-953 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Prasongchean W, Ferretti P. (2012). Autologous stem cells for personalised medicine. N Biotechnol 29:641-650 [DOI] [PubMed] [Google Scholar]
  55. Prockop DJ. (2009). Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther 17:939-946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Raaijmakers MH, Scadden DT. (2008). Evolving concepts on the microenvironmental niche for hematopoietic stem cells. Curr Opin Hematol 15:301-306 [DOI] [PubMed] [Google Scholar]
  57. Raff M. (2003). Adult stem cell plasticity: fact or artifact? Annu Rev Cell Dev Biol 19:1-22 [DOI] [PubMed] [Google Scholar]
  58. Rebelatto CK, Aguiar AM, Moretao MP, Senegaglia AC, Hansen P, Barchiki F, Oliveira J, Martins J, Kuligovski C, Mansur F, Christofis A, Amaral VF, Brofman PS, Goldenberg S, Nakao LS, Correa A. (2008). Dissimilar differentiation of mesenchymal stem cells from bone marrow, umbilical cord blood, and adipose tissue. Exp Biol Med (Maywood) 233:901-913 [DOI] [PubMed] [Google Scholar]
  59. Richardson GD, Arnott EC, Whitehouse CJ, Lawrence CM, Reynolds AJ, Hole N, Jahoda CA. (2005). Plasticity of rodent and human hair follicle dermal cells: implications for cell therapy and tissue engineering. J Investig Dermatol Symp Proc 10:180-183 [DOI] [PubMed] [Google Scholar]
  60. Rostovskaya M, Anastassiadis K. (2012). Differential expression of surface markers in mouse bone marrow mesenchymal stromal cell subpopulations with distinct lineage commitment. PLoS One 7:e51221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. (2008). Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol 180:2581-2587 [DOI] [PubMed] [Google Scholar]
  62. Seppanen E, Roy E, Ellis R, Bou-Gharios G, Fisk NM, Khosrotehrani K. (2013). Distant mesenchymal progenitors contribute to skin wound healing and produce collagen: evidence from a murine fetal microchimerism model. PLoS One 8:e62662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Sheridan CM, Rice D, Hiscott PS, Wong D, Kent DL. (2006). The presence of AC133-positive cells suggests a possible role of endothelial progenitor cells in the formation of choroidal neovascularization. Invest Ophthalmol Vis Sci 47:1642-1645 [DOI] [PubMed] [Google Scholar]
  64. Skube SB, Chaverri JM, Goodson HV. (2010). Effect of GFP tags on the localization of EB1 and EB1 fragments in vivo. Cytoskeleton (Hoboken) 67:1-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Smith S, Neaves W, Teitelbaum S. (2007). Adult versus embryonic stem cells: treatments. Science 316:1422-1423; author reply 1422-1423. [DOI] [PubMed] [Google Scholar]
  66. Snippert HJ, Clevers H. (2011). Tracking adult stem cells. EMBO Rep 12:113-122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Spangrude GJ, Heimfeld S, Weissman IL. (1988). Purification and characterization of mouse hematopoietic stem cells. Science 241:58-62 [DOI] [PubMed] [Google Scholar]
  68. Stepanovic V, Awad O, Jiao C, Dunnwald M, Schatteman GC. (2003). Leprdb diabetic mouse bone marrow cells inhibit skin wound vascularization but promote wound healing. Circ Res 92:1247-1253 [DOI] [PubMed] [Google Scholar]
  69. Taghizadeh RR, Sherley JL. (2008). CFP and YFP, but not GFP, provide stable fluorescent marking of rat hepatic adult stem cells. J Biomed Biotechnol 2008:453590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Tian H, Lu Y, Shah SP, Hong S. (2011). 14S,21R-Dihydroxydocosahexaenoic Acid Remedies Impaired Healing and Mesenchymal Stem Cell Functions in Diabetic Wounds. J Biol Chem 286:4443-4453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Tian H, Lu Y, Shah SP, Hong S. (2010). Novel 14S,21-dihydroxy-docosahexaenoic acid rescues wound healing and associated angiogenesis impaired by acute ethanol intoxication/exposure. J Cell Biochem 111:266-273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Tobita M, Orbay H, Mizuno H. (2011). Adipose-derived stem cells: current findings and future perspectives. Discov Med 11:160-170 [PubMed] [Google Scholar]
  73. Toma JG, McKenzie IA, Bagli D, Miller FD. (2005). Isolation and characterization of multipotent skin-derived precursors from human skin. Stem Cells 23:727-737 [DOI] [PubMed] [Google Scholar]
  74. Untergasser G, Koeck R, Wolf D, Rumpold H, Ott H, Debbage P, Koppelstaetter C, Gunsilius E. (2006). CD34+/CD133- circulating endothelial precursor cells (CEP): characterization, senescence and in vivo application. Exp Gerontol 41:600-608 [DOI] [PubMed] [Google Scholar]
  75. Vaculik C, Schuster C, Bauer W, Iram N, Pfisterer K, Kramer G, Reinisch A, Strunk D, Elbe-Burger A. (2012). Human dermis harbors distinct mesenchymal stromal cell subsets. J Invest Dermatol 132:563-574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Wu Y, Chen L, Scott PG, Tredget EE. (2007). Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 25:2648-2659 [DOI] [PubMed] [Google Scholar]
  77. Wu Y, Zhao RC, Tredget EE. (2010). Concise review: bone marrow-derived stem/progenitor cells in cutaneous repair and regeneration. Stem Cells 28:905-915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Yang ZX, Han ZB, Ji YR, Wang YW, Liang L, Chi Y, Yang SG, Li LN, Luo WF, Li JP, Chen DD, Du WJ, Cao XC, Zhuo GS, Wang T, Han ZC. (2013). CD106 identifies a subpopulation of mesenchymal stem cells with unique immunomodulatory properties. PLoS One 8:e59354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Zhang X, Semon JA, Zhang S, Strong AL, Scruggs BA, Gimble JM, Bunnell BA. (2013). Characterization of adipose-derived stromal/stem cells from the twitcher mouse model of krabbe disease. BMC Cell Biol 14:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Zimmerlin L, Donnenberg AD, Rubin JP, Basse P, Landreneau RJ, Donnenberg VS. (2011). Regenerative therapy and cancer: in vitro and in vivo studies of the interaction between adipose-derived stem cells and breast cancer cells from clinical isolates. Tissue Eng Part A 17:93-106 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Histochemistry and Cytochemistry are provided here courtesy of The Histochemical Society

RESOURCES