Advertisement
Original Article| Volume 11, ISSUE 1, P118-123, September 2006

Download started.

Ok

Epidermal Stem Cells Have the Potential to Assist in Healing Damaged Tissues

  • Jackie R. Bickenbach
    Correspondence
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, 1-457 BSB, 51 Newton Road, Iowa City, Iowa 52240, USA
    Affiliations
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA

    Department of Dermatology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
    Search for articles by this author
  • Matthew M. Stern
    Affiliations
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA

    Department of Dermatology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
    Search for articles by this author
  • Katie L. Grinnell
    Affiliations
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA

    Department of Dermatology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
    Search for articles by this author
  • Antonio Manuel
    Affiliations
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA

    Department of Dermatology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
    Search for articles by this author
  • Sathivel Chinnathambi
    Affiliations
    Department of Anatomy and Cell Biology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA

    Department of Dermatology, Caver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
    Search for articles by this author
      Homeostasis of continuously renewing tissues, such as the epidermis, is maintained by somatic undifferentiated, self-renewing stem cells, which are thought to persist throughout life. Through a series of labeling experiments, we previously showed that stem cells from mouse skin did not divide often, but they did divide at a steady rate in vivo. Using our recently redefined sorting method, we isolated epidermal stem and transit amplifying (TA) cells from mouse skin. When injected into a developing blastocyst or into damaged tissues, the stem cells, but not the TA cells, could participate in the formation of new tissues. We hypothesize that all tissues contain reserved undifferentiated stem cells that are primed to react if needed. These reserve stem cells could restore the tissue in which they reside or they could be called upon to help restore another tissue that was severely damage.
      LRC
      label-retaining cell
      TA
      transit amplifying

      Somatic Stem Cells of the Epidermis

      Overall, it is generally agreed that homeostasis of the continuously renewing interfollicular epidermis is maintained by a small population of stem cells. Basically, a stem cell is considered to be an undifferentiated cell, which is both self-renewing and the source of all cells in that tissue. This is thought to be accomplished by a mechanism of differential cell division, where the undifferentiated stem cells divide to produce daughter cells that maintain the stem cell phenotype and daughter cells that are somehow changed in that they now can undergo only a finite number of cell divisions before they differentiate and leave the proliferative basal compartment. How this differential cell division is accomplished is not entirely understood, but in the epithelia of the small intestine, cells in the stem cell niche appear to segregate their DNA during cell division such that the stem cells retain the original DNA template while the transit amplifying (TA) daughter cells obtain the newly replicated DNA (
      • Potten C.S.
      • Owen G.
      • Booth D.
      Intestinal stem cells protect their genome by selective segregation of template DNA strands.
      ). Thus, any mutations that occurred during replication would be passed on to the TA daughter cells, leaving the stem cell DNA unperturbed. Whether this is also true for stem cells in skin epidermis has yet to be proven (for a review, see
      • Sherley J.L.
      Asymmetric cell kinetics genes: the key to expansion of adult stem cells in culture.
      ).
      Until recently, it was thought that somatic stem cells contributed cells only to their particular tissue. However, it is clear from several reports that many somatic stem cells, including epidermal stem cells, have the capacity to transdifferentiate into other cell types (e.g., see
      • Krause D.S.
      • Theise N.D.
      • Collector M.I.
      • Henegariu O.
      • Hwang S.
      • Gardner R.
      • et al.
      Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell.
      ;
      • Janes S.M.
      • Lowell S.
      • Hutter C.
      Epidermal stem cells.
      ;
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Verfaillie C.M.
      Adult stem cells: assessing the case for pluripotency.
      ;
      • Rosenthal N.
      Prometheus's vulture and the stem-cell promise.
      ). This bodes well for the use of epidermal stem cells to repair tissues. The skin is the largest organ of the body and the most easily accessible, making epidermal stem cell isolation less problematic for the patient, provided we can identify and isolate the appropriate cells. This latter has been under investigation for several decades.
      Before the early 1980s, information available from radiation and structural studies of the skin (
      • Withers H.R.
      Recovery and repopulation in vivo by mouse skin epithelial cells during fractionated irradiation.
      ;
      • Mackenzie I.C.
      Ordered structure of the stratum corneum of mammalian skin.
      ) suggested that the continuously renewing epidermis should have a self-renewing stem cell population. However, no direct evidence existed. Heterogeneity of the epidermal proliferative compartment was first identified through a series of nuclear label-retention studies (
      • Bickenbach J.R.
      Identification of label-retaining cells in oral mucosa and skin.
      ;
      • Bickenbach J.R.
      • McCutcheon J.
      • Mackenzie I.C.
      Rate of loss of tritiated thymidine label in basal cells in mouse epithelial tissue.
      ). Young growing mice were given several injections of tritiated thymidine over 2 days, then chased for up to 240 days. Samples of various epithelia were fixed, sectioned, and coated with liquid emulsion. The number of silver grains over each labeled cell was counted for each time point and for each tissue (for example of epidermis, see Figure 1). We showed that a few cells in the interfollicular epidermis retained a nuclear label for up to 240 days. We called these label-retaining cells (LRCs). The presence of LRCs with their slow loss of tritiated thymidine over time (a reduction of the number of grains per cell) proved that the epidermal proliferative population contained cells which cycled intermittently. Since that time LRCs have been identified in several tissues, and to date, this is an accepted method to identify epidermal stem cells (
      • Tumbar T.
      • Guasch G.
      • Greco V.
      • Blanpain C.
      • Lowry W.E.
      • Rendl M.
      • et al.
      Defining the epithelial stem cell niche in skin.
      ).
      Figure thumbnail gr1
      Figure 1Percentage of LRCs at 15, 30, 60, 90, and 240 days after labeling. Cells were graphed and clustered according to the number of grains counted per nucleus. Note, the gradual decrease in the number of grains per nuclei with increasing time (purple to dark blue bar in each cluster). Also note that at each time point after labeling, a few cells were heavily labeled (>30 grains/nucleus).

      Isolation of Stem Cells from the Interfollicular Epidermis

      To isolate LRCs, we used a modification and combination of two previously published techniques (
      • Barrandon Y.
      • Green H.
      Cell size as a determinant of the clone-forming ability of human keratinocytes.
      ;
      • Goodell M.A.
      • Brose K.
      • Paradis G.
      • Conner A.S.
      • Mulligan R.C.
      Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
      ,
      • Goodell M.A.
      • Rosenzweig M.
      • Kim H.
      • Marks D.F.
      • DeMaria M.
      • Paradis G.
      • et al.
      Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species.
      ). The first was a method for isolating hematopoietic stem cells, which worked on the principle that stem cells had more ABCG2 cell surface pumps. This allowed them to easily exclude the vital Hoechst 33342 red/blue dye. Hematopoietic cells sorted by this method proved to be highly enriched for the long-term in vivo repopulative stem cells (
      • Goodell M.A.
      • Brose K.
      • Paradis G.
      • Conner A.S.
      • Mulligan R.C.
      Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
      ,
      • Goodell M.A.
      • Rosenzweig M.
      • Kim H.
      • Marks D.F.
      • DeMaria M.
      • Paradis G.
      • et al.
      Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species.
      ). We modified this method and combined it with a method for isolating very small but highly proliferative keratinocytes (
      • Barrandon Y.
      • Green H.
      Cell size as a determinant of the clone-forming ability of human keratinocytes.
      ). The combination of both methods allowed us to use a flow cytometer to sort basal keratinocytes into several populations with different recapitulation and proliferative characteristics (
      • Dunnwald M.
      • Tomanek-Chalkley A.
      • Alexandrunas D.
      • Fishbaugh J.
      • Bickenbach J.R.
      Isolating a pure population of epidermal stem cells for gene use in tissue engineering.
      ;
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Liang L.
      • Chinnathambi S.
      • Stern M.
      • Tomanek-Chalkley A.
      • Manuel T.D.
      • Bickenbach J.R.
      As epidermal stem cells age they do not substantially change their characteristics.
      ;
      • Bickenbach J.R.
      • Stern M.
      Plasticity of epidermal stem cells (EpiSC): survival in various environments.
      ). One population that we called TA cells showed rapid in vitro proliferation, but inability to maintain an epithelium, and no long-term expression of a recombinant gene. Another population that we call stem cells was ∼0.1% of the total basal cell population, expressed keratin 14 (K14), formed large clones in culture, exhibited much slower proliferation but much greater growth potential in vitro than TA cells, reformed and maintained an epidermis in organotypic culture, and expressed a recombinant gene for a long period of time (
      • Dunnwald M.
      • Tomanek-Chalkley A.
      • Alexandrunas D.
      • Fishbaugh J.
      • Bickenbach J.R.
      Isolating a pure population of epidermal stem cells for gene use in tissue engineering.
      ;
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Liang L.
      • Chinnathambi S.
      • Stern M.
      • Tomanek-Chalkley A.
      • Manuel T.D.
      • Bickenbach J.R.
      As epidermal stem cells age they do not substantially change their characteristics.
      ;
      • Bickenbach J.R.
      • Stern M.
      Plasticity of epidermal stem cells (EpiSC): survival in various environments.
      ). Additionally, we showed that both neonatal and older adult epidermal stem cells, but not the TA cells, could produce cell lineages from all three germ layers in the developing mouse (
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Liang L.
      • Chinnathambi S.
      • Stern M.
      • Tomanek-Chalkley A.
      • Manuel T.D.
      • Bickenbach J.R.
      As epidermal stem cells age they do not substantially change their characteristics.
      ). These cells could also assist in tissue recovery after injury (
      • Bickenbach J.R.
      • Stern M.
      Plasticity of epidermal stem cells (EpiSC): survival in various environments.
      ), suggesting that the stem cells retain a high level of tissue plasticity, whereas the TA cells do not.

      Plasticity of Interfollicular Epidermal Stem Cells

      The first evidence that somatic adult stem cells might be pluripotent was reported in 1961 for the hematopoietic system (
      • Till J.E.
      • McCulloch E.A.
      A direct measurement of the radiation sensitivity of normal mouse bone marrow cells.
      ). Hematopoietic progenitor cells are identified by expression of the cell surface antigen CD34 for human or Sca-1 and c-kit for mouse (
      • Orlic D.
      • Anderson D.
      • Bodine D.M.
      Biological properties of subpopulations of pluripotent hematopoietic stem cells enriched by elutriation and flow cytometry.
      ;
      • Veer Reddy G.P.
      • Tiarks C.Y.
      • Wuu J.
      • Hsieh C-C.
      • Quesenberry P.J.
      Cell cycle analysis and synchronization of pluripotent hematopoietic progenitor stem cells.
      ). Hematopoietic early progenitor cells have also been identified as side population cells by their ability to exclude Hoechst 33342 dye, and thus located on the low side of the fluorescence in the scatter plots from the flow cytometer (
      • Goodell M.A.
      • Brose K.
      • Paradis G.
      • Conner A.S.
      • Mulligan R.C.
      Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
      ,
      • Goodell M.A.
      • Rosenzweig M.
      • Kim H.
      • Marks D.F.
      • DeMaria M.
      • Paradis G.
      • et al.
      Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species.
      ;
      • Jackson K.A.
      • Mi T.
      • Goodell M.A.
      Hematopoietic potential of stem cells isolated from murine skeletal muscle.
      ). These hematopoietic cells express the three basic stem cell characteristics of self-renewal, unlimited proliferative capacity, and the ability to differentiate into various mature circulating blood cells (lymphocytes, macrophages, myelomonocytes, granulocytes, erythroid, etc) (
      • Lemischka I.R.
      • Raulet D.H.
      • Mulligan R.C.
      Development potential and dynamic behavior of hematopoietic stem cells.
      ;
      • Keller G.
      • Snodgrass R.
      Life span of multipotential hematopoietic stem cells in vivo.
      ;
      • Uchida N.
      • Fleming W.H.
      • Alpern E.J.
      • Weissman I.L.
      Heterogeneity of hematopoietic stem cells.
      ;
      • Goodell M.A.
      • Brose K.
      • Paradis G.
      • Conner A.S.
      • Mulligan R.C.
      Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
      ;
      • Iscove N.N.
      • Nawa K.
      Hematopoietic stem cells expand during serial transplantation in vivo without apparent exhaustion.
      ). In the past decade, it has been shown that pluripotentiality is not limited to hematopoietic stem cells. For example, stem cells located in intestinal crypts provide all four cell types of the small intestine (paneth, enteroendocrine, goblet, and intestinal epithelial cells) (
      • Potten C.S.
      • Loeffler M.
      Stem cells: attributes, cycles, spirals, pitfalls, and uncertainties. Lessons for and from the crypt.
      ). In the eye, both epithelial and goblet cells are derived from conjunctival stem cells, and corneal cells are derived from limbal stem cells (
      • Wei Z.-G.
      • Cotsarelis G.
      • Sun T.-T.
      • Lavker R.M.
      Label-retaining cells are preferentially located in fornical epithelium: implications on conjunctival epithelial homeostasis.
      ,
      • Wei Z.-G.
      • Sun T.-T.
      • Lavker R.M.
      Rabbit conjunctival and corneal epithelial cells belong to two separate lineages.
      ;
      • Miller S.J.
      • Lavker R.M.
      • Sun T.-T.
      Keratinocyte stem cells of cornea, skin, and hair follicles.
      ;
      • Pellegrini G.
      • Golisano O.
      • Paterna P.
      • Lambiase A.
      • Bonini S.
      • Rama P.
      • et al.
      Location and clonal analysis of stem cells and their differentiated progeny in the human ocular surface.
      ). In addition, stem cells can transdifferentiate into multiple tissues. For example, skeletal muscle cells have the capacity to differentiate into hematopoietic cells (
      • Jackson K.A.
      • Mi T.
      • Goodell M.A.
      Hematopoietic potential of stem cells isolated from murine skeletal muscle.
      ), and adipose cells differentiated into chondrogenic, myogenic, and osteogenic cells in the presence of lineage-specific induction factors (
      • Zuk P.A.
      • Zhu M.
      • Mizuno H.
      • Huang J.
      • Futrell W.
      • Katz A.J.
      • et al.
      Multilineage cells from human adipose tissue: implications for cell-based therapies.
      ). Multipotent mesenchymal cells were also reported (
      • Pittenger M.F.
      • Mackay A.M.
      • Beck S.C.
      • Jaiswal R.K.
      • Douglas R.
      • Mosca J.D.
      • et al.
      Multilineage potential of adult human mesenchymal stem cells.
      ). Adult neural stem cells, injected into an irradiated host, produced blood cells and early hematopoietic cells (
      • Bjornson C.R.R.
      • Rietze R.L.
      • Reynolds B.A.
      • Magli M.C.
      • Vescovi A.L.
      Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo.
      ), and hematopoietic stem cells injected into the tail vein expressed neuronal phenotypes in the brain (
      • Brazelton T.R.
      • Rossi F.M.V.
      • Keshet G.I.
      • Blau H.M.
      From marrow to brain: expression of neuronal phenotypes in adult mice.
      ;
      • Mezey E.
      • Chandras K.J.
      • Harta G.
      • Maki R.A.
      • McKercher S.R.
      Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow.
      ). A single bone marrow stem cells produce cells that incorporate into all three germ layers (
      • Krause D.S.
      • Theise N.D.
      • Collector M.I.
      • Henegariu O.
      • Hwang S.
      • Gardner R.
      • et al.
      Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell.
      ), and neonatal and old adult epidermal stem cells can do the same (
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Liang L.
      • Chinnathambi S.
      • Stern M.
      • Tomanek-Chalkley A.
      • Manuel T.D.
      • Bickenbach J.R.
      As epidermal stem cells age they do not substantially change their characteristics.
      ;
      • Bickenbach J.R.
      • Stern M.
      Plasticity of epidermal stem cells (EpiSC): survival in various environments.
      ). There is some evidence from the Drosophila field that transdifferentiation may be a general phenomenon that is evolutionarily inherited (
      • Wei G.
      • Schubiger G.
      • Harder F.
      • Muller A.M.
      Stem cell plasticity in mammals and transdetermination in Drosophila: common themes?.
      ). Thus, given the appropriate environmental stimulus, somatic stem cells may have the plasticity to form a variety of tissues.
      The plasticity of epithelial cells isolated from whisker and hair follicles have been extensively studied. It has been suggested that cells from the bulge of the hair follicle produce all the epithelial cells in the outer root sheath, inner root sheath, and matrix, as well as cells in the interfollicular epidermis, and cells in the sebaceous glands. Whether this is a typical homeostatic mechanism is not clear. We do know that when the skin is damaged, cells from the hair follicle migrate into the interfollicular epidermis and into the sebaceous gland (
      • Taylor G.
      • Lehrer M.S.
      • Jensen P.J.
      • Sun T.-T.
      • Lavker R.M.
      Involvement of follicular stem cells in forming not only the follicle but also the epidermis.
      ;
      • Ghazizadeh S.
      • Taichman L.B.
      Multiple classes of stem cells in cutaneous epithelium: a lineage analysis of adult mouse skin.
      ). Recently, it was shown that a single-cell cultured from either the bulge or the matrix of the rat whisker follicle could contribute all the cell lineages of the hair follicle and the sebaceous gland if grafted to the basement membrane zone in neonatal mouse skin (
      • Claudinot S.
      • Nicolas M.
      • Oshima H.
      • Rochat A.
      • Barrandon Y.
      Long-term renewal of hair follicles from clonogenic multipotent stem cells.
      ). However, the grafting technique in itself creates a wound, and thus may not truly represent homeostasis. Furthermore, with an ever-increasing array of molecular markers staining different epithelial cell population, it is becoming apparent that follicular and interfollicular stem cells may be distinct (
      • Blanpain C.
      • Lowry W.E.
      • Geoghegan A.
      • Polak L.
      • Fuchs E.
      Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche.
      ;
      • Morris R.J.
      • Liu Y.
      • Marles L.
      • Yang Z.
      • Trempus C.
      • Li S.
      • et al.
      Capturing and profiling adult hair follicle stem cells.
      ;
      • Tumbar T.
      • Guasch G.
      • Greco V.
      • Blanpain C.
      • Lowry W.E.
      • Rendl M.
      • et al.
      Defining the epithelial stem cell niche in skin.
      ;
      • Levy V.
      • Lindon C.
      • Harfe B.D.
      • Morgan B.A.
      Distinct stem cell populations regenerate the follicle and interfollicular epidermis.
      )
      Both embryonic and somatic stem cells have been classified by their potential to contribute to other cell types or tissues as totipotent (able to give rise to all embryonic and extraembryonic cell types), pluripotent (able to give rise to all cell types of the embryo proper), multipotent (able to give rise to a subset cell lineages, usually in a tissue) (for a review, see
      • Wagers A.J.
      • Weissman I.L.
      Plasticity of adult stem cells.
      ). Somatic stem cells have also been classified as plastic (able to change their phenotype or to transdifferentiate into another cell type). However, the mechanism of plasticity is poorly understood. It may be that transdifferentiation is a phenomenon of one type of cell fusing with another type of cell as suggested for the hematopoietic and neural stem cells (
      • Spees J.L.
      • Olson S.D.
      • Ylostalo J.
      • Lynch P.J.
      • Smith J.
      • Perry A.
      • et al.
      Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone stroma.
      ;
      • Terada N.
      • Hamazaki T.
      • Oka M.
      • Hoki M.
      • Mastalerz D.
      • Nakano Y.
      • et al.
      Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion.
      ;
      • Ying Q.
      • Nichols J.
      • Evans E.
      • Smith A.
      Changing potency by spontaneous fusion.
      ;
      • Vassilopoulos G.
      • Wang P.
      • Russell D.
      Transplanted bone marrow regenerates liver by cell fusion.
      ;
      • Wang X.J.
      • Willenbring H.
      • Akkari Y.
      • Torimaru Y.
      • Fostyer M.
      • Al-Dhallmy M.
      • et al.
      Cell fusion is the principal source of bone-marrow-derived hepatocytes.
      ). In our hands, neonatal epidermal stem cells did not appear to fuse with embryonic stem cells when grown together in culture. If fusion is not what is causing plasticity of somatic stem cells, then three other possibilities exist: (1) somatic stem cells might be disbursed throughout other tissues; (2) genetic reprogramming might take place when a somatic stem cell is placed in a new environment; or (3) pluripotent cells left undifferentiated from initial development might persist and be distributed throughout all tissues. The latter possibility would require a shift in the paradigm of stem cells. Furthermore, it appears clear that an inductive influence may be required, such as that found in a developmental environment or a damaged tissue (
      • Levy V.
      • Lindon C.
      • Harfe B.D.
      • Morgan B.A.
      Distinct stem cell populations regenerate the follicle and interfollicular epidermis.
      ).

      Potential of Epidermal Stem Cells for Tissue Replacement

      Follicular cells that were positive for CD34, but negative for keratin 15 were shown to form several types of cells in vitro, including neurons, glia, keratinocytes, smooth muscle cells, and melanocytes (
      • Toma J.G.
      • Akhavan M.
      • Fernandes J.L.
      • Barnabe-Heider F.
      • Sadikot A.
      • Kaplan D.R.
      • et al.
      Isolation of multipotent adult stem cells from the dermis of mammalian skin.
      ). These data suggested that such cells could be used to regenerate tissues after wounding. When CD34-positive, keratin 15-negative, nestin-positive cells were grafted into a gap in a severed sciatic nerve, the cells transdifferentiated primarily into Schwann cells. Importantly, they appeared to completely restore the function of the severed nerve, for example, the rats could walk again (
      • Amoh Y.
      • Lingna Li L.
      • Campillo R.
      • Kawahara K.
      • Katsuoka K.
      • Penman S.
      • et al.
      Implanted hair follicle stem cells form Schwann cells that support repair of severed peripheral nerves.
      ). Thus, even though transdifferentiation may not be a typical homeostatic function of epithelial stem cells, these findings suggest that using them for tissue regeneration is viable.
      To examine the transdifferentiation potential of epidermal stem cells under stress, we created full-thickness skin wounds (6 mm punch biopsies) in the backs of young adult C57BL/6 mice. After 2 days, we injected green fluorescence protein (GFP)+ epidermal stem cells just beneath the wound bed. The GFP+ epidermal stem cells came from the back skin of GFP transgenic neonatal mice. Controls received TA cells or buffer only. The wounds that received the neonatal epidermal stem cells appeared to heal faster in that by 4 days after injection, the gross appearance of the wounds appeared healed, whereas both sets of control wounds did not appear healed until 7 days after injection. At 21 days, histological analysis indicated that all the wounds were fully healed. In wounds that received epidermal stem cells, we found GFP+ cells in the underlying dermal tissues, including cells with an adipocyte morphology within the fat layer (Figure 2). These findings suggest that under the stress of wounding, mouse epidermal stem cells have the ability to incorporate into the healing dermal tissues.
      Figure thumbnail gr2
      Figure 2Wounds receiving neonatal epidermal stem cells retain GFP+ cells. Full-thickness skin wounds were created in the back skin of C57BL/6 non-transgenic mice. After 2 days, epidermal stem cells were isolated from the back skin of neonatal GFP transgenic mice, and injected beneath the wound beds. Shown here are adjacent sections through the middle of the healed wound bed 21 days after GFP+ epidermal stem cells were injected. (a) Hematoxylin- and eosin-stained section. (b) Unstained adjacent section showing GFP fluorescence. Circle surrounds a cluster of GFP+ cells of varying types. Arrows point to single GFP+ cell.
      Epidermal stem cells also show potential for non-skin tissue replacement therapy. When GFP+ epidermal stem cells were injected into the blood stream of lethally irradiated mice, they helped the mice survive and appeared to help repair their damaged bone marrow (Figures 3 and 4). Recipient mice were irradiated with 1,100 cGy whole-body γ-radiation from a Cesium-137 source (broken into two doses of 600 and 500 cGy, 3 h apart). After 24 h, 1 × 105 GFP+ epidermal stem cells were injected. For radioprotection, each mouse received 1 × 105 total bone marrow cells from non-GFP-expressing mice. Control mice received TA epidermal cells, bone marrow cells only, or buffer. We monitored the mice for 8 months. All control mice receiving 2 × 105 bone marrow cells lived for the entire 8 months; those who received 1 × 105 bone marrow cells died at 2 months. All mice receiving epidermal stem cells lived the entire 8 months (Figure 3). Mice receiving 1 × 105 TA cells did not fare as well. By 4 months after irradiation, 60% (12 of 20 mice) were dead, and by 8 months after injection, 80% were dead (Figure 3). At 8 months, surviving mice that received TA cells had no GFP+ cells in cell smears made from total bone marrow isolates. In the bone marrow cell smears from mice that received epidermal stem cells, only ∼1 in 1,000,000 were GFP+. Although this is a very low percentage of engraftment, some GFP+ cells in the circulating blood were also positive for a cocktail of lineage markers (Figure 4), indicating that at least a few epidermal stem cells contributed to repopulating the hematopoietic tissue. These data suggest that epidermal stem cells aid in the overall recovery of the irradiated mice.
      Figure thumbnail gr3
      Figure 3Survival graph of irradiated mice receiving epidermal stem (EpiSC) or epidermal transit amplifying cells (EpiTA). Note that all the mice receiving epidermal stem cells lived, whereas most of the mice receiving epidermal TA cells died, suggesting that epidermal stem cells participated in hematopoietic repopulation.
      Figure thumbnail gr4
      Figure 4GFP+ Lin+ cells present in peripheral blood of irradiated mice 8 months after receiving GFP+ epidermal stem cells. C57BL/6 mice were lethally irradiated, then injected with GFP-expressing epidermal stem cells, and the bone marrow examined. Panels show two pictures of the same area. (a) Smear stained with an antibody cocktail of hematopoietic lineage markers for macrophages/monocytes (CD11b, Mac-1), T-lymphocytes (CD5, CD3, CD4, CD8), B-lymphocytes (CD45R,B220), granulocytes (Gr-1), and erythrocytes (TER119), and then alkaline phosphatase-labeled secondary antibody. (b) Smear stained with an antibody to GFP, and then horseradish peroxidase-labeled secondary antibody. Arrows point to GFP+ cells that are also hematopoietic lineage marker positive.

      Potential to Improve Targeting of Epidermal Stem Cells to Non-Skin Tissues

      To improve engraftment of epidermal stem cells to non-skin tissue, it appears that we will have to alter the cells before injection. One way to approach this is to change the cell's surface receptors. This could be carried out by transfecting or transducing the cells with the appropriate gene. However, the efficiency of most transfection methods is only 10–20% in epidermal keratinocytes, thus requiring either injection of many more cells or selection of only the cells that have been transfected. Furthermore, if a viral vector is used to transduce the gene, then the added risk of transferring viral proteins might preclude these cells for future use in humans. We have taken a different approach, and allowed the epidermal stem cells to alter their own cell surface receptors in response to a change in their environment. We isolated and cultured mouse epidermal stem cells and epidermal TA cells. Both cell types were grown in low calcium medium with 10% chelexed serum, or in keratinocyte serum-free medium with bovine pituitary extract in place of the serum, or in defined keratinocyte serum-free medium, which is a defined medium. To each cell type grown in each medium, we added 100 ng/ml of IL-3, IL-6, stem cell factor, or stromal-derived factor-1α, or a combination of 100 ng/ml IL-6 plus 100 ng/ml stromal-derived factor-1α. These factors have been shown to influence hematopoietic progenitor cells to enter specific lineage pathways. After 3 days in culture, cells were double-stained with antibodies to Sca-1 and K14. Sca-1 is found on hematopoietic progenitors cells that have been in contact with bone marrow stromal cells, and K14 is an intermediate filament, which is expressed by basal keratinocytes. Cells were scored as expressing Sca-1, K14, or both markers (Figure 5). Five separate experiments were performed, and 200 cells counted in each experiment. TA cells did not express Sca-1. Epidermal stem cells responded by expressed Sca-1 in all treatment groups, with cells exposed to stromal-derived factor-1α showing the most complete change. Approximately, 97% of cells expressed Sca-1+ and no longer expressing K14 (Figure 5). These data indicate that under certain in vitro growth conditions, epidermal stem cells can specifically respond to cytokines and alter their cell surface expression to that of a hematopoietic cell surface marker. It is interesting that the expression of Sca-1 on the cell's surface appears to be incompatible with the intermediate filament K14, which was always seen in partial collapse in co-expressing cells. Surprisingly, the type of medium in which the cells were grown appeared to make no difference in the overall results. We had expected that the presence of serum in the medium might drastically upregulate the Sca-1 expression. However, the control cultures grown in serum, but without the addition of the cytokines, showed continual expression of K14 and no upregulation of Sca-1. Whether changing the epidermal expression of Sca-1 in vitro before injecting the cells results in an increased bone marrow engraftment of these cells remains to be determined.
      Figure thumbnail gr5
      Figure 5Percentage of cells expressing Sca-1, K14, or both after exposure to various cytokines in culture. Cells were cultured for 3 days in defined keratinocyte serum-free medium plus 100 ng/ml of stem cell factor, IL-3, IL-6, stromal-derived factor-α, or 100 ng/ml each of IL6+stromal-derived factor-1α, and then fixed and double-labeled with antibodies to K14 and Sca-1. The percentage of cells stained with only K14 (green bars), only Sca-1 (red bars), or both (yellow bars) antibodies were calculated. Experiments were performed five times and 200 cells were counted in each experiment. Means and standard deviations were determined for each group.

      Paradigm Shift

      In conclusion, we have shown that stem cells can be highly enriched from the epidermis of the skin, and that given the appropriate stimulus, these cells appear to be induced to transdifferentiate into cell types in tissues other than the epidermis of the skin. How this takes place is not clear. It may be that all tissues contain a small population of uncommitted stem cells. Thus, we are not seeing true transdifferentiation. Instead, we are inducing the uncommitted stem cells population. We speculate that these cells remain sequestered as reserved stem cells that sit in each tissue primed to react if needed. These reserved stem cells could restore the tissue in which they reside or they could be called upon to migrate and help restore another tissue under severe damage. It has been shown that in amphibians pluripotent epi-like stem cells in the adult can form all types of cells and can express typical embryonic markers (
      • Young H.E.
      Existence of reserve quiescent stem cells in adults, from amphibians to humans.
      ). Such cells were not lineage committed in that 1% of these cells could form any somatic tissue at any given time. They did not follow the typical lineage pathway established in the embryo during organogenesis. Tissue regeneration in the amphibian is owing to the activation of the reserved population of quiescent uncommitted stem cells to proliferate and commit to a specific tissue lineage (
      • Young H.E.
      Existence of reserve quiescent stem cells in adults, from amphibians to humans.
      ). If this is true, then the mechanism underlying the plasticity (fusion, or transdifferentiation) of the stem cells is immaterial. For example, somatic neural stem cells have a very broad developmental capacity and the ability to contribute to the formation of a variety of organs in both mouse and chick embryos (
      • Clarke D.L.
      • Johansson C.B.
      • Wilbertz J.
      • Veress B.
      • Nilsson E.
      • Karlstrom H.
      • et al.
      Generalized potential of adult neural stem cells.
      ). These mouse cells were cultured as neurospheres, then injected as adherent neurospheres into chick amniotic cavities or dissociated and injected directly into mouse blastocysts. Both chick and mouse embryos showed a mosaic pattern of cells derived from the neural stem cells, indicating that the cells could commit to an altered lineage pathway in development. More interestingly, these cells appear to be able to form a committed lineage in a different organism, suggesting that they may be more uncommitted than previously thought. Our data suggest that epidermal stem cells may also have this ability. We found that somatic epidermal stem cells could contribute to a variety of tissues during the development of a mouse and that they persisted into adulthood (
      • Liang L.
      • Bickenbach J.R.
      Somatic epidermal stem cells can produce multiple cell lineages during development.
      ;
      • Liang L.
      • Chinnathambi S.
      • Stern M.
      • Tomanek-Chalkley A.
      • Manuel T.D.
      • Bickenbach J.R.
      As epidermal stem cells age they do not substantially change their characteristics.
      ). We have determined that they survive in zebrafish embryos (
      • Bickenbach J.R.
      • Stern M.
      Plasticity of epidermal stem cells (EpiSC): survival in various environments.
      ), but we have not tested whether they are interacting with the zebrafish cells. Hematopoietic stem cells have been shown to incorporate and function in a variety of murine tissues (
      • Krause D.S.
      • Theise N.D.
      • Collector M.I.
      • Henegariu O.
      • Hwang S.
      • Gardner R.
      • et al.
      Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell.
      ). Furthermore, these uncommitted cells appear to exist in all tissues and persist into adult tissues. Long-term persistence of grafted male cells in females have been reported (
      • Flowers M.E.D.
      • Stockschlaeder M.A.R.
      • Schuening F.G.
      • Niederwieser D.
      • Hackman R.
      • Miller A.D.
      • et al.
      Long-term transplantation of canine keratinocytes made resistant to G418 through retrovirus-mediated gene transfer.
      ), as well as 27-year persistence of male cells in the blood of females who had given birth to male offspring (
      • Bianchi D.W.
      • Zickwolf G.K.
      • Weil G.J.
      • Sylvester S.
      • DeMaria M.A.
      Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum.
      ). Although lifelong persistence of single uncommitted stem cell has not been proved, it is an attractive hypothesis and leads us to wonder whether stem cells change with increasing age or whether they remain as uncommitted, undifferentiated cells in all tissues – just waiting to be called to act.

      Conflict of Interest

      The authors state no conflict of interest.

      ACKNOWLEDGMENTS

      We thank the current members of the Bickenbach Laboratory for their helpful discussions, and the members of The University of Iowa Flow Cytometry Core for their excellent technical help. We also thank Dr Luchuan Liang, Jason Marley, and Dr Yubin Kang for their help, and Dr Ian Mackenzie for his support and intelligent arguments. This work was supported by the National Institutes of Health (NIAMS, NIA) and a focused giving award from Johnson & Johnson.

      REFERENCES

        • Amoh Y.
        • Lingna Li L.
        • Campillo R.
        • Kawahara K.
        • Katsuoka K.
        • Penman S.
        • et al.
        Implanted hair follicle stem cells form Schwann cells that support repair of severed peripheral nerves.
        Proc Natl Acad Sci USA. 2005; 102: 17734-17738
        • Barrandon Y.
        • Green H.
        Cell size as a determinant of the clone-forming ability of human keratinocytes.
        Proc Natl Acad Sci USA. 1985; 82: 5390-5394
        • Bianchi D.W.
        • Zickwolf G.K.
        • Weil G.J.
        • Sylvester S.
        • DeMaria M.A.
        Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum.
        Proc Natl Acad Sci USA. 1996; 93: 705-708
        • Bickenbach J.R.
        Identification of label-retaining cells in oral mucosa and skin.
        J Dent Res. 1981; 122C: 1611-1620
        • Bickenbach J.R.
        • McCutcheon J.
        • Mackenzie I.C.
        Rate of loss of tritiated thymidine label in basal cells in mouse epithelial tissue.
        Cell Tissue Kinet. 1986; 19: 325-333
        • Bickenbach J.R.
        • Stern M.
        Plasticity of epidermal stem cells (EpiSC): survival in various environments.
        Stem Cell Rev. 2005; 1: 71-77
        • Bjornson C.R.R.
        • Rietze R.L.
        • Reynolds B.A.
        • Magli M.C.
        • Vescovi A.L.
        Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo.
        Science. 1999; 283: 534-537
        • Blanpain C.
        • Lowry W.E.
        • Geoghegan A.
        • Polak L.
        • Fuchs E.
        Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche.
        Cell. 2004; 118: 635-648
        • Brazelton T.R.
        • Rossi F.M.V.
        • Keshet G.I.
        • Blau H.M.
        From marrow to brain: expression of neuronal phenotypes in adult mice.
        Science. 2000; 290: 1775-1779
        • Clarke D.L.
        • Johansson C.B.
        • Wilbertz J.
        • Veress B.
        • Nilsson E.
        • Karlstrom H.
        • et al.
        Generalized potential of adult neural stem cells.
        Science. 2000; 288: 1660-1663
        • Claudinot S.
        • Nicolas M.
        • Oshima H.
        • Rochat A.
        • Barrandon Y.
        Long-term renewal of hair follicles from clonogenic multipotent stem cells.
        Proc Natl Acad Sci USA. 2005; 102: 14677-14682
        • Dunnwald M.
        • Tomanek-Chalkley A.
        • Alexandrunas D.
        • Fishbaugh J.
        • Bickenbach J.R.
        Isolating a pure population of epidermal stem cells for gene use in tissue engineering.
        Exp Dermatol. 2001; 10: 45-54
        • Flowers M.E.D.
        • Stockschlaeder M.A.R.
        • Schuening F.G.
        • Niederwieser D.
        • Hackman R.
        • Miller A.D.
        • et al.
        Long-term transplantation of canine keratinocytes made resistant to G418 through retrovirus-mediated gene transfer.
        Proc Natl Acad Sci USA. 1990; 87: 2349-2353
        • Ghazizadeh S.
        • Taichman L.B.
        Multiple classes of stem cells in cutaneous epithelium: a lineage analysis of adult mouse skin.
        EMBO J. 2001; 20: 1215-1222
        • Goodell M.A.
        • Brose K.
        • Paradis G.
        • Conner A.S.
        • Mulligan R.C.
        Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
        J Exp Med. 1996; 183: 1797-1806
        • Goodell M.A.
        • Rosenzweig M.
        • Kim H.
        • Marks D.F.
        • DeMaria M.
        • Paradis G.
        • et al.
        Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species.
        Nat Med. 1997; 3: 1337-1345
        • Iscove N.N.
        • Nawa K.
        Hematopoietic stem cells expand during serial transplantation in vivo without apparent exhaustion.
        Curr Biol. 1997; 7: 805-808
        • Jackson K.A.
        • Mi T.
        • Goodell M.A.
        Hematopoietic potential of stem cells isolated from murine skeletal muscle.
        Proc Natl Acad Sci USA. 1999; 96: 14482-14486
        • Janes S.M.
        • Lowell S.
        • Hutter C.
        Epidermal stem cells.
        J Pathol. 2002; 197: 479-491
        • Keller G.
        • Snodgrass R.
        Life span of multipotential hematopoietic stem cells in vivo.
        J Exp Med. 1990; 171: 1407-1418
        • Krause D.S.
        • Theise N.D.
        • Collector M.I.
        • Henegariu O.
        • Hwang S.
        • Gardner R.
        • et al.
        Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell.
        Cell. 2001; 105: 369-377
        • Lemischka I.R.
        • Raulet D.H.
        • Mulligan R.C.
        Development potential and dynamic behavior of hematopoietic stem cells.
        Cell. 1986; 45: 917-927
        • Levy V.
        • Lindon C.
        • Harfe B.D.
        • Morgan B.A.
        Distinct stem cell populations regenerate the follicle and interfollicular epidermis.
        Dev Cell. 2005; 9: 855-861
        • Liang L.
        • Bickenbach J.R.
        Somatic epidermal stem cells can produce multiple cell lineages during development.
        Stem Cells. 2002; 20: 3-10
        • Liang L.
        • Chinnathambi S.
        • Stern M.
        • Tomanek-Chalkley A.
        • Manuel T.D.
        • Bickenbach J.R.
        As epidermal stem cells age they do not substantially change their characteristics.
        J Investig Dermatol Symp Proc. 2004; 9: 229-237
        • Mackenzie I.C.
        Ordered structure of the stratum corneum of mammalian skin.
        Nature. 1969; 222: 881-882
        • Mezey E.
        • Chandras K.J.
        • Harta G.
        • Maki R.A.
        • McKercher S.R.
        Turning blood into brain: cells bearing neuronal antigens generated in vivo from bone marrow.
        Science. 2000; 290: 1779-1782
        • Miller S.J.
        • Lavker R.M.
        • Sun T.-T.
        Keratinocyte stem cells of cornea, skin, and hair follicles.
        in: Potten C.S. Stem Cells. London, Academic Press1997: 331-362
        • Morris R.J.
        • Liu Y.
        • Marles L.
        • Yang Z.
        • Trempus C.
        • Li S.
        • et al.
        Capturing and profiling adult hair follicle stem cells.
        Nat Biotechnol. 2004; 22: 411-417
        • Orlic D.
        • Anderson D.
        • Bodine D.M.
        Biological properties of subpopulations of pluripotent hematopoietic stem cells enriched by elutriation and flow cytometry.
        Blood Cells. 1994; 20: 107-117
        • Pellegrini G.
        • Golisano O.
        • Paterna P.
        • Lambiase A.
        • Bonini S.
        • Rama P.
        • et al.
        Location and clonal analysis of stem cells and their differentiated progeny in the human ocular surface.
        J Cell Biol. 1999; 145: 769-782
        • Pittenger M.F.
        • Mackay A.M.
        • Beck S.C.
        • Jaiswal R.K.
        • Douglas R.
        • Mosca J.D.
        • et al.
        Multilineage potential of adult human mesenchymal stem cells.
        Science. 1999; 284: 143-147
        • Potten C.S.
        • Loeffler M.
        Stem cells: attributes, cycles, spirals, pitfalls, and uncertainties. Lessons for and from the crypt.
        Development. 1990; 110: 1001-1020
        • Potten C.S.
        • Owen G.
        • Booth D.
        Intestinal stem cells protect their genome by selective segregation of template DNA strands.
        J Cell Sci. 2002; 115: 2381-2388
        • Rosenthal N.
        Prometheus's vulture and the stem-cell promise.
        N Engl J Med. 2003; 349: 267-274
        • Sherley J.L.
        Asymmetric cell kinetics genes: the key to expansion of adult stem cells in culture.
        Stem Cells. 2002; 20: 561-572
        • Spees J.L.
        • Olson S.D.
        • Ylostalo J.
        • Lynch P.J.
        • Smith J.
        • Perry A.
        • et al.
        Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone stroma.
        Proc Natl Acad Sci USA. 2002; 100: 2397-2402
        • Taylor G.
        • Lehrer M.S.
        • Jensen P.J.
        • Sun T.-T.
        • Lavker R.M.
        Involvement of follicular stem cells in forming not only the follicle but also the epidermis.
        Cell. 2000; 102: 451-461
        • Terada N.
        • Hamazaki T.
        • Oka M.
        • Hoki M.
        • Mastalerz D.
        • Nakano Y.
        • et al.
        Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion.
        Nature. 2002; 416: 482-485
        • Till J.E.
        • McCulloch E.A.
        A direct measurement of the radiation sensitivity of normal mouse bone marrow cells.
        Radiat Res. 1961; 14: 213-219
        • Toma J.G.
        • Akhavan M.
        • Fernandes J.L.
        • Barnabe-Heider F.
        • Sadikot A.
        • Kaplan D.R.
        • et al.
        Isolation of multipotent adult stem cells from the dermis of mammalian skin.
        Nat Cell Biol. 2001; 3: 778-784
        • Tumbar T.
        • Guasch G.
        • Greco V.
        • Blanpain C.
        • Lowry W.E.
        • Rendl M.
        • et al.
        Defining the epithelial stem cell niche in skin.
        Science. 2004; 303: 359-363
        • Uchida N.
        • Fleming W.H.
        • Alpern E.J.
        • Weissman I.L.
        Heterogeneity of hematopoietic stem cells.
        Curr Opin Immunol. 1993; 5: 177-184
        • Vassilopoulos G.
        • Wang P.
        • Russell D.
        Transplanted bone marrow regenerates liver by cell fusion.
        Nature. 2003; 422: 901-904
        • Veer Reddy G.P.
        • Tiarks C.Y.
        • Wuu J.
        • Hsieh C-C.
        • Quesenberry P.J.
        Cell cycle analysis and synchronization of pluripotent hematopoietic progenitor stem cells.
        Blood. 1997; 90: 2293-2299
        • Verfaillie C.M.
        Adult stem cells: assessing the case for pluripotency.
        Trends Cell Biol. 2002; 12: 502-508
        • Wagers A.J.
        • Weissman I.L.
        Plasticity of adult stem cells.
        Cell. 2004; 116: 639-648
        • Wang X.J.
        • Willenbring H.
        • Akkari Y.
        • Torimaru Y.
        • Fostyer M.
        • Al-Dhallmy M.
        • et al.
        Cell fusion is the principal source of bone-marrow-derived hepatocytes.
        Nature. 2003; 422: 897-901
        • Wei G.
        • Schubiger G.
        • Harder F.
        • Muller A.M.
        Stem cell plasticity in mammals and transdetermination in Drosophila: common themes?.
        Stem Cells. 2000; 18: 409-414
        • Wei Z.-G.
        • Cotsarelis G.
        • Sun T.-T.
        • Lavker R.M.
        Label-retaining cells are preferentially located in fornical epithelium: implications on conjunctival epithelial homeostasis.
        Invest Ophthal Vis Sci. 1995; 36: 236-246
        • Wei Z.-G.
        • Sun T.-T.
        • Lavker R.M.
        Rabbit conjunctival and corneal epithelial cells belong to two separate lineages.
        Invest Ophthal Vis Sci. 1996; 37: 523-533
        • Withers H.R.
        Recovery and repopulation in vivo by mouse skin epithelial cells during fractionated irradiation.
        Radiat Res. 1967; 32: 227-239
        • Ying Q.
        • Nichols J.
        • Evans E.
        • Smith A.
        Changing potency by spontaneous fusion.
        Nature. 2002; 416: 485-487
        • Young H.E.
        Existence of reserve quiescent stem cells in adults, from amphibians to humans.
        Curr Top Microbiol Immunol. 2004; 280: 71-109
        • Zuk P.A.
        • Zhu M.
        • Mizuno H.
        • Huang J.
        • Futrell W.
        • Katz A.J.
        • et al.
        Multilineage cells from human adipose tissue: implications for cell-based therapies.
        Tissue Eng. 2001; 7: 211-228