251
|
Hair follicle bulge: a fascinating reservoir of epithelial stem cells. J Dermatol Sci 2007; 46:81-9. [PMID: 17207970 DOI: 10.1016/j.jdermsci.2006.12.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 11/29/2006] [Accepted: 12/06/2006] [Indexed: 12/17/2022]
Abstract
Hair follicles reconstitute themselves though the hair cycle, suggesting the presence of intrinsic stem cells. In contrast to the previous belief that stem cells reside in the bulbar region of hair follicles, stem cells were detected in the bulge area, a contiguous part of outer root sheath, that provides the insertion point for arrector pili muscle and marks the bottom of the permanent portion of hair follicles. The bulge cells are morphologically undifferentiated and slow-cycling under the normal conditions. Later, studies successively demonstrated that bulge cells possess stem cell properties such as high proliferative capacity and multipotency to regenerate not only hair follicles but also sebaceous glands and epidermis. Our knowledge of the bulge cell biology is rapidly increasing because of the identification of novel cell surface markers, the ability to isolate living bulge cells, and microarray analysis of multiple gene expression. Importantly, novel cell surface markers were identified on human bulge cells using precise laser capture microdissection and microarray analyses. Use of these markers enabled the successful enrichment of living human bulge cells, raising the possibility of future treatments of hair disorders using stem cells. Additional clinical relevance of bulge cell biology includes the importance of bulge cells as a gene therapy target and their possible roles in tumorigenesis.
Collapse
|
252
|
Motlík J, Klíma J, Dvoránková B, Smetana K. Porcine epidermal stem cells as a biomedical model for wound healing and normal/malignant epithelial cell propagation. Theriogenology 2007; 67:105-11. [PMID: 17055565 DOI: 10.1016/j.theriogenology.2006.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
This article summarizes research using cells derived from epidermis of the miniature pigs for use as a cell therapy for skin repair and as a model for squamous carcinoma of the head and neck. Stem cells are an important "tool" for biomedical research. Adult stem cells are defined functionally, as cells that have the capacity to self-renew as well as the ability to generate differentiated cells. They are present in defined tissue microenvironments called niches. Asymmetric mitosis allows them to produce one daughter cell with the properties of stem cells (self-renewal) and a second cell with characteristics of progenitor cells, or transit amplifying cells, which proliferate quickly but with a limited number of mitotic divisions. Porcine epidermal stem cells, located in the bulge region of the outer root sheath of hair follicles, migrate in vitro from hair sheaths and because they are resistant to anoikis (detachment induced apoptosis), survive in non-adhesive conditions to form spheroids. These cells express keratins, galectin-1 and their nuclei are rich in DeltaNp63alpha. Interestingly, the multiple phenotype analysis of the human tumor cells in squamous carcinoma of head and neck revealed similarities with epidermal stem cells. These cancer stem cells are usually located on the periphery of the tumor where the invasive front of the tumor responsible for its aggressive behavior is located. In contrast, extensive expression of markers of terminal differentiation such as expression of glycoligands reactive for the endogenous lectin, galectin-3, indicates better tumor prognosis.
Collapse
Affiliation(s)
- J Motlík
- Institute of Animal Physiology and Genetics, Academy of Sciences, Libechov, Czech Republic.
| | | | | | | |
Collapse
|
253
|
Gu LH, Coulombe PA. Keratin expression provides novel insight into the morphogenesis and function of the companion layer in hair follicles. J Invest Dermatol 2006; 127:1061-73. [PMID: 17170733 DOI: 10.1038/sj.jid.5700673] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hair follicles cycle between stages of growth (anagen) and metabolic quiescence (telogen) throughout life. In mature follicles, transition from telogen back into anagen involves the activation, proliferation, and differentiation of epithelial stem cells located in the bulge, a specialization of the outer root sheath. Recent studies identified keratin 6a (K6a) transcripts as enriched in bulge epithelial stem cells in mouse skin. We used messenger RNA probes, antibodies, a LacZ reporter mouse model, and whole-mount staining assays to investigate the regulation of mK6a during mouse postnatal hair cycling, and compare it to mK75, a companion layer (Cl) marker. We find that mK75 regulation parallels that of inner root sheath (IRS) markers, with expression onset at anagen IIIa above the new hair bulb and subsequent spreading towards the bulge. Although also occurring in the Cl, mK6a expression begins at anagen IIIb in differentiating cells located proximal to the bulge, and subsequently spreads towards the hair bulb. mK6a and mK75 thus exhibit temporally distinct, and spatially opposed, expression patterns in the Cl during postnatal anagen. These findings provide novel insight into the morphogenesis and properties of the Cl, and raise the distinct possibility that it is an integral part of the IRS compartment.
Collapse
Affiliation(s)
- Li-Hong Gu
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
254
|
Pascucci L, Mercati F, Gargiulo AM, Pedini V, Sorbolini S, Ceccarelli P. CD34 glycoprotein identifies putative stem cells located in the isthmic region of canine hair follicles. Vet Dermatol 2006; 17:244-51. [PMID: 16827667 DOI: 10.1111/j.1365-3164.2006.00527.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It is widely documented that a pool of multipotent stem cells located in humans and mice hair follicle outer root sheath (bulge region) is involved in the restoration of the whole follicular unit during each anagen phase. To the authors' knowledge, data regarding the location and characterization of hair follicle stem compartment in dogs have not been reported in the recent relevant literature. In this study, we investigated the haematopoietic stem and progenitor cell antigen CD34 as a marker of putative stem cells located in a bulge-like region of canine hair follicles. The presence of CD34 mRNA and glycoprotein was assessed on formalin-fixed, paraffin-embedded canine skin samples by in situ hybridization technique and by standard immunohistochemistry, respectively. A strong expression of CD34 mRNA and glycoprotein was observed in a well-defined area of the hair follicle isthmic region and appeared uniformly concentrated at the level of the basal layer of the outer root sheath. These findings provide compelling support to the hypothesis that in dogs, a subpopulation of basal keratinocytes located in the hair follicle isthmic region and characterized by the selective expression of CD34 is potentially associated with the stem cell compartment of this skin appendage.
Collapse
Affiliation(s)
- Luisa Pascucci
- Dipartimento di Scienze Biopatologiche ed Igiene delle Produzioni Animali ed Alimentari - Facoltà di Medicina Veterinaria, Università di Perugia, Perugia, Italy.
| | | | | | | | | | | |
Collapse
|
255
|
Zhan Q, Signoretti S, Whitaker-Menezes D, Friedman TM, Korngold R, Murphy GF. Cytokeratin15-positive basal epithelial cells targeted in graft-versus-host disease express a constitutive antiapoptotic phenotype. J Invest Dermatol 2006; 127:106-15. [PMID: 17039241 DOI: 10.1038/sj.jid.5700583] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The normal gene expression profile of rete-tip keratinocytes targeted in human graft-versus-host disease (GVHD) remains unexplored. Murine lingual epithelium, unlike murine skin, consists of a basal layer that resembles human cutaneous rete ridges and harbors rete tip-associated cells that express cytokeratin 15 (K15), a marker for epithelial stem cells. Target cell apoptosis in murine GVHD preferentially involves subpopulations of basal cells that (1) reside at tips of lingual rete ridge-like prominences (RLPs), (2) constitutively express K15 protein, (3) express the proapoptotic protein Bax early in disease progression, and (4) coincide spatially with putative epithelial stem cells. Here, we show by real-time reverse transcription-PCR that immunohistochemistry-guided laser-captured K15-positive (K15+) murine basal cells constitutively express quantitatively higher mRNA levels for K15 but lower mRNA levels of Bax than do K15- basal cells, consistent with the presumed stem cell nature of K15+ basal cells. Moreover, apoptosis gene array screening of K15+ microdissected basal cells demonstrated a dominant trend toward the preferential expression of genes associated with protection from apoptosis. Accordingly, genes that regulate apoptotic vulnerability are differentially expressed in basal layer subpopulations distinguishable by K15 expression.
Collapse
Affiliation(s)
- Qian Zhan
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
256
|
Hayashi R, Yamato M, Sugiyama H, Sumide T, Yang J, Okano T, Tano Y, Nishida K. N-Cadherin is expressed by putative stem/progenitor cells and melanocytes in the human limbal epithelial stem cell niche. Stem Cells 2006; 25:289-96. [PMID: 17008425 DOI: 10.1634/stemcells.2006-0167] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Corneal epithelial stem cells are known to be localized to the basal layer of the limbal epithelium, providing a model system for epithelial stem cell biology; however, the mechanisms regarding the maintenance of these stem cells in their specialized niche remain poorly understood. N-cadherin is a member of the classic cadherin family and has previously been demonstrated to be expressed by hematopoietic stem cells. In the present study, we demonstrate that N-cadherin is expressed by putative stem/progenitor cells, as well as melanocytes, in the human limbal epithelial stem cell niche. In addition, we demonstrate that upon in vitro culture using 3T3 feeder layers, loss of N-cadherin expression occurs with cell proliferation. These results indicate that N-cadherin may be a critical cell-to-cell adhesion molecule between corneal epithelial stem/progenitor cells and their corresponding niche cells in the limbal epithelium.
Collapse
Affiliation(s)
- Ryuhei Hayashi
- Department of Ophthalmology, Tohoku University School of Medicine, 1-1 Seiryomachi, Aoba-ku, Sendai 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
257
|
Zhang J, He XC, Tong WG, Johnson T, Wiedemann LM, Mishina Y, Feng JQ, Li L. Bone morphogenetic protein signaling inhibits hair follicle anagen induction by restricting epithelial stem/progenitor cell activation and expansion. Stem Cells 2006; 24:2826-39. [PMID: 16960130 DOI: 10.1634/stemcells.2005-0544] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Epithelial stem cells (EP-SCs) located in the bulge region of a hair follicle (HF) have the potential to give rise to hair follicle stem/progenitor cells that migrate down to regenerate HFs. Bone morphogenetic protein (BMP) signaling has been shown to regulate the HF cycle by inhibiting anagen induction. Here we show that active BMP signaling functions to prevent EP-SC activation and expansion. Dynamic expression of Noggin, a BMP antagonist, releases EP-SCs from BMP-mediated restriction, leading to EP-SC activation and initiation of the anagen phase. Experimentally induced conditional inactivation of the BMP type IA receptor (Bmpr1a) in EP-SCs leads to overproduction of HF stem/progenitor cells and the eventual formation of matricomas. This genetic manipulation of the BMP signaling pathway also reveals unexpected activation of beta-catenin, a major mediator of Wnt signaling. We propose that BMP activity controls the HF cycle by antagonizing Wnt/beta-catenin activity. This is at least partially achieved by BMP-mediated enhancement of transforming growth factor-beta-regulated epithelial cell-specific phosphatase (PTEN) function. Subsequently, PTEN, through phosphatidyl inositol 3-kinase-Akt, inhibits the activity of beta-catenin, the convergence point of the BMP and Wnt signaling pathways.
Collapse
Affiliation(s)
- Jiwang Zhang
- Stowers Institute for Medical Research, 1000 E 50th Street, Kansas City, Missouri 64110, USA
| | | | | | | | | | | | | | | |
Collapse
|
258
|
Abstract
BACKGROUND Recent studies have demonstrated that the cytokeratin 15 (CK15)-positive stem cells located in the hair follicle bulge are also involved in sebaceous gland renewal. No previous studies have dealt with the CK15 expression in sebaceous neoplasms. METHODS We studied the CK15 expression in 30 sebaceous neoplasms including 10 sebaceomas (sebaceoma defined as a distinct benign neoplasm with sebaceous differentiation), 10 sebaceous neoplasms of Muir-Torre syndrome, and 10 sebaceous carcinomas, in addition to that in the mantles of normal hair follicles. RESULTS CK15 was positive in the undifferentiated sebocytes of the mantles. All 10 sebaceomas showed CK15 expression in the basaloid, germinative cells. Both sebaceous neoplasms in Muir-Torre syndrome and sebaceous carcinomas demonstrated negative or only a focal positive reaction, including the occasional aberrant expression in matured sebocytes, to CK15. CONCLUSIONS CK15 may be a useful marker for stem cells with a sebaceous fate, and a constant CK15 expression in sebaceomas supported the hypothesis that sebaceoma is a benign neoplasm of sebaceous germinative cells in the mantles. The similar staining pattern of CK15 between sebaceous neoplasms in Muir-Torre syndrome and sebaceous carcinomas may be one piece of evidence supporting the hypothesis that most sebaceous neoplasms in Muir-Torre syndrome are low-grade sebaceous carcinomas.
Collapse
Affiliation(s)
- Noriyuki Misago
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan.
| | | |
Collapse
|
259
|
Orringer JS, Hammerberg C, Lowe L, Kang S, Johnson TM, Hamilton T, Voorhees JJ, Fisher GJ. The effects of laser-mediated hair removal on immunohistochemical staining properties of hair follicles. J Am Acad Dermatol 2006; 55:402-7. [PMID: 16908343 DOI: 10.1016/j.jaad.2006.04.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 04/05/2006] [Accepted: 04/16/2006] [Indexed: 12/17/2022]
Abstract
BACKGROUND The mechanisms involved in laser-mediated hair removal remain unclear. One means of reducing hair growth is alteration of follicular stem cells. OBJECTIVE We sought to examine the effects of laser hair removal on the immunohistochemical staining properties of human hair follicles, including the putative stem cells of the bulge region. METHODS Treatment of unwanted axillary hair was performed on one side using an 800 nm-wavelength diode laser and on the other side using a 1064 nm-wavelength neodymium:yttrium-aluminum-garnet laser. Serial skin samples were obtained at baseline and various times after treatment and stained using immunohistochemical techniques. RESULTS Hair shafts were thermally altered, but the immunostaining properties of much of the follicle, including the bulge region, remained generally unchanged. LIMITATIONS This study only addressed the acute immunohistochemical changes found after a single treatment using specific laser parameters. CONCLUSIONS Laser-mediated hair removal does not appear to work by frank destruction of follicular stem cells. Other mechanisms including functional alteration of these cells may underlie the clinical efficacy of the procedure.
Collapse
Affiliation(s)
- Jeffrey S Orringer
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0314, USA.
| | | | | | | | | | | | | | | |
Collapse
|
260
|
Raposio E, Guida C, Baldelli I, Curto M, Fiocca R, Kunkl A, Robello G, Santi PL. Characterization of multipotent cells from human adult hair follicles. Toxicol In Vitro 2006; 21:320-3. [PMID: 17027224 DOI: 10.1016/j.tiv.2006.07.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 07/19/2006] [Accepted: 07/26/2006] [Indexed: 11/20/2022]
Abstract
Recent works demonstrated the presence of a multipotent epithelial cell population in the bulge region of adult human hair follicles. These cells can be cultured in vitro, thus leading to the preparation of dermal-epidermal substitutes which are applicable in the treatment of burns and ulcers. We evaluated the main marker expression in cells obtained from stripped human hair follicles. A pool of hair follicles were incubated at 37 degrees C and 5% CO(2) in a growth medium. The cells were then labelled with antibodies (anti-CD34, anti-CD38, anti-CD45, anti-CD90, anti-CD133, anti-CD146) and analysed by cytometry. We also used hair follicles for immunohistochemical studies, employing antibodies such as CD34, Actin Smooth Muscle, Filaggrin, Desmin, Vimentin, Glial Fibrillary Acidic Protein, Ki-67, PanCytokeratin, CK15, CK19. The cytometry results revealed that a part of bulge cells were CD34+ (1-2%). CD34+ population comprises both large, CD45-, CD133-, CD146- cells and small, CD45+, CD133+, CD146+ cells. Thus, a part of CD34+ cells present a mature endothelial marker (CD146). An expression of the proliferation marker Ki-67 and the stem cell marker CD34 is present in the follicle bulge region. In conclusion, we observed that the stripped hair follicle has the same multipotent cell population as adult and fetal scalp hair follicles.
Collapse
|
261
|
Raveh E, Cohen S, Levanon D, Negreanu V, Groner Y, Gat U. Dynamic expression of Runx1 in skin affects hair structure. Mech Dev 2006; 123:842-50. [PMID: 17011173 DOI: 10.1016/j.mod.2006.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 08/01/2006] [Accepted: 08/03/2006] [Indexed: 12/17/2022]
Abstract
The three mammalian Runx transcription factors, some of which are known to be involved in human genetic diseases and cancer, are pivotal players in embryo development and function as key regulators of cell fate determination and organogenesis. Here, we report the expression of Runx1 during the development of hair and other skin appendages in the mouse and describe the effect of Runx1 on the structural hair output. In hair follicles, where the three Runx proteins are expressed, Runx1 expression is most prominent in both mesenchymal and epithelial compartments. The epithelial expression includes the hair keratin forming layers of the hair shaft and the bulge, where interestingly, Runx1 is co-expressed with keratin 15, a putative hair follicle stem cell marker. In the hair mesenchyme, during early stages of hair morphogenesis, Runx1 is expressed in a discrete dermal sub-epithelial layer, while at later stages it is found in a hair cycle dependent pattern in the dermal papilla. To elucidate the function of Runx1 in the hair follicle we have generated a Runx1 epidermal conditional knockout and found that the mutant mice display a remarkable structural deformation of the zigzag hair type. The data delineate Runx1 as a novel specific marker of several hair follicle cell types and sheds light on its role in hair morphogenesis and differentiation.
Collapse
Affiliation(s)
- Eli Raveh
- Department of Cell and Animal Biology, Silberman Life Sciences Institute, Edmond Safra Campus at Givat-Ram, The Hebrew University, Jerusalem 91904, Israel
| | | | | | | | | | | |
Collapse
|
262
|
Abstract
Putative epithelial stem cells were identified in the hair follicle bulge as quiescent "label retaining cells". The study of these cells was hindered until the identification of bulge cell molecular markers, such as CD34 expression and K15 promoter activity. This allowed for the isolation and characterization of bulge cells from mouse follicles. Bulge cells possess stem cell characteristics, including multipotency, high proliferative potential, and their cardinal feature of quiescence. Lineage analysis demonstrated that all epithelial layers within the adult follicle and hair originated from bulge cells. Bulge cells only contribute to the epidermis during wound healing, but after isolation, when combined with neonatal dermal cells, they regenerate new hair follicles, epidermis, and sebaceous glands. Bulge cells maintain their stem cell characteristics after propagation in vitro, thus ultimately they may be useful for tissue engineering applications. Understanding the signals important for directing movement and differentiation of bulge cells into different lineages will be important for developing treatments based on stem cells as well as clarifying their role in skin disease.
Collapse
Affiliation(s)
- George Cotsarelis
- Department of Dermatology, Kligman Laboratories, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
263
|
Gardner H, Shearstone JR, Bandaru R, Crowell T, Lynes M, Trojanowska M, Pannu J, Smith E, Jablonska S, Blaszczyk M, Tan FK, Mayes MD. Gene profiling of scleroderma skin reveals robust signatures of disease that are imperfectly reflected in the transcript profiles of explanted fibroblasts. ACTA ACUST UNITED AC 2006; 54:1961-73. [PMID: 16736506 DOI: 10.1002/art.21894] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To determine whether biopsy specimens obtained from systemic sclerosis (SSc) lesions show a distinctive gene profile, whether that gene profile is maintained in fibroblasts cultured from SSc skin biopsy specimens, and whether results from tissue obtained from multiple clinical centers can be combined to yield useful observations in this rare disease. METHODS Biopsy samples and passaged fibroblasts were stored in RNAlater solution prior to processing for RNA. RNA from SSc and control skin biopsy specimens, as well as SSc and control explanted passage 4 fibroblasts, from 9 patients and 9 controls was hybridized to Affymetrix HG-U133A arrays. Data were analyzed using the BRB ArrayTools system. When appropriate, findings were followed up with immunohistochemical analysis or TaqMan studies. RESULTS Biopsy samples obtained from patients with SSc had a robust and distinctive gene profile, with approximately 1,800 qualifiers distinguishing normal skin from SSc skin at a significant level. The SSc phenotype was the major driver of sample clusters, independent of origin. Alterations in transforming growth factor beta and Wnt pathways, extracellular matrix proteins, and the CCN family were prominent. Explanted fibroblasts from SSc biopsy samples showed a far smaller subset of changes that were relatively variable between samples, suggesting that either nonfibroblast cell types or other aspects of the dermal milieu are required for full expression of the SSc phenotype. CONCLUSION SSc has a distinct gene profile that is not confounded by geographic location, indicating that extended multicenter studies may be worthwhile to identify distinct subsets of disease by transcript profiling. Explanted SSc fibroblasts show an incomplete reflection of the SSc phenotype.
Collapse
|
264
|
Yu H, Fang D, Kumar SM, Li L, Nguyen TK, Acs G, Herlyn M, Xu X. Isolation of a novel population of multipotent adult stem cells from human hair follicles. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1879-88. [PMID: 16723703 PMCID: PMC1606635 DOI: 10.2353/ajpath.2006.051170] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hair follicles are known to contain a well-characterized niche for adult stem cells: the bulge, which contains epithelial and melanocytic stem cells. Using human embryonic stem cell culture conditions, we isolated a population of adult stem cells from human hair follicles that are distinctively different from known epithelial or melanocytic stem cells. These cells do not express squamous or melanocytic markers but express neural crest and neuron stem cell markers as well as the embryonic stem cell transcription factors Nanog and Oct4. These precursor cells proliferate as spheres, are capable of self-renewal, and can differentiate into multiple lineages. Differentiated cells not only acquire lineage-specific markers but also demonstrate appropriate functions in ex vivo conditions. Most of the Oct4-positive cells in human skin were located in the area highlighted by cytokeratin 15 staining in vivo. Our data suggest that human embryonic stem cell medium can be used to isolate and expand human adult stem cells. Using this method, we isolated a novel population of multipotent adult stem cells from human hair follicles, and these cells appear to be located in the bulge area. Human hair follicles may provide an accessible, autologous source of adult stem cells for therapeutic application.
Collapse
Affiliation(s)
- Hong Yu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
265
|
Nijhof JGW, Braun KM, Giangreco A, van Pelt C, Kawamoto H, Boyd RL, Willemze R, Mullenders LHF, Watt FM, de Gruijl FR, van Ewijk W. The cell-surface marker MTS24 identifies a novel population of follicular keratinocytes with characteristics of progenitor cells. Development 2006; 133:3027-37. [PMID: 16818453 DOI: 10.1242/dev.02443] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We describe a novel murine progenitor cell population localised to a previously uncharacterised region between sebaceous glands and the hair follicle bulge, defined by its reactivity to the thymic epithelial progenitor cell marker MTS24. MTS24 labels a membrane-bound antigen present during the early stages of hair follicle development and in adult mice. MTS24 co-localises with expression of alpha6-integrin and keratin 14, indicating that these cells include basal keratinocytes. This novel population does not express the bulge-specific stem cell markers CD34 or keratin 15, and is infrequently BrdU label retaining. MTS24-positive and -negative keratinocyte populations were isolated by flow cytometry and assessed for colony-forming efficiency. MTS24-positive keratinocytes exhibited a two-fold increase in colony formation and colony size compared to MTS24-negative basal keratinocytes. In addition, both the MTS24-positive and CD34-positive subpopulations were capable of producing secondary colonies after serial passage of individual cell clones. Finally, gene expression profiles of MTS24 and CD34 subpopulations were compared. These results showed that the overall gene expression profile of MTS24-positive cells resembles the pattern previously reported in bulge stem cells. Taken together, these data suggest that the cell-surface marker MTS24 identifies a new reservoir of hair follicle keratinocytes with a proliferative capacity and gene expression profile suggestive of progenitor or stem cells.
Collapse
Affiliation(s)
- Joanne G W Nijhof
- Department of Dermatology, Leiden University Medical Center (LUMC The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
266
|
Andl T, Murchison EP, Liu F, Zhang Y, Yunta-Gonzalez M, Tobias JW, Andl CD, Seykora JT, Hannon GJ, Millar SE. The miRNA-processing enzyme dicer is essential for the morphogenesis and maintenance of hair follicles. Curr Biol 2006; 16:1041-9. [PMID: 16682203 PMCID: PMC2996092 DOI: 10.1016/j.cub.2006.04.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 03/27/2006] [Accepted: 04/12/2006] [Indexed: 12/17/2022]
Abstract
The discovery that microRNAs (miRNAs) play important roles in regulating gene expression via posttranscriptional repression has revealed a previously unsuspected mechanism controlling development and progenitor-cell function (reviewed in ); however, little is known of miRNA functions in mammalian organogenesis. Processing of miRNAs and their assembly into the RNA-induced silencing (RISC) complex requires the essential multifunctional enzyme Dicer . We found that Dicer mRNA and multiple miRNAs are expressed in mouse skin, suggesting roles in skin- and hair-follicle biology. In newborn mice carrying an epidermal-specific Dicer deletion, hair follicles were stunted and hypoproliferative. Hair-shaft and inner-root-sheath differentiation was initiated, but the mutant hair follicles were misoriented and expression of the key signaling molecules Shh and Notch1 was lost by postnatal day 7. At this stage, hair-follicle dermal papillae were observed to evaginate, forming highly unusual structures within the basal epidermis. Normal hair shafts were not produced in the Dicer mutant, and the follicles lacked stem cell markers and degenerated. In contrast to decreased follicular proliferation, the epidermis became hyperproliferative. These results reveal critical roles for Dicer in the skin and implicate miRNAs in key aspects of epidermal and hair-follicle development and function.
Collapse
Affiliation(s)
- Thomas Andl
- Departments of Dermatology and Cell and Developmental Biology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
| | - Elizabeth P. Murchison
- Watson School of Biological Sciences Howard Hughes Medical Institute Cold Spring Harbor Laboratory Cold Spring Harbor, New York 11724
| | - Fei Liu
- Departments of Dermatology and Cell and Developmental Biology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
| | - Yuhang Zhang
- Departments of Dermatology and Cell and Developmental Biology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
| | - Monica Yunta-Gonzalez
- Departments of Dermatology and Cell and Developmental Biology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
| | - John W. Tobias
- Bioinformatics Core University of Pennsylvania Philadelphia, Pennsylvania 19104
| | - Claudia D. Andl
- Gastroenterology Division Department of Medicine Abramson Cancer Center and Family Research Institute University of Pennsylvania Philadelphia, Pennsylvania 19104
| | - John T. Seykora
- Department of Dermatology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
| | - Gregory J. Hannon
- Watson School of Biological Sciences Howard Hughes Medical Institute Cold Spring Harbor Laboratory Cold Spring Harbor, New York 11724
| | - Sarah E. Millar
- Departments of Dermatology and Cell and Developmental Biology University of Pennsylvania School of Medicine Philadelphia Pennsylvania 19104
- Correspondence:
| |
Collapse
|
267
|
Abstract
AIM To investigate a possible follicular origin of extramammary Paget's disease (EPD). EPD is a predominantly intraepidermal tumour with extensive involvement of adnexal structures and high recurrence rates suggesting a follicular stem cell origin. Cytokeratin (CK) 15 and CK19 are considered markers for follicular stem cells located in the hair follicle bulge region. METHODS AND RESULTS Formalin-fixed paraffin-embedded tissues of 12 cases of primary EPD (three anal, nine vulvar) were studied immunohistochemically with antibodies to CK15 and CK19. All cases of EPD showed polygonal Paget cells in the interfollicular epidermis, hair follicles, sebaceous and apocrine glands distributed individually, in nests and in gland-like areas. The polygonal Paget cells were intimately associated with small, flat, mitotically active, 'compressed' keratinocytes. The large Paget cells uniformly expressed CK19 in 12/12 EPD. The small 'compressed' keratinocytes showed strong cytoplasmic CK15 staining in 9/12 EPD with focal accentuation, while the polygonal Paget cells were negative. CONCLUSIONS These histological and immunohistochemical observations allow the following conclusions: (i) the small, flat, 'compressed' keratinocytes are an integral part of EPD; (ii) the dual cell population is reminiscent of sebaceous glands with mature sebocytes and germinative keratinocytes; (iii) since both cell types express cytokeratins typical for follicular differentiation, EPD may be a proliferation of adnexal stem cells residing in the infundibulo-sebaceous unit of hair follicles and adnexal structures.
Collapse
Affiliation(s)
- S Regauer
- Institute of Pathology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
268
|
Abstract
In the last two decades, there has been a tremendous increase in the understanding of stem cell biology, including the field of cutaneous stem cells. Extensive stem cell research and potential clinical applications have provided new perspectives in the use of stem cells in the treatment of human skin disorders such as severe burns and wounds, as well as skin cancer and alopecia. Adult, tissue-specific stem cells are required for tissue homeostasis as well as for the ability to respond to insults such as during wound healing. Fetal wounds can heal rapidly without scars, while in adults wound healing decreases with aging, and this likely represents changes in the functional status of stem cells. In this review, we summarize the main characteristics of cutaneous stem cells in general and present the most recent knowledge in our understanding of these stem cells. We also address the difference in tissue regeneration between fetal and adult tissues in the aspect of stem cell biology.
Collapse
Affiliation(s)
- Cecilia Roh
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | |
Collapse
|
269
|
Essayem S, Kovacic-Milivojevic B, Baumbusch C, McDonagh S, Dolganov G, Howerton K, Larocque N, Mauro T, Ramirez A, Ramos DM, Fisher SJ, Jorcano JL, Beggs HE, Reichardt LF, Ilic D. Hair cycle and wound healing in mice with a keratinocyte-restricted deletion of FAK. Oncogene 2006; 25:1081-9. [PMID: 16247468 PMCID: PMC2710133 DOI: 10.1038/sj.onc.1209130] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Focal adhesion kinase (FAK) is a critical component in transducing signals downstream of both integrins and growth factor receptors. To determine how the loss of FAK affects the epidermis in vivo, we have generated a mouse model with a keratinocyte-restricted deletion of fak (FAKK5 KO mice). FAK(K5 KO) mice displayed three major phenotypes--irregularities of hair cycle, sebaceous glands hypoplasia, and a thinner epidermis--pointing to defects in the proliferative capacity of multipotent stem cells found in the bulge. FAK-null keratinocytes in conventional primary culture undergo massive apoptosis hindering further analyses, whereas the defects observed in vivo do not shorten the mouse lifespan. These results suggest that the structure and the signaling environment of the native tissue may overcome the lack of signaling through FAK. Our findings point to the importance of in vivo and three-dimensional in vitro models in analyses of cell migration, proliferation, and survival. Surprisingly, the difference between FAKloxP/+ and FAKK5 KO mice in wound closure was not statistically significant, suggesting that in vivo loss of FAK does not affect migration/proliferation of basal keratinocytes in the same way as it affects multipotent stem cells of the skin.
Collapse
Affiliation(s)
- S Essayem
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - B Kovacic-Milivojevic
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - C Baumbusch
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - S McDonagh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - G Dolganov
- Department of Pulmonary, University of California San Francisco, San Francisco, CA, USA
| | - K Howerton
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - N Larocque
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - T Mauro
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - A Ramirez
- Department of Epithelial Damage, Repair and Tissue Engineering Program, CIEMAT, Madrid, Spain
| | - DM Ramos
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
| | - SJ Fisher
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - JL Jorcano
- Department of Epithelial Damage, Repair and Tissue Engineering Program, CIEMAT, Madrid, Spain
| | - HE Beggs
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - LF Reichardt
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - D Ilic
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
270
|
Cotsarelis G. Gene expression profiling gets to the root of human hair follicle stem cells. J Clin Invest 2006; 116:19-22. [PMID: 16395398 PMCID: PMC1323274 DOI: 10.1172/jci27490] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Hair follicle stem cells sustain growth and cycling of the hair follicle and are located in the permanent portion of the follicle known as the bulge. In this issue of the JCI, Ohyama et al. report the characterization of global gene expression patterns of human hair follicle stem cells after their isolation using sophisticated laser capture techniques to microdissect out bulge cells. They discovered a panel of cell surface markers useful for isolating living hair follicle stem cells, a finding with potential therapeutic implications since isolated stem cells in mice can generate new hair follicles when transplanted to other mice. The findings of Ohyama et al. validate the use of the mouse for studying hair follicle biology but also underscore critical differences between mouse and human stem cell markers. In particular, CD34, which delineates hair follicle stem cells in the mouse, is not expressed by human hair follicle stem cells, while CD200 is expressed by stem cells in both species. Ultimately, this information will assist efforts to develop cell-based and cell-targeted treatments for skin disease.
Collapse
Affiliation(s)
- George Cotsarelis
- University of Pennsylvania School of Medicine, M8 Stellar-Chance Laboratories, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
271
|
Roh C, Tao Q, Photopoulos C, Lyle S. In vitro differences between keratinocyte stem cells and transit-amplifying cells of the human hair follicle. J Invest Dermatol 2006; 125:1099-105. [PMID: 16354178 DOI: 10.1111/j.0022-202x.2005.23958.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Epithelial stem cells within the human hair follicle are critical for hair development, hair cycling, wound healing, and tumorigenesis. We and others have previously shown that the hair follicle bulge area contains keratinocyte stem cells, whereas the hair matrix represents the proliferating and differentiating transit-amplifying (TA) cell compartment. In order to better characterize the phenotypic differences between human keratinocyte stem cells and their daughter TA cells, we compared the in vitro properties of cell adhesion, cell migration, clonogenicity, and in vitro life span. Epithelial outgrowths from the hair matrix appeared within 2 d of explant, whereas stem cell outgrowths appeared between 7 and 10 d after explant. Both populations form colonies; however, stem cells from telogen follicles formed more total colonies, and more colonies greater than 3 mm. Upon subculture, stem cells formed colonies until passage 6 and terminally differentiated at passage 7, whereas TA cells only formed colonies until passage 2. Stem cells express more beta1 integrin and adhere more rapidly to collagen IV. Most strikingly, TA cells showed a 7-fold greater mobility on migration assays than stem cells (0.704 vs 0.102 microm per min). These results help define the human hair follicle stem cell and TA cell phenotypes and correlate with the in vivo properties of these compartments.
Collapse
Affiliation(s)
- Cecilia Roh
- Pathology Department, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
272
|
Levy V, Lindon C, Harfe BD, Morgan BA. Distinct stem cell populations regenerate the follicle and interfollicular epidermis. Dev Cell 2006; 9:855-61. [PMID: 16326396 DOI: 10.1016/j.devcel.2005.11.003] [Citation(s) in RCA: 339] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2005] [Revised: 10/19/2005] [Accepted: 11/07/2005] [Indexed: 12/17/2022]
Abstract
The regeneration of the skin and its appendages is thought to occur by the regulated activation of a dedicated stem cell population. A population of cells in the bulge region of the hair follicle has been identified as the putative stem cell of both the follicle and the interfollicular epidermis. While this assertion is supported by a variety of surrogate assays, there has been no direct confirmation of the normal contribution of these cells to the regeneration of structures other than the cycling portion of the hair follicle. Here, we report lineage analysis revealing that the follicular epithelium is derived from cells in the epidermal placode that express Sonic hedgehog. This analysis also demonstrates that the stem cells resident in the follicular bulge that regenerate the follicle are neither the stem cells of the epidermis nor the source of the stem cells of the epidermis in the absence of trauma.
Collapse
Affiliation(s)
- Vered Levy
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
273
|
Vodicka P, Smetana K, Dvoránková B, Emerick T, Xu YZ, Ourednik J, Ourednik V, Motlík J. The Miniature Pig as an Animal Model in Biomedical Research. Ann N Y Acad Sci 2006; 1049:161-71. [PMID: 15965115 DOI: 10.1196/annals.1334.015] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Crucial prerequisites for the development of safe preclinical protocols in biomedical research are suitable animal models that would allow for human-related validation of valuable research information gathered from experimentation with lower mammals. In this sense, the miniature pig, sharing many physiological similarities with humans, offers several breeding and handling advantages (when compared to non-human primates), making it an optimal species for preclinical experimentation. The present review offers several examples taken from current research in the hope of convincing the reader that the porcine animal model has gained massively in importance in biomedical research during the last few years. The adduced examples are taken from the following fields of investigation: (a) the physiology of reproduction, where pig oocytes are being used to study chromosomal abnormalities (aneuploidy) in the adult human oocyte; (b) the generation of suitable organs for xenotransplantation using transgene expression in pig tissues; (c) the skin physiology and the treatment of skin defects using cell therapy-based approaches that take advantage of similarities between pig and human epidermis; and (d) neurotransplantation using porcine neural stem cells grafted into inbred miniature pigs as an alternative model to non-human primates xenografted with human cells.
Collapse
Affiliation(s)
- Petr Vodicka
- Institute of Animal Physiology and Genetics, Department of Physiology of Reproduction, Rumburská 89, Libechov 277 21, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
274
|
Abstract
Major progress in understanding epithelial skin stem cells has been accomplished. This has been possible by developing new methods for labeling, tracking, isolating, and characterizing enriched populations of stem cells. This chapter summarizes in vivo and in vitro assays that are currently employed to analyze skin epithelial stem cells. Despite progress, the definition of a stem cell is currently a functional one. Unambiguous identification of a stem cell in intact tissue is still not possible. These limitations hamper molecular studies aimed at unraveling the cellular mechanisms operating in the stem cell compartment. This chapter emphasizes current methods for analyzing hair follicle stem cells, as opposed to other epithelial compartments, because the hair follicle has been most intensively studied up to date.
Collapse
Affiliation(s)
- Tudorita Tumbar
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
275
|
Misago N, Narisawa Y. Cytokeratin 15 expression in apocrine mixed tumors of the skin and other benign neoplasms with apocrine differentiation. J Dermatol 2006; 33:2-9. [PMID: 16469077 DOI: 10.1111/j.1346-8138.2006.00001.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To clarify the features of apocrine mixed tumors (AMT) of the skin among benign neoplasms with apocrine differentiation in their relationship to follicular stem cells, we investigated the immunohistochemical expression of CK15 (LHK15 and C8/144B), which is a relatively specific marker of hair follicle stem cells in the bulge, in 35 cases of eight different benign neoplasms with presumed apocrine differentiation. All eight cases of AMT of the skin showed CK15 immunostaining of the neoplastic cells, and all four cases of syringocystadenoma papilliferum, all five cases of spiradenoma, and both cases of cylindroma also showed a focally positive reaction to CK15. None of the other benign neoplasms with presumed apocrine differentiation showed CK15 expression. In AMT of the skin, the proportion of CK15-positive cells in the follicular or sebaceous differentiation group (78.8%, average of four cases) was significantly higher than the group without this differentiation (8.8%, average of four cases). AMT of the skin are unique among benign neoplasms with apocrine differentiation in their substantial and constant CK15 expression, suggesting that they derive from multipotent epithelial stem cells in the bulge. AMT of the skin with follicular or sebaceous differentiation are considered to show an immature stage of apocrine differentiation still rich in stem cells or to originate from stem cells with an incompletely established apocrine fate. The partially positive reaction for CK15 in syringocystadenomas papilliferum and spiradenoma/cylindroma may depend on the ability to express CK15 in stem cells with an apocrine fate or result from the follicular and apocrine nature of this neoplasm.
Collapse
Affiliation(s)
- Noriyuki Misago
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan.
| | | |
Collapse
|
276
|
Abstract
Current understanding of the biology of epidermal stem cells opens a totally new perspective in the function of the epidermis and adjacent epithelial structures. A number of pathogenetic as well as clinical-therapeutic approaches against a variety of dermatoses may become possible with knowledge about keratinocyte proliferation, differentiation and regeneration. The reservoir of epidermal stem cells is located in the interfollicular epidermis, the hair follicle area and the germinal hair follicle matrix. Endogenous stem cell clones exist here, giving rise to transient amplifying cells and postmitotic cells. The stem cell clones are organized in clusters and display high expression of adhesion proteins, which guarantee their stability in a specific environment consisting of different cell types and extracellular substrates in the stratum basale. Differentiation is determined by a specific cascade of chemical signals from the stem cell environment and from the genetic program of the cell. The clinical relevance of stem cells lies primarily in their therapeutic potential with reconstruction of epithelia by reimplantation of autologous stem cells or gene therapeutic applications such as targeted transfection. However, the benefit-to-risk ratio cannot yet be accurately estimated.
Collapse
Affiliation(s)
- Karin Rzepka
- Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg
| | | | | | | |
Collapse
|
277
|
Ito M, Liu Y, Yang Z, Nguyen J, Liang F, Morris RJ, Cotsarelis G. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat Med 2005; 11:1351-4. [PMID: 16288281 DOI: 10.1038/nm1328] [Citation(s) in RCA: 948] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Accepted: 10/18/2005] [Indexed: 12/16/2022]
Abstract
The discovery of long-lived epithelial stem cells in the bulge region of the hair follicle led to the hypothesis that epidermal renewal and epidermal repair after wounding both depend on these cells. To determine whether bulge cells are necessary for epidermal renewal, here we have ablated these cells by targeting them with a suicide gene encoding herpes simplex virus thymidine kinase (HSV-TK) using a Keratin 1-15 (Krt1-15) promoter. We show that ablation leads to complete loss of hair follicles but survival of the epidermis. Through fate-mapping experiments, we find that stem cells in the hair follicle bulge do not normally contribute cells to the epidermis which is organized into epidermal proliferative units, as previously predicted. After epidermal injury, however, cells from the bulge are recruited into the epidermis and migrate in a linear manner toward the center of the wound, ultimately forming a marked radial pattern. Notably, although the bulge-derived cells acquire an epidermal phenotype, most are eliminated from the epidermis over several weeks, indicating that bulge stem cells respond rapidly to epidermal wounding by generating short-lived 'transient amplifying' cells responsible for acute wound repair. Our findings have implications for both gene therapy and developing treatments for wounds because it will be necessary to consider epidermal and hair follicle stem cells as distinct populations.
Collapse
Affiliation(s)
- Mayumi Ito
- Department of Dermatology, Kligman Laboratories, University of Pennsylvania School of Medicine, M8 Stellar-Chance Building, 422 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
278
|
Selleri S, Arnaboldi F, Palazzo M, Hussein U, Balsari A, Rumio C. Caveolin-1 is expressed on multipotent cells of hair follicles and might be involved in their resistance to chemotherapy. Br J Dermatol 2005; 153:506-13. [PMID: 16120134 DOI: 10.1111/j.1365-2133.2005.06746.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Caveolin-1 is the principal protein that composes caveolae, which are vesicular invaginations present on the plasma membrane of different cell types. Caveolae are involved in a variety of cellular functions including regulation of proliferation rate and resistance to chemotherapeutic drugs. Chemotherapy frequently induces alopecia which is reversible most probably due to the low proliferative rate of hair follicle stem cells and due to the expression of proteins which confer resistance. OBJECTIVES Using a specific animal model and immunohistochemistry, we analysed the expression of both caveolin-1 and the cell proliferation marker beta-catenin, at different stages of the hair follicle cycle, both before and after doxorubicin (DXR) -induced alopecia. METHODS Seven-week-old C57BL/6 mice were depilated in order to synchronize hair follicle cycle in the anagen phase. Chemotherapy with DXR 15 mg kg(-1) was used to induce alopecia. Control and treated mice were then sacrificed at precise time points and caveolin-1 expression in hairs at different stages of the cycle were analysed by immunohistochemistry. By double immunofluorescence, colocalization of caveolin-1 and cytokeratin-15 was confirmed in the bulge region. The state of proliferation of cells composing hair follicle was assessed by beta-catenin immunohistochemistry. RESULTS Caveolin-1 was expressed by the cells of the bulge area, the multipotent compartment of the hair follicle, during all phases of growth (anagen), regression (catagen) and resting (telogen). During the anagen phases, nuclear beta-catenin labelling was not observed in bulge cells, but rather in the deeper portion of the follicle. Damaged hair follicles from DXR-treated mice presented bulge cells which still expressed caveolin-1, suggesting that this protein might play a role in their drug resistance. As expected, no beta-catenin nuclear staining was detectable in DXR-treated hair follicles, indicating the complete lack of proliferative processes. The differential localization of caveolin-1 and beta-catenin suggests that the mutually exclusive expression of these proteins is useful for correct hair regrowth, whether during the physiological cycle or after chemotherapy-induced alopecia. CONCLUSIONS Expression of caveolin-1 within the multipotent cell compartment of the hair follicle can explain the resistance of bulge cells to many chemotherapeutics, suggested by the reversibility of chemotherapy-induced alopecia.
Collapse
Affiliation(s)
- S Selleri
- Department of Human Morphology, National Cancer Institute, Università degli Studi di Milano, Via Mangiagalli, 31, 20133 Milan, Italy
| | | | | | | | | | | |
Collapse
|
279
|
Bianchi N, Depianto D, McGowan K, Gu C, Coulombe PA. Exploiting the keratin 17 gene promoter to visualize live cells in epithelial appendages of mice. Mol Cell Biol 2005; 25:7249-59. [PMID: 16055733 PMCID: PMC1190235 DOI: 10.1128/mcb.25.16.7249-7259.2005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Keratin genes afford, given their large number (>50) and differential regulation, a unique opportunity to study the mechanisms underlying specification and differentiation in epithelia of higher metazoans. Moreover, the small size and regulation in cis of many keratin genes enable the use of their regulatory sequence to achieve targeted gene expression in mice. Here we show that 2 kilobases of 5' upstream region from the mouse keratin 17 gene (mK17) confers expression of green fluorescent protein (GFP) in major epithelial appendages of transgenic mice. Like that of mK17, onset of [mK17 5']-GFP reporter expression coincides with the appearance of ectoderm-derived epithelial appendages during embryonic development. In adult mice, [mK17 5']-GFP is appropriately regulated within hair, nail, glands, and oral papilla. Tracking of GFP fluorescence allows for the visualization of growth cycle-related changes in hair follicles, and the defects engendered by the hairless mutation, in live skin tissue. Deletion of an internal 48-bp interval, which encompasses a Gli-responsive element, from this promoter results in loss of GFP fluorescence in most appendages in vivo, suggesting that sonic hedgehog participates in K17 regulation. The compact mK17 gene promoter provides a novel tool for appendage-preferred gene expression and manipulation in transgenic mice.
Collapse
Affiliation(s)
- Nicholas Bianchi
- Predoctoral Program in Human Genetics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 212105, USA
| | | | | | | | | |
Collapse
|
280
|
Soma T, Tajima M, Kishimoto J. Hair cycle-specific expression of versican in human hair follicles. J Dermatol Sci 2005; 39:147-54. [PMID: 15871917 DOI: 10.1016/j.jdermsci.2005.03.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 03/22/2005] [Accepted: 03/28/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Versican, a large chondroitin sulfate proteoglycan molecule, is implicated in the induction of hair morphogenesis, the initiation of hair regeneration, and the maintenance of hair growth in mouse species. In contrast, in human hair follicles, the distribution and the roles of versican remains obscure. OBJECTIVES To elucidate the implication of versican in normal human hair growth. METHODS Versican expression was examined by in situ hybridization (mRNA) and immunohistochemistry (protein). RESULTS The results clearly showed specific versican gene expression in the dermal papilla of anagen, which apparently decreased in the dermal papilla of catagen hair follicles. No specific signal was detectable in telogen hair follicles. Consistent with ISH results, versican immunoreactivity was extended over the dermal papilla of anagen hair follicles, and again, this staining diminished in the catagen phase of human hair follicles. Interestingly, versican proteins were deposited outside K15-positive epithelial cells in the bulge throughout the hair cycle. Versican immunoreactivity in the dermal papilla was almost lost in vellus-like hair follicles affected by male pattern baldness. CONCLUSION Specific expression of versican in the anagen hair follicles suggests its importance to maintain the normal growing phase of human as well as mouse.
Collapse
Affiliation(s)
- Tsutomu Soma
- Shiseido Life Science Research Center, 2-12-1 Fukuura, Kanazawa-ku, Yokohama 236-8643, Japan.
| | | | | |
Collapse
|
281
|
Morasso MI, Tomic-Canic M. Epidermal stem cells: the cradle of epidermal determination, differentiation and wound healing. Biol Cell 2005; 97:173-83. [PMID: 15715523 PMCID: PMC1283090 DOI: 10.1042/bc20040098] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The field of epidermal stem cells has dramatically advanced in the last decade, leading to a better understanding of the molecular factors, signalling pathways and cellular events that identify and characterize stem cells, thus revealing their immense potential for therapeutic use. Furthermore, multipotent epidermal stem cells present the major advantage of easy accessibility with the discovery of their specific location within the bulge of the hair follicle. This review focuses on the most recent findings on epidermal stem cells, and their potential role in initial epidermal commitment, differentiation and wound healing processes in the skin.
Collapse
Affiliation(s)
- Maria I Morasso
- Developmental Skin Biology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
282
|
|
283
|
Sinha S. Regulation of intermediate filament gene expression. Methods Cell Biol 2005; 78:267-96. [PMID: 15646622 DOI: 10.1016/s0091-679x(04)78010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Affiliation(s)
- Satrajit Sinha
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, New York 14214, USA
| |
Collapse
|
284
|
Hutchin ME, Kariapper MST, Grachtchouk M, Wang A, Wei L, Cummings D, Liu J, Michael LE, Glick A, Dlugosz AA. Sustained Hedgehog signaling is required for basal cell carcinoma proliferation and survival: conditional skin tumorigenesis recapitulates the hair growth cycle. Genes Dev 2004; 19:214-23. [PMID: 15625189 PMCID: PMC545881 DOI: 10.1101/gad.1258705] [Citation(s) in RCA: 453] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Temporally and spatially constrained Hedgehog (Hh) signaling regulates cyclic growth of hair follicle epithelium while constitutive Hh signaling drives the development of basal cell carcinomas (BCCs), the most common cancers in humans. Using mice engineered to conditionally express the Hh effector Gli2, we show that continued Hh signaling is required for growth of established BCCs. Transgene inactivation led to BCC regression accompanied by reduced tumor cell proliferation and increased apoptosis, leaving behind a small subset of nonproliferative cells that could form tumors upon transgene reactivation. Nearly all BCCs arose from hair follicles, which harbor cutaneous epithelial stem cells, and reconstitution of regressing tumor cells with an inductive mesenchyme led to multilineage differentiation and hair follicle formation. Our data reveal that continued Hh signaling is required for proliferation and survival of established BCCs, provide compelling support for the concept that these tumors represent an aberrant form of follicle organogenesis, and uncover potential limitations to treating BCCs using Hh pathway inhibitors.
Collapse
Affiliation(s)
- Mark E Hutchin
- Department of Dermatology and Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
285
|
Fernandes KJL, McKenzie IA, Mill P, Smith KM, Akhavan M, Barnabé-Heider F, Biernaskie J, Junek A, Kobayashi NR, Toma JG, Kaplan DR, Labosky PA, Rafuse V, Hui CC, Miller FD. A dermal niche for multipotent adult skin-derived precursor cells. Nat Cell Biol 2004; 6:1082-93. [PMID: 15517002 DOI: 10.1038/ncb1181] [Citation(s) in RCA: 569] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A fundamental question in stem cell research is whether cultured multipotent adult stem cells represent endogenous multipotent precursor cells. Here we address this question, focusing on SKPs, a cultured adult stem cell from the dermis that generates both neural and mesodermal progeny. We show that SKPs derive from endogenous adult dermal precursors that exhibit properties similar to embryonic neural-crest stem cells. We demonstrate that these endogenous SKPs can first be isolated from skin during embryogenesis and that they persist into adulthood, with a niche in the papillae of hair and whisker follicles. Furthermore, lineage analysis indicates that both hair and whisker follicle dermal papillae contain neural-crest-derived cells, and that SKPs from the whisker pad are of neural-crest origin. We propose that SKPs represent an endogenous embryonic precursor cell that arises in peripheral tissues such as skin during development and maintains multipotency into adulthood.
Collapse
Affiliation(s)
- Karl J L Fernandes
- Department of Cancer Research, Hospital For Sick Children Research Institute, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
286
|
Abstract
With the development of advanced cell-labeling technologies, fluorescence activated cell sorting (FACS), as well as improved understanding of mammalian gene expression, it is now possible to identify and isolate specific sub-populations of adult mammalian cells with good accuracy. Recent publications by Morris et al. and Tumbar et al. demonstrate the isolation of putative epithelial stem cells from the hair follicle bulge and Affymetrix expression arrays were employed to elucidate putative genes that might control stem cell fates.
Collapse
Affiliation(s)
- Po-Lin So
- Department of Dermatology, University of California-San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
287
|
Abstract
Skin cancers as seen in the clinic are the result of a long history of events of which only the final stages are easily observed. As normal cells progress to the neoplastic and later metastatic stages, multiple changes in gene expression and cellular phenotypes occur. Nevertheless, the early events in the pathway leading from the first exposure to carcinogenic or mutagenic agents to a frank tumor are thought to involve a two-step process of tumor initiation and tumor promotion. In experimental two-stage skin carcinogenesis in mice, benign and malignant neoplasms can be induced on the backs of mice following a low, or sub-threshold, exposure to a carcinogen (initiation) and subsequent chronic regenerative epidermal hyperplasia caused by a variety of physical, chemical, or biological agents (promotion). Tumor initiation is thought to involve conversion of some of the epidermal cells into latent neoplastic cells, whereas promotion elicits expression of the neoplastic change. Many questions remain about this process, in particular the identity and biological properties of the cells that are specifically the targets of tumor initiation and promotion. Conceivably, any proliferative cell could become and remain initiated; however, these rare cells in the cutaneous epithelium able to become neoplastic cells after exposure to carcinogens and tumor promoters have many of the properties of stem cells. Although this concept that stem cells are the target cells in the development of cancer is not new, I will consider here the evidence that the target cells are indeed stem cells in the cutaneous epithelium.
Collapse
Affiliation(s)
- Rebecca J Morris
- Department of Dermatology, Columbia University, College of Physicians and Surgeons, 630 West 168th Street, VC-15, New York, NY 10032, USA.
| |
Collapse
|
288
|
Roh C, Tao Q, Lyle S. Dermal papilla-induced hair differentiation of adult epithelial stem cells from human skin. Physiol Genomics 2004; 19:207-17. [PMID: 15292489 DOI: 10.1152/physiolgenomics.00134.2004] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The epithelial-mesenchymal interactions between keratinocyte stem cells and dermal papilla (DP) cells are crucial for normal development of the hair follicle as well as during hair cycling. During the cyclical regrowth of a new lower follicle, the multipotent hair follicle stem cells are stimulated to proliferate and differentiate through interactions with the underlying mesenchymal DP cells. To characterize the events occurring during the process of epithelial stem cell fate determination, we utilized a coculture system by incubating human hair follicle keratinocyte stem cells with DP cells. Using GeneChip microarrays, we analyzed changes in gene expression within the stem cells upon coculture with the DP over a 5-day time course. A number of important signaling pathways and growth factors were regulated. The hair-specific keratin 6hf (K6hf) gene proved a particularly good marker of hair differentiation, with a 7.9-fold increase in mRNA and resulting increased protein levels. The high expression of K6hf was unique to DP-induced keratinocyte differentiation, since expression of K6hf was not induced by high calcium. Since the beta-catenin signaling pathway has been implicated in hair follicle development, we examined the role of beta-catenin in our system and demonstrated that beta-catenin/lef-1 signaling is required for DP-induced hair differentiation.
Collapse
Affiliation(s)
- Cecilia Roh
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
289
|
Morris RJ, Liu Y, Marles L, Yang Z, Trempus C, Li S, Lin JS, Sawicki JA, Cotsarelis G. Capturing and profiling adult hair follicle stem cells. Nat Biotechnol 2004; 22:411-7. [PMID: 15024388 DOI: 10.1038/nbt950] [Citation(s) in RCA: 986] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Accepted: 02/06/2004] [Indexed: 12/16/2022]
Abstract
The hair follicle bulge possesses putative epithelial stem cells. Characterization of these cells has been hampered by the inability to target bulge cells genetically. Here, we use a Keratin1-15 (Krt1-15, also known as K15) promoter to target mouse bulge cells with an inducible Cre recombinase construct or with the gene encoding enhanced green fluorescent protein (EGFP), which allow for lineage analysis and for isolation of the cells. We show that bulge cells in adult mice generate all epithelial cell types within the intact follicle and hair during normal hair follicle cycling. After isolation, adult Krt1-15-EGFP-positive cells reconstituted all components of the cutaneous epithelium and had a higher proliferative potential than Krt1-15-EGFP-negative cells. Genetic profiling of hair follicle stem cells revealed several known and unknown receptors and signaling pathways important for maintaining the stem cell phenotype. Ultimately, these findings provide potential targets for the treatment of hair loss and other disorders of skin and hair.
Collapse
Affiliation(s)
- Rebecca J Morris
- Department of Dermatology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|