101
|
Zhang H, Zhang S, Zhao H, Qiao J, Liu S, Deng Z, Lei X, Ning L, Cao Y, Zhao Y, Duan E. Ovine Hair Follicle Stem Cells Derived from Single Vibrissae Reconstitute Haired Skin. Int J Mol Sci 2015; 16:17779-97. [PMID: 26247934 PMCID: PMC4581221 DOI: 10.3390/ijms160817779] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 12/17/2022] Open
Abstract
Hair follicle stem cells (HFSCs) possess fascinating self-renewal capacity and multipotency, which play important roles in mammalian hair growth and skin wound repair. Although HFSCs from other mammalian species have been obtained, the characteristics of ovine HFSCs, as well as the methods to isolate them have not been well addressed. Here, we report an efficient strategy to obtain multipotent ovine HFSCs. Through microdissection and organ culture, we obtained keratinocytes that grew from the bulge area of vibrissa hair follicles, and even abundant keratinocytes were harvested from a single hair follicle. These bulge-derived keratinocytes are highly positive for Krt15, Krt14, Tp63, Krt19 and Itga6; in addition to their strong proliferation abilities in vitro, these keratinocytes formed new epidermis, hair follicles and sebaceous glands in skin reconstitution experiments, showing that these are HFSCs from the bulge outer root sheath. Taken together, we developed an efficient in vitro system to enrich ovine HFSCs, providing enough HFSCs for the investigations about the ovine hair cycle, aiming to promote wool production in the future.
Collapse
Affiliation(s)
- Huishan Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shoubing Zhang
- Department of Histology & Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Huashan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jingqiao Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shuang Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhili Deng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xiaohua Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Lina Ning
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yujing Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Enkui Duan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
102
|
Li A, Lai YC, Figueroa S, Yang T, Widelitz RB, Kobielak K, Nie Q, Chuong CM. Deciphering principles of morphogenesis from temporal and spatial patterns on the integument. Dev Dyn 2015; 244:905-20. [PMID: 25858668 PMCID: PMC4520785 DOI: 10.1002/dvdy.24281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/04/2015] [Accepted: 04/03/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND How tissue patterns form in development and regeneration is a fundamental issue remaining to be fully understood. The integument often forms repetitive units in space (periodic patterning) and time (cyclic renewal), such as feathers and hairs. Integument patterns are visible and experimentally manipulatable, helping us reveal pattern formative processes. Variability is seen in regional phenotypic specificities and temporal cycling at different physiological stages. RESULTS Here we show some cellular/molecular bases revealed by analyzing integument patterns. (1) Localized cellular activity (proliferation, rearrangement, apoptosis, differentiation) transforms prototypic organ primordia into specific shapes. Combinatorial positioning of different localized activity zones generates diverse and complex organ forms. (2) Competitive equilibrium between activators and inhibitors regulates stem cells through cyclic quiescence and activation. CONCLUSIONS Dynamic interactions between stem cells and their adjacent niche regulate regenerative behavior, modulated by multi-layers of macro-environmental factors (dermis, body hormone status, and external environment). Genomics studies may reveal how positional information of localized cellular activity is stored. In vivo skin imaging and lineage tracing unveils new insights into stem cell plasticity. Principles of self-assembly obtained from the integumentary organ model can be applied to help restore damaged patterns during regenerative wound healing and for tissue engineering to rebuild tissues. Developmental Dynamics 244:905-920, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ang Li
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Yung-Chih Lai
- Department of Pathology, University of Southern California, Los Angeles, California
- Center for Developmental Biology and Regenerative Medicine, Taiwan University, Taipei, Taiwan
| | - Seth Figueroa
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Tian Yang
- Department of Cell Biology, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Krzysztof Kobielak
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, California
| | - Cheng Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California
- Center for Developmental Biology and Regenerative Medicine, Taiwan University, Taipei, Taiwan
- Stem Cell and Regenerative Medicine Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
103
|
Zgheib C, Xu J, Mallette AC, Caskey RC, Zhang L, Hu J, Liechty KW. SCF increases in utero-labeled stem cells migration and improves wound healing. Wound Repair Regen 2015; 23:583-90. [PMID: 26032674 DOI: 10.1111/wrr.12318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/27/2015] [Indexed: 12/17/2022]
Abstract
Diabetic skin wounds lack the ability to heal properly and constitute a major and significant complication of diabetes. Nontraumatic lower extremity amputations are the number one complication of diabetic skin wounds. The complexity of their pathophysiology requires an intervention at many levels to enhance healing and wound closure. Stem cells are a promising treatment for diabetic skin wounds as they have the ability to correct abnormal healing. Stem cell factor (SCF), a chemokine expressed in the skin, can induce stem cells migration, however the role of SCF in diabetic skin wound healing is still unknown. We hypothesize that SCF would correct the impairment and promote the healing of diabetic skin wounds. Our results show that SCF improved wound closure in diabetic mice and increased HIF-1α and vascular endothelial growth factor (VEGF) expression levels in these wounds. SCF treatment also enhanced the migration of red fluorescent protein (RFP)-labeled skin stem cells via in utero intra-amniotic injection of lenti-RFP at E8. Interestingly these RFP+ cells are present in the epidermis, stain negative for K15, and appear to be distinct from the already known hair follicle stem cells. These results demonstrate that SCF improves diabetic wound healing in part by increasing the recruitment of a unique stem cell population present in the skin.
Collapse
Affiliation(s)
- Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Junwang Xu
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | | | - Robert C Caskey
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Liping Zhang
- University of Mississippi Medical Center, Jackson, Mississippi
| | - Junyi Hu
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
104
|
Goodell MA, Nguyen H, Shroyer N. Somatic stem cell heterogeneity: diversity in the blood, skin and intestinal stem cell compartments. Nat Rev Mol Cell Biol 2015; 16:299-309. [PMID: 25907613 PMCID: PMC5317203 DOI: 10.1038/nrm3980] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Somatic stem cells replenish many tissues throughout life to repair damage and to maintain tissue homeostasis. Stem cell function is frequently described as following a hierarchical model in which a single master cell undergoes self-renewal and differentiation into multiple cell types and is responsible for most regenerative activity. However, recent data from studies on blood, skin and intestinal epithelium all point to the concomitant action of multiple types of stem cells with distinct everyday roles. Under stress conditions such as acute injury, the surprising developmental flexibility of these stem cells enables them to adapt to diverse roles and to acquire different regeneration capabilities. This paradigm shift raises many new questions about the developmental origins, inter-relationships and molecular regulation of these multiple stem cell types.
Collapse
Affiliation(s)
- Margaret A Goodell
- Stem Cells and Regenerative Medicine Center and Departments of Pediatrics, Molecular and Cellular Biology, and Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Hoang Nguyen
- Stem Cells and Regenerative Medicine Center and Departments of Pediatrics, Molecular and Cellular Biology, and Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Noah Shroyer
- Stem Cells and Regenerative Medicine Center and Departments of Pediatrics, Molecular and Cellular Biology, and Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
105
|
Bai X, Lei M, Shi J, Yu Y, Qiu W, Lai X, Liu Y, Yang T, Yang L, Widelitz RB, Chuong CM, Lian X. Roles of GasderminA3 in Catagen-Telogen Transition During Hair Cycling. J Invest Dermatol 2015; 135:2162-2172. [PMID: 25860385 PMCID: PMC4537385 DOI: 10.1038/jid.2015.147] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 03/03/2015] [Accepted: 03/30/2015] [Indexed: 01/08/2023]
Abstract
Hair follicles undergo cyclic behavior through regression (catagen), rest (telogen) and regeneration (anagen) during postnatal life. The hair cycle transition is strictly regulated by the autonomous and extrinsic molecular environment. However, whether there is a switch controlling catagen-telogen transition remains largely unknown. Here we show that hair follicles cycle from catagen to the next anagen without transitioning through a morphologically typical telogen after Gsdma3 mutation. This leaves an ESLS (epithelial strand-like structure) during the time period corresponding to telogen phase in WT mice. Molecularly, Wnt10b is upregulated in Gsdma3 mutant mice. Restoration of Gsdma3 expression in AE (alopecia and excoriation) mouse skin rescues hair follicle telogen entry and significantly decreases the Wnt10b-mediated Wnt/β-catenin signaling pathway. Overexpression of Wnt10b inhibits telogen entry by increasing epithelial strand cell proliferation. Subsequently, hair follicles with a Gsdma3 mutation enter the second anagen simultaneously as WT mice. Hair follicles cannot enter the second anagen with ectopic WT Gsdma3 overexpression. A luciferase reporter assay proves Gsdma3 directly suppresses Wnt signaling. Our findings suggest Gsdma3 plays an important role in catagen-telogen transition by balancing the Wnt signaling pathway, and that morphologically typical telogen is not essential for the initiation of a new hair cycle.
Collapse
Affiliation(s)
- Xiufeng Bai
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Mingxing Lei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, People's Republic of China; '111' Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing, People's Republic of China; Department of Pathology, University of Southern California, Los Angeles, California, USA.
| | - Jiazhong Shi
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yu Yu
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Weiming Qiu
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiangdong Lai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, People's Republic of China; '111' Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing, People's Republic of China
| | - Yingxin Liu
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Tian Yang
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, People's Republic of China; '111' Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing, People's Republic of China
| | - Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California, USA
| | - Xiaohua Lian
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
106
|
Segrelles C, García-Escudero R, Garín MI, Aranda JF, Hernández P, Ariza JM, Santos M, Paramio JM, Lorz C. Akt signaling leads to stem cell activation and promotes tumor development in epidermis. Stem Cells 2015; 32:1917-28. [PMID: 24504902 DOI: 10.1002/stem.1669] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 01/18/2014] [Indexed: 01/25/2023]
Abstract
Hair follicle stem cells (HF-SCs) alternate between periods of quiescence and proliferation, to finally differentiate into all the cell types that constitute the hair follicle. Also, they have been recently identified as cells of origin in skin cancer. HF-SCs localize in a precise region of the hair follicle, the bulge, and molecular markers for this population have been established. Thus, HF-SCs are good model to study the potential role of oncogenic activations on SC physiology. Expression of a permanently active form of Akt (myrAkt) in basal cells leads to Akt hyperactivation specifically in the CD34(+)Itga6(H) population. This activation causes bulge stem cells to exit from quiescence increasing their response to proliferative stimuli and affecting some functions such as cell migration. HF-SC identity upon Akt activation is preserved; in this sense, increased proliferation does not result in stem cell exhaustion with age suggesting that Akt activation does not affect self-renewal an important aspect for normal tissue maintenance and cancer development. Genome-wide transcriptome analysis of HF-SC isolated from myrAkt and wild-type epidermis underscores changes in metabolic pathways characteristic of cancer cells. These differences manifest during a two-step carcinogenesis protocol in which Akt activation in HF-SCs results in increased tumor development and malignant transformation.
Collapse
Affiliation(s)
- Carmen Segrelles
- Molecular Oncology Unit and, Department of Basic Research, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Interfering with stem cell-specific gatekeeper functions controls tumour initiation and malignant progression of skin tumours. Nat Commun 2015; 6:5874. [PMID: 25608467 PMCID: PMC4354047 DOI: 10.1038/ncomms6874] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/14/2014] [Indexed: 12/17/2022] Open
Abstract
Epithelial cancer constitutes a major clinical challenge and molecular mechanisms underlying the process of tumour initiation are not well understood. Here we demonstrate that hair follicle bulge stem cells (SCs) give rise to well-differentiated sebaceous tumours and show that SCs are not only crucial in tumour initiation, but are also involved in tumour plasticity and heterogeneity. Our findings reveal that SC-specific expression of mutant Lef1, which mimics mutations found in human sebaceous tumours, drives sebaceous tumour formation. Mechanistically, we demonstrate that mutant Lef1 abolishes p53 activity in SCs. Intriguingly, mutant Lef1 induces DNA damage and interferes with SC-specific gatekeeper functions normally protecting against accumulations of DNA lesions and cell loss. Thus, normal control of SC proliferation is disrupted by mutant Lef1, thereby allowing uncontrolled propagation of tumour-initiating SCs. Collectively, these findings identify underlying molecular and cellular mechanisms of tumour-initiating events in tissue SCs providing a potential target for future therapeutic strategies. The presence of multiple stem and progenitor cells in the skin has a major impact on the formation of different epidermal tumours. Here the authors identify bulge stem cells as the cells of origin of sebaceous tumours through genetic lineage tracing and molecular studies in a mouse model.
Collapse
|
108
|
Zhou SB, Wang J, Chiang CA, Sheng LL, Li QF. Mechanical stretch upregulates SDF-1α in skin tissue and induces migration of circulating bone marrow-derived stem cells into the expanded skin. Stem Cells 2015; 31:2703-13. [PMID: 23836581 DOI: 10.1002/stem.1479] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/23/2013] [Accepted: 06/14/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Skin and soft tissue expansion is a procedure that stimulates skin regeneration by applying continuous mechanical stretching of normal donor skin for reconstruction purposes. We have reported that topical transplantation of bone marrow-derived mesenchymal stem cells (MSCs) can accelerate mechanical stretch induced skin regeneration. However, it is unclear how circulating MSCs respond to mechanical stretch in skin tissue. METHODS MSCs from luciferase-Tg Lewis rats were transplanted into a rat tissue expansion model and tracked in vivo by luminescence imaging. Expression levels of chemokines including macrophage inflammatory protein-1α, thymus and activation-regulated chemokine, secondary lymphoid tissue chemokine, cutaneous T-cell attracting chemokine, and stromal-derived factor-1α (SDF-1α) were elevated in mechanically stretched tissues, as were their related chemokine receptors in MSCs. Chemotactic assays were conducted in vitro and in vivo to assess the impact of chemokine expression on MSC migration. RESULTS MSC migration was observed in mechanically stretched skin. Mechanical stretching induced temporal upregulation of chemokine expression. Among all the tested chemokines, SDF-1α showed the most significant increase in stretched skin, suggesting a strong connection to migration of MSCs. The in vitro chemotactic assay showed that conditioned medium from mechanically stretched cells induced MSC migration, which could be blocked with the CXCR4 antagonist AMD3100, as effectively as medium containing 50 ng/ml rat recombinant SDF-1α. Results from in vivo study also showed that MSC migration to mechanically stretched skin was significantly blocked by AMD3100. Moreover, migrating MSCs expressed differentiation markers, suggesting a contribution of MSCs to skin regeneration through differentiation. CONCLUSION Mechanical stretching can upregulate SDF-1α in skin and recruit circulating MSCs through the SDF-1α/CXCR4 pathway.
Collapse
Affiliation(s)
- Shuang-Bai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
109
|
Castellana D, Paus R, Perez-Moreno M. Macrophages contribute to the cyclic activation of adult hair follicle stem cells. PLoS Biol 2014; 12:e1002002. [PMID: 25536657 PMCID: PMC4275176 DOI: 10.1371/journal.pbio.1002002] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022] Open
Abstract
Castellana, Paus, and Perez-Moreno discover that skin resident macrophages signal to skin stem cells via Wnt ligands to activate the hair follicle life cycle. Skin epithelial stem cells operate within a complex signaling milieu that orchestrates their lifetime regenerative properties. The question of whether and how immune cells impact on these stem cells within their niche is not well understood. Here we show that skin-resident macrophages decrease in number because of apoptosis before the onset of epithelial hair follicle stem cell activation during the murine hair cycle. This process is linked to distinct gene expression, including Wnt transcription. Interestingly, by mimicking this event through the selective induction of macrophage apoptosis in early telogen, we identify a novel involvement of macrophages in stem cell activation in vivo. Importantly, the macrophage-specific pharmacological inhibition of Wnt production delays hair follicle growth. Thus, perifollicular macrophages contribute to the activation of skin epithelial stem cells as a novel, additional cue that regulates their regenerative activity. This finding may have translational implications for skin repair, inflammatory skin diseases and cancer. The cyclic life of hair follicles consists of recurring phases of growth, decay, and rest. Previous studies have identified signals that prompt a new phase of hair growth through the activation of resting hair follicle stem cells (HF-SCs). In addition to these signals, recent findings have shown that cues arising from the neighboring skin environment, in which hair follicles dwell, also participate in controlling hair follicle growth. Here we show that skin resident macrophages surround and signal to resting HF-SCs, regulating their entry into a new phase of hair follicle growth. This process involves the death and activation of a fraction of resident macrophages— resulting in Wnt ligand release —that in turn activate HF-SCs. These findings reveal additional mechanisms controlling endogenous stem cell pools that are likely to be relevant for modulating stem cell regenerative capabilities. The results provide new insights that may have implications for the development of technologies with potential applications in regeneration, aging, and cancer.
Collapse
Affiliation(s)
- Donatello Castellana
- Epithelial Cell Biology Group, BBVA Foundation-CNIO Cancer Cell Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ralf Paus
- Institute of Inflammation and Repair, University of Manchester, Manchester, United Kingdom
- Department of Dermatology, University of Münster, Münster, Germany
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group, BBVA Foundation-CNIO Cancer Cell Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- * E-mail:
| |
Collapse
|
110
|
Immune status, strain background, and anatomic site of inoculation affect mouse papillomavirus (MmuPV1) induction of exophytic papillomas or endophytic trichoblastomas. PLoS One 2014; 9:e113582. [PMID: 25474466 PMCID: PMC4256377 DOI: 10.1371/journal.pone.0113582] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/25/2014] [Indexed: 12/14/2022] Open
Abstract
Papillomaviruses (PVs) induce papillomas, premalignant lesions, and carcinomas in a wide variety of species. PVs are classified first based on their host and tissue tropism and then their genomic diversities. A laboratory mouse papillomavirus, MmuPV1 (formerly MusPV), was horizontally transmitted within an inbred colony of NMRI-Foxn1(nu)/Foxn1nu (nude; T cell deficient) mice of an unknown period of time. A ground-up, filtered papilloma inoculum was not capable of infecting C57BL/6J wild-type mice; however, immunocompetent, alopecic, S/RV/Cri-ba/ba (bare) mice developed small papillomas at injection sites that regressed. NMRI-Foxn1(nu) and B6.Cg-Foxn1(nu), but not NU/J-Foxn1(nu), mice were susceptible to MmuPV1 infection. B6 congenic strains, but not other congenic strains carrying the same allelic mutations, lacking B- and T-cells, but not B-cells alone, were susceptible to infection, indicating that mouse strain and T-cell deficiency are critical to tumor formation. Lesions initially observed were exophytic papillomas around the muzzle, exophytic papillomas on the tail, and condylomas of the vaginal lining which could be induced by separate scarification or simultaneous scarification of MmuPV1 at all four sites. On the dorsal skin, locally invasive, poorly differentiated tumors developed with features similar to human trichoblastomas. Transcriptome analysis revealed significant differences between the normal skin in these anatomic sites and in papillomas versus trichoblastomas. The primarily dysregulated genes involved molecular pathways associated with cancer, cellular development, cellular growth and proliferation, cell morphology, and connective tissue development and function. Although trichoepitheliomas are benign, aggressive tumors, few of the genes commonly associated with basal cell carcinoma or squamous cells carcinoma were highly dysregulated.
Collapse
|
111
|
Torkamani N, Rufaut NW, Jones L, Sinclair RD. Beyond goosebumps: does the arrector pili muscle have a role in hair loss? Int J Trichology 2014; 6:88-94. [PMID: 25210331 PMCID: PMC4158628 DOI: 10.4103/0974-7753.139077] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The arrector pili muscle (APM) consists of a small band of smooth muscle that connects the hair follicle to the connective tissue of the basement membrane. The APM mediates thermoregulation by contracting to increase air-trapping, but was thought to be vestigial in humans. The APM attaches proximally to the hair follicle at the bulge, a known stem cell niche. Recent studies have been directed toward this muscle's possible role in maintaining the follicular integrity and stability. This review summarizes APM anatomy and physiology and then discusses the relationship between the follicular unit and the APM. The potential role of the APM in hair loss disorders is also described, and a model explaining APM changes in hair loss is proposed.
Collapse
Affiliation(s)
- Niloufar Torkamani
- University of Melbourne, Melbourne, Victoria, Australia ; Epworth Hospital, Melbourne, Victoria, Australia
| | - Nicholas W Rufaut
- University of Melbourne, Melbourne, Victoria, Australia ; Epworth Hospital, Melbourne, Victoria, Australia
| | - Leslie Jones
- University of Melbourne, Melbourne, Victoria, Australia ; Epworth Hospital, Melbourne, Victoria, Australia
| | - Rodney D Sinclair
- University of Melbourne, Melbourne, Victoria, Australia ; Epworth Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
112
|
Schepeler T, Page ME, Jensen KB. Heterogeneity and plasticity of epidermal stem cells. Development 2014; 141:2559-67. [PMID: 24961797 PMCID: PMC4067958 DOI: 10.1242/dev.104588] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The epidermis is an integral part of our largest organ, the skin, and protects us against the hostile environment. It is a highly dynamic tissue that, during normal steady-state conditions, undergoes constant turnover. Multiple stem cell populations residing in autonomously maintained compartments facilitate this task. In this Review, we discuss stem cell behaviour during normal tissue homeostasis, regeneration and disease within the pilosebaceous unit, an integral structure of the epidermis that is responsible for hair growth and lubrication of the epithelium. We provide an up-to-date view of the pilosebaceous unit, encompassing the heterogeneity and plasticity of multiple discrete stem cell populations that are strongly influenced by external cues to maintain their identity and function.
Collapse
Affiliation(s)
- Troels Schepeler
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen N DK-2200, Denmark
| | - Mahalia E Page
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kim B Jensen
- BRIC - Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen N DK-2200, Denmark Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
113
|
Overexpression of epigen during embryonic development induces reversible, epidermal growth factor receptor-dependent sebaceous gland hyperplasia. Mol Cell Biol 2014; 34:3086-95. [PMID: 24891618 DOI: 10.1128/mcb.00302-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) system is a key regulator of epithelial development and homeostasis. Its functions in the sebaceous gland (SG), however, remain poorly characterized. In this study, using a transgenic mouse line with tissue-specific and inducible expression of the EGFR ligand epigen, we showed that increased activation of the EGFR in skin keratinocytes results in enlarged SGs and increased sebum production. The phenotype can be reverted by interrupting transgene expression and is EGFR dependent, as gland size and sebum levels return to normal values after crossing to the EGFR-impaired mouse line Wa5. Intriguingly, however, the SG enlargement appears only if EGFR activation occurs before birth. Importantly, the enlarged sebaceous glands are associated with an increased expression of the transcription factor MYC and of the transmembrane proteins LRIG1, an established negative-feedback regulator of the EGFR/ERBB tyrosine kinase receptors and a stem cell marker. Our findings identify EGFR signaling as a major pathway determining SG activity and suggest a functional relationship between the EGFR/ERBB system and MYC/LRIG1 in the commitment of stem cells toward specific progenitor cell types, with implications for our understanding of their role in tissue development, homeostasis, and disease.
Collapse
|
114
|
Lee SH, Chung MK, Sohn YJ, Lee YS, Kang KS. Human Hair Follicle Cells with the Cell Surface Marker CD34 Can Regenerate New Mouse Hair Follicles and Located in the Outer Root Sheath of Immunodeficient Nude Mice. Int J Stem Cells 2014; 1:70-81. [PMID: 24855511 DOI: 10.15283/ijsc.2008.1.1.70] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2008] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The bulge region of hair follicle has been reported as a putative reservoir of hair follicle stem cells. The purpose of this study was to compare hair follice CD34 negative (CD34-) cell with CD34 positive (CD34+) cell and to evaluate the ability to regenerate new hair of immunodeficient nude mouse. METHODS AND RESULTS In this report, we isolated the cells with CD34, known as bulge-negative cell surface marker from cultured human hair follicle cells using by magnetic cell sorting (MACS), injected the cells to immunodeficient nude mouse. To determine immunological characterization, human hair follicle CD34+ cells and CD34- cells were assessed by flow cytometry. The localization of injected-CD34+ cells was assessed on formalin-fixed, paraffin-embedded mouse skin samples by in situ hybridization technique. Our findings show that the human hair follicle cells with cell surface marker CD34 were located in the outer root sheath of nude mouse after transplantation and the cells were able to regenerate new hair follicle in immunodeficient nude mouse. CD34- cells also were able to regenerate follicles in the mouse, however, CD34+ cells were able to regenerate much more hair follicle than CD34- cells. CONCLUSIONS Therefore, the results of this study add new insight into the investigation of CD34 stem cell-related molecule in human hair follicles and suggest that not all human hair follicle stem cells reside in bulge region, but in a lager niche.
Collapse
Affiliation(s)
- Sung-Hoon Lee
- Laboratory of Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University ; Adult Stem Cell Research, Seoul, Korea
| | | | | | - Yong-Soon Lee
- Laboratory of Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University ; Adult Stem Cell Research, Seoul, Korea
| | - Kyung-Sun Kang
- Laboratory of Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, and BK21 Program for Veterinary Science, Seoul National University ; Adult Stem Cell Research, Seoul, Korea
| |
Collapse
|
115
|
The acquisition of cancer stem cell-like properties and neoplastic transformation of human keratinocytes induced by arsenite involves epigenetic silencing of let-7c via Ras/NF-κB. Toxicol Lett 2014; 227:91-8. [PMID: 24704393 DOI: 10.1016/j.toxlet.2014.03.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/22/2014] [Accepted: 03/24/2014] [Indexed: 10/25/2022]
Abstract
Exposure of humans to inorganic arsenic can cause skin cancer. The acquisition of cancer stem cell-like properties is involved in the initiation of some cancers, and there are changes in let-7 levels in some tumors. The mechanisms of action, however, remain obscure. Here, we report that there are decreased levels of let-7a, let-7b, and let-7c in human keratinocyte HaCaT cells during malignant transformation induced by a low concentration (1.0μM) of arsenite. The process by which arsenite reduces the level of let-7c apparently involves methylation, for 5-aza-2'-deoxycytidine, an inhibitor of methyltransferases, prevents arsenite-induced hypermethylation, decreases the level of let-7c, and thereby blocks arsenite-induced activation of the Ras/NF-κB signal pathway. Let-7c is an up-stream regulator of the Ras/NF-κB signal pathway and down-regulates activation of this pathway. In arsenite-transformed HaCaT cells, the acquisition of cancer stem cell-like properties is prevented by over-expression of let-7c, and over-expression of let-7c decreases the malignancy of transformed HaCaT cells. Thus, we conclude that epigenetic silencing of let-7c via Ras/NF-κB is involved in the acquisition of cancer stem cell-like properties and neoplastic transformation of HaCaT cells induced by arsenite, which contribute to the tumorigenesis of arsenite.
Collapse
|
116
|
Abstract
The melanocyte stem cells of the hair follicle provide an attractive system for the study of the stem cells. Successful regeneration of a functional organ relies on the organized and timely orchestration of molecular events among distinct stem/progenitor cell populations. The stem cells are regulated by communication with their specialized microenvironment known as the niche. Despite remarkable progress in understanding stem cell-intrinsic behavior, the molecular nature of the extrinsic factors provided to the stem cells by the niche microenvironment remains poorly understood. In this regard, the bulge niche of the mammalian hair follicle offers an excellent model for study. It holds two resident populations of SCs: epidermal stem cells and melanocyte stem cells. While their behavior is tightly coordinated, very little of the crosstalk involved is known. This review summarized the recent development in trying to understand the regulation of melanocyte and melanocyte stem cells. A better understanding of the normal regulation and behaviors of the melanocytes and the melanocyte stem cells will help to improve the clinical applications in regenerative medicine, cancer therapy, and aging.
Collapse
Affiliation(s)
- Ang Li
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, PO 300, New York, NY 10065, USA
| |
Collapse
|
117
|
Purba TS, Haslam IS, Poblet E, Jiménez F, Gandarillas A, Izeta A, Paus R. Human epithelial hair follicle stem cells and their progeny: current state of knowledge, the widening gap in translational research and future challenges. Bioessays 2014; 36:513-25. [PMID: 24665045 DOI: 10.1002/bies.201300166] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epithelial hair follicle stem cells (eHFSCs) are required to generate, maintain and renew the continuously cycling hair follicle (HF), supply cells that produce the keratinized hair shaft and aid in the reepithelialization of injured skin. Therefore, their study is biologically and clinically important, from alopecia to carcinogenesis and regenerative medicine. However, human eHFSCs remain ill defined compared to their murine counterparts, and it is unclear which murine eHFSC markers really apply to the human HF. We address this by reviewing current concepts on human eHFSC biology, their immediate progeny and their molecular markers, focusing on Keratin 15 and 19, CD200, CD34, PHLDA1, and EpCAM/Ber-EP4. After delineating how human eHFSCs may be selectively targeted experimentally, we close by defining as yet unmet key challenges in human eHFSC research. The ultimate goal is to transfer emerging concepts from murine epithelial stem cell biology to human HF physiology and pathology.
Collapse
Affiliation(s)
- Talveen S Purba
- The Dermatology Centre, Salford Royal NHS Foundation Trust and Institute of Inflammation and Repair, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
118
|
Alternative lengthening of telomeres in cancer stem cells in vivo. Oncogene 2014; 34:611-20. [PMID: 24531712 PMCID: PMC4135038 DOI: 10.1038/onc.2013.603] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 11/01/2013] [Accepted: 12/18/2013] [Indexed: 12/17/2022]
Abstract
Chromosome ends are protected by telomeres which prevent DNA damage response and degradation. Telomerase expression extends telomeres and inhibits DNA damage response. Telomeres are also maintained by the recombination based alternative lengthening pathway. Telomerase is believed to be the sole mechanism for telomere maintenance in epidermis. We show that basal cells in epidermis maintain telomeres both by telomerase and ALT mechanisms in vivo. ALT was detected in epidermal stem cells in Terc−/− mice, and normal human epidermal keratinocytes are also ALT positive. ALT pathway is suppressed in primary but not metastatic epidermal squamous cell carcinomas (SCC) in Terc+/+ mice. ALT pathway is expressed in stem and basal cells in epidermal SCC in Terc−/− mice, and some telomerase positive human SCC lines. Telomeres shorten dramatically in stem and basal cells in epidermal SCC in vivo. Telomere shortening is associated with telomeric DNA damage response and apoptosis in stem and basal cells. Stem cells were transformed in both primary and metastatic epidermal SCC. Genetic ablation of this small cell population resulted in significant tumor regression in vivo. We concluded that alternative lengthening of telomeres is important in epidermal homeostasis and tumorigenesis in vivo.
Collapse
|
119
|
Shen Y, Dai L, Li X, Liang R, Guan G, Zhang Z, Cao W, Liu Z, Mei S, Liang W, Qin S, Xu J, Chen H. Epidermal stem cells cultured on collagen-modified chitin membrane induce in situ tissue regeneration of full-thickness skin defects in mice. PLoS One 2014; 9:e87557. [PMID: 24516553 PMCID: PMC3917838 DOI: 10.1371/journal.pone.0087557] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 12/22/2013] [Indexed: 12/17/2022] Open
Abstract
A Large scale of full-thickness skin defects is lack of auto-grafts and which requires the engineered skin substitutes for repair and regeneration. One major obstacle in skin tissue engineering is to expand epidermal stem cells (ESCs) and develop functional substitutes. The other one is the scaffold of the ESCs. Here, we applied type I collagen-modified chitin membrane to form collagen-chitin biomimetic membrane (C-CBM), which has been proved to have a great biocompatibility and degraded totally when it was subcutaneously transplanted into rat skin. ESCs were cultured, and the resulting biofilm was used to cover full-thickness skin defects in nude mice. The transplantation of ESCs- collagen- chitn biomimetic membrane (ESCs-C-CBM) has achieved in situ skin regeneration. In nude mice, compared to controls with collagen-chitin biomimetic membrane (C-CBM) only, the ESCs-C-CBM group had significantly more dermatoglyphs on the skin wound 10 w after surgery, and the new skin was relatively thick, red and elastic. In vivo experiments showed obvious hair follicle cell proliferation in the full-thickness skin defect. Stem cell markers examination showed active ESCs in repair and regeneration of skin. The results indicate that the collagen-modified chitin membrane carry with ESCs has successfully regenerated the whole skin with all the skin appendages and function.
Collapse
Affiliation(s)
- Yan Shen
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Libing Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Xiaojian Li
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
- * E-mail:
| | - Rong Liang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Guangxiong Guan
- Department of Medical Laboratory, Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, People’s Republic of China
| | - Zhi Zhang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Wenjuan Cao
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Shirley Mei
- Student of Sophie Davis School of Biomedical Education, Mack Lipkin Fellowship, New York, New York, United States of America
| | - Weiguo Liang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Shennan Qin
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Honghui Chen
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital Medical College, Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
120
|
Han B, Chen SY, Zhu YT, Tseng SCG. Integration of BMP/Wnt signaling to control clonal growth of limbal epithelial progenitor cells by niche cells. Stem Cell Res 2014; 12:562-73. [PMID: 24530980 DOI: 10.1016/j.scr.2014.01.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/07/2014] [Accepted: 01/08/2014] [Indexed: 12/13/2022] Open
Abstract
Both BMP and Wnt signaling control stem cells in bulge/dermal papilla, intestinal crypt, and bone marrow. To explore their roles in the limbal niche, which govern corneal epithelial homeostasis, we established an in vitro model of sphere growth by reunion between single limbal epithelial progenitor cells (LEPCs) and aggregates of limbal niche cells (LNCs) in 3D Matrigel. Compared to LEPCs alone, spheres formed by LEPC+LNC exhibited higher clonal growth and less corneal epithelial differentiation. Furthermore, pSmad1/5/8 was in the nucleus of LEPCs, but not LNCs, and correlated with upregulation of BMP1, BMP3, BMP4, all three BMP receptors, and BMP target genes. Inactivation of BMP signaling in LNCs was correlated with upregulation of noggin preferentially expressed by LNCs. Additionally, β-catenin was stabilized in the perinuclear cytoplasm in LEPCs and correlated with upregulation of Wnt7A and FZD5 preferentially expressed by LEPCs. Inactivation of Wnt signaling in LNCs was correlated with upregulation of DKK1/2 by LNCs. Addition of XAV939 that expectedly downregulated perinuclear β-catenin in LEPCs led to significant reduction of epithelial clonal growth, but upregulated all three BMP receptors and downregulated LNC-derived noggin, resulting in activation of BMP signaling in LNCs. Addition of noggin that expectedly downregulated nuclear localization of pSmad1/5/8 in LEPCs led to nuclear localization of β-catenin in larger LEPCs but membrane relocation of β-catenin in smaller LEPCs and significant upregulation of DKK1/2. Hence, balancing acts between Wnt signaling and BMP signaling exist not only within LEPCs but also between LEPCs and LNCs to regulate clonal growth of LEPCs.
Collapse
Affiliation(s)
- Bo Han
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Ocular Surface Research & Education Foundation, Miami, FL, USA
| | - Szu-Yu Chen
- R&D Department, TissueTech, Inc., Miami, FL, USA
| | | | - Scheffer C G Tseng
- R&D Department, TissueTech, Inc., Miami, FL, USA; Ocular Surface Center, Miami, FL, USA; Ocular Surface Research & Education Foundation, Miami, FL, USA.
| |
Collapse
|
121
|
Rompolas P, Greco V. Stem cell dynamics in the hair follicle niche. Semin Cell Dev Biol 2013; 25-26:34-42. [PMID: 24361866 DOI: 10.1016/j.semcdb.2013.12.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 11/27/2013] [Accepted: 12/11/2013] [Indexed: 12/17/2022]
Abstract
Hair follicles are appendages of the mammalian skin that have the ability to periodically and stereotypically regenerate in order to continuously produce new hair over our lifetime. The ability of the hair follicle to regenerate is due to the presence of stem cells that along with other cell populations and non-cellular components, including molecular signals and extracellular material, make up a niche microenvironment. Mounting evidence suggests that the niche is critical for regulating stem cell behavior and thus the process of regeneration. Here, we review the literature concerning past and current studies that have utilized mouse genetic models, combined with other approaches to dissect the molecular and cellular composition of the hair follicle niche. We also discuss our current understanding of how stem cells operate within the niche during the process of tissue regeneration and the factors that regulate their behavior.
Collapse
Affiliation(s)
- Panteleimon Rompolas
- Department of Genetics, Department of Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Valentina Greco
- Department of Genetics, Department of Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
122
|
Tai G, Ranjzad P, Marriage F, Rehman S, Denley H, Dixon J, Mitchell K, Day PJR, Woolf AS. Cytokeratin 15 marks basal epithelia in developing ureters and is upregulated in a subset of urothelial cell carcinomas. PLoS One 2013; 8:e81167. [PMID: 24260555 PMCID: PMC3832456 DOI: 10.1371/journal.pone.0081167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 10/09/2013] [Indexed: 11/29/2022] Open
Abstract
The mammalian ureter contains a water-tight epithelium surrounded by smooth muscle. Key molecules have been defined which regulate ureteric bud initiation and drive the differentiation of ureteric mesenchyme into peristaltic smooth muscle. Less is known about mechanisms underlying the developmental patterning of the multilayered epithelium characterising the mature ureter. In skin, which also contains a multilayered epithelium, cytokeratin 15 (CK15), an acidic intermediate filament protein, marks cells whose progeny contribute to epidermal regeneration following wounding. Moreover, CK15+ precursor cells in skin can give rise to basal cell carcinomas. In the current study, using transcriptome microarrays of embryonic wild type mouse ureters, Krt15, coding for CK15, was detected. Quantitative polymerase chain reaction analyses confirmed the initial finding and demonstrated that Krt15 levels increased during the fetal period when the ureteric epithelium becomes multilayered. CK15 protein was undetectable in the ureteric bud, the rudiment from which the ureter grows. Nevertheless, later in fetal development, CK15 was immunodetected in a subset of basal urothelial cells in the ureteric stalk. Superficial epithelial cells, including those positive for the differentiation marker uroplakin III, were CK15-. Transformation-related protein 63 (P63) has been implicated in epithelial differentiation in murine fetal urinary bladders. In wild type fetal ureters, CK15+ cells were positive for P63, and p63 homozygous null mutant ureters lacked CK15+ cells. In these mutant ureters, sections of the urothelium were monolayered versus the uniform multilayering found in wild type littermates. Human urothelial cell carcinomas account for considerable morbidity and mortality. CK15 was upregulated in a subset of invasive ureteric and urinary bladder cancers. Thus, in ureter development, the absence of CK15 is associated with a structurally simplified urothelium whereas, postnatally, increased CK15 levels feature in malignant urothelial overgrowth. CK15 may be a novel marker for urinary tract epithelial precursor cells.
Collapse
Affiliation(s)
- Guangping Tai
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom ; Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
Mesenchymal cells play a role in controlling the number of hair follicles. However, the precise molecules involved are unclear. Absence in mesenchymal cells of the expression of the secreted matricellular protein CTGF/CCN2 results in an increased number of hair follicles, concomitant with increased β-catenin activity. It is critical to understand how stem cell activity is regulated during regeneration. Hair follicles constitute an important model for organ regeneration because, throughout adult life, they undergo cyclical regeneration. Hair follicle stem cells—epithelial cells located in the follicle bulge—are activated by periodic β-catenin activity, which is regulated not only by epithelial-derived Wnt, but also, through as-yet-undefined mechanisms, the surrounding dermal microenvironment. The matricellular protein connective tissue growth factor (CCN2) is secreted into the microenvironment and acts as a multifunctional signaling modifier. In adult skin, CCN2 is largely absent but is unexpectedly restricted to the dermal papillae and outer root sheath. Deletion of CCN2 in dermal papillae and the outer root sheath results in a shortened telogen-phase length and elevated number of hair follicles. Recombinant CCN2 causes decreased β-catenin stability in keratinocytes. In vivo, loss of CCN2 results in elevated numbers of K15-positive epidermal stem cells that possess elevated β-catenin levels and β-catenin–dependent reporter gene expression. These results indicate that CCN2 expression by dermal papillae cells is a physiologically relevant suppressor of hair follicle formation by destabilization of β-catenin and suggest that CCN2 normally acts to maintain stem cell quiescence.
Collapse
Affiliation(s)
- Shangxi Liu
- Departments of Dentistry and Physiology and Pharmacology, University of Western Ontario, London, ON N6A 5C1, Canada
| | | |
Collapse
|
124
|
Genetic correction of stem cells in the treatment of inherited diseases and focus on xeroderma pigmentosum. Int J Mol Sci 2013; 14:20019-36. [PMID: 24113582 PMCID: PMC3821600 DOI: 10.3390/ijms141020019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/11/2013] [Accepted: 09/17/2013] [Indexed: 02/05/2023] Open
Abstract
Somatic stem cells ensure tissue renewal along life and healing of injuries. Their safe isolation, genetic manipulation ex vivo and reinfusion in patients suffering from life threatening immune deficiencies (for example, severe combined immunodeficiency (SCID)) have demonstrated the efficacy of ex vivo gene therapy. Similarly, adult epidermal stem cells have the capacity to renew epidermis, the fully differentiated, protective envelope of our body. Stable skin replacement of severely burned patients have proven life saving. Xeroderma pigmentosum (XP) is a devastating disease due to severe defects in the repair of mutagenic DNA lesions introduced upon exposure to solar radiations. Most patients die from the consequences of budding hundreds of skin cancers in the absence of photoprotection. We have developed a safe procedure of genetic correction of epidermal stem cells isolated from XP patients. Preclinical and safety assessments indicate successful correction of XP epidermal stem cells in the long term and their capacity to regenerate a normal skin with full capacities of DNA repair.
Collapse
|
125
|
Spatial organization within a niche as a determinant of stem-cell fate. Nature 2013; 502:513-8. [PMID: 24097351 PMCID: PMC3895444 DOI: 10.1038/nature12602] [Citation(s) in RCA: 312] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/23/2013] [Indexed: 12/17/2022]
Abstract
Stem cell niches in mammalian tissues are often heterogeneous and compartmentalized, however whether distinct niche locations determine different stem cell fates remains unclear. To test this hypothesis, we utilized the mouse hair follicle niche and devised a novel approach by combining intravital microscopy with genetic lineage tracing to re-visit the same stem cell lineages, from their exact place of origin, throughout regeneration in live mice. Using this method, we show directly that the position of a stem cell within the hair follicle niche can predict whether it is likely to remain uncommitted, generate precursors or commit to a differentiated fate. Furthermore, using laser ablation we demonstrate that hair follicle stem cells are dispensable for regeneration and that epithelial cells, which do not normally participate in hair growth, re-populate the lost stem cell compartment and sustain hair regeneration. This study provides a general paradigm for niche-induced fate determination in adult tissues.
Collapse
|
126
|
Solanas G, Benitah SA. Regenerating the skin: a task for the heterogeneous stem cell pool and surrounding niche. Nat Rev Mol Cell Biol 2013; 14:737-48. [DOI: 10.1038/nrm3675] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
127
|
Bose A, Teh MT, Mackenzie IC, Waseem A. Keratin k15 as a biomarker of epidermal stem cells. Int J Mol Sci 2013; 14:19385-98. [PMID: 24071939 PMCID: PMC3821562 DOI: 10.3390/ijms141019385] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/05/2013] [Accepted: 09/10/2013] [Indexed: 01/21/2023] Open
Abstract
Keratin 15 (K15) is type I keratin protein co-expressed with the K5/K14 pair present in the basal keratinocytes of all stratified epithelia. Although it is a minor component of the cytoskeleton with a variable expression pattern, nonetheless its expression has been reported as a stem cell marker in the bulge of hair follicles. Conversely, suprabasal expression of K15 has also been reported in both normal and diseased tissues, which is inconsistent with its role as a stem cell marker. Our recently published work has given evidence of the molecular pathways that seem to control the expression of K15 in undifferentiated and differentiated cells. In this article, we have critically reviewed the published work to establish the reliability of K15 as an epidermal stem cell marker.
Collapse
Affiliation(s)
- Amrita Bose
- Centre for Clinical and Diagnostic Oral Sciences, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Turner Street, London E1 2AD, UK.
| | | | | | | |
Collapse
|
128
|
Rho O, Kiguchi K, Jiang G, DiGiovanni J. Impact of mTORC1 inhibition on keratinocyte proliferation during skin tumor promotion in wild-type and BK5.AktWT mice. Mol Carcinog 2013; 53:871-82. [PMID: 24114993 DOI: 10.1002/mc.22046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/03/2013] [Accepted: 04/17/2013] [Indexed: 01/22/2023]
Abstract
In this study, we examined the impact of rapamycin on mTORC1 signaling during 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced keratinocyte proliferation and skin tumor promotion in both wild-type (FVB/N) and BK5.Akt(WT) mice. TPA activated mTORC1 signaling in a time-dependent manner in cultured primary mouse keratinocytes and a mouse keratinocyte cell line. Early activation (15-30 min) of mTORC1 signaling induced by TPA was mediated in part by PKC activation, whereas later activation (2-4 h) was mediated by activation of EGFR and Akt. BK5.Akt(WT) transgenic mice, where Akt1 is overexpressed in basal epidermis, are highly sensitive to TPA-induced epidermal proliferation and two-stage skin carcinogenesis. Targeting mTORC1 with rapamycin effectively inhibited TPA-induced epidermal hyperplasia and hyperproliferation as well as tumor promotion in a dose-dependent manner in both wild-type and BK5.Akt(WT) mice. A significant expansion (∼threefold) of the label retaining cell (LRC) population per hair follicle was observed in BK5.Akt(WT) mice compared to FVB/N mice. There was also a significant increase in K15 expressing cells in the hair follicle of transgenic mice that coincided with expression of phospho-Akt, phospho-S6K, and phospho-PRAS40, suggesting an important role of mTORC1 signaling in bulge-region keratinocyte stem cell (KSC) homeostasis. After 2 weeks of TPA treatment, LRCs had moved upward into the interfollicular epidermis from the bulge region of both wild-type and BK5.Akt(WT) mice. TPA-mediated LRC proliferation and migration was significantly inhibited by rapamycin. Collectively, the current data indicate that signaling through mTORC1 contributes significantly to the process of skin tumor promotion through effects on proliferation of the target cells for tumor development.
Collapse
Affiliation(s)
- Okkyung Rho
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, Austin, Texas
| | | | | | | |
Collapse
|
129
|
White RA, Neiman JM, Reddi A, Han G, Birlea S, Mitra D, Dionne L, Fernandez P, Murao K, Bian L, Keysar SB, Goldstein NB, Song N, Bornstein S, Han Z, Lu X, Wisell J, Li F, Song J, Lu SL, Jimeno A, Roop DR, Wang XJ. Epithelial stem cell mutations that promote squamous cell carcinoma metastasis. J Clin Invest 2013; 123:4390-404. [PMID: 23999427 DOI: 10.1172/jci65856] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 07/11/2013] [Indexed: 12/15/2022] Open
Abstract
Squamous cell carcinomas (SCCs) originate in stratified epithelia, with a small subset becoming metastatic. Epithelial stem cells are targets for driver mutations that give rise to SCCs, but it is unknown whether they contribute to oncogenic multipotency and metastasis. We developed a mouse model of SCC by targeting two frequent genetic mutations in human SCCs, oncogene Kras(G12D) activation and Smad4 deletion, to mouse keratin 15-expressing (K15+) stem cells. We show that transgenic mice developed multilineage tumors, including metastatic SCCs. Among cancer stem cell-enriched (CSC-enriched) populations, those with increased side population (SP) cells correlated with epithelial-mesenchymal transition (EMT) and lung metastasis. We show that microRNA-9 (miR-9) contributed to SP expansion and metastasis, and miR-9 inhibition reduced the number of SP cells and metastasis. Increased miR-9 was detected in metastatic human primary SCCs and SCC metastases, and miR-9-transduced human SCC cells exhibited increased invasion. We identified α-catenin as a predominant miR-9 target. Increased miR-9 in human SCC metastases correlated with α-catenin loss but not E-cadherin loss. Our results demonstrate that stem cells with Kras(G12D) activation and Smad4 depletion can produce tumors that are multipotent and susceptible to EMT and metastasis. Additionally, tumor initiation and metastatic properties of CSCs can be uncoupled, with miR-9 regulating the expansion of metastatic CSCs.
Collapse
|
130
|
Jones KB, Klein OD. Oral epithelial stem cells in tissue maintenance and disease: the first steps in a long journey. Int J Oral Sci 2013; 5:121-9. [PMID: 23887128 PMCID: PMC3967329 DOI: 10.1038/ijos.2013.46] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 06/08/2013] [Indexed: 12/11/2022] Open
Abstract
The identification and characterization of stem cells is a major focus of developmental biology and regenerative medicine. The advent of genetic inducible fate mapping techniques has made it possible to precisely label specific cell populations and to follow their progeny over time. When combined with advanced mathematical and statistical methods, stem cell division dynamics can be studied in new and exciting ways. Despite advances in a number of tissues, relatively little attention has been paid to stem cells in the oral epithelium. This review will focus on current knowledge about adult oral epithelial stem cells, paradigms in other epithelial stem cell systems that could facilitate new discoveries in this area and the potential roles of epithelial stem cells in oral disease.
Collapse
Affiliation(s)
- Kyle B Jones
- Program in Craniofacial and Mesenchymal Biology, University of California, San Francisco, San Francisco, USA
| | | |
Collapse
|
131
|
Hayes CS, DeFeo-Mattox K, Woster PM, Gilmour SK. Elevated ornithine decarboxylase activity promotes skin tumorigenesis by stimulating the recruitment of bulge stem cells but not via toxic polyamine catabolic metabolites. Amino Acids 2013; 46:543-52. [PMID: 23884694 DOI: 10.1007/s00726-013-1559-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/03/2013] [Indexed: 12/17/2022]
Abstract
Elevated expression of ornithine decarboxylase (ODC), the regulatory enzyme in polyamine biosynthesis, targeted to the epidermis is sufficient to promote skin tumor development following a single subthreshold dose of dimethylbenz(a)anthracene (DMBA). Since skin tumor promotion involves recruitment of hair follicle bulge stem cells harboring genetic lesions, we assessed the effect of increased epidermal ODC on recruitment of bulge stem cells in ODC-ER transgenic mice in which ODC activity is induced de novo in adult skin with 4-hydroxytamoxifen (4OHT). Bromodeoxyuridine-pulse labeling and use of K15.CrePR1;R26R;ODC-ER triple transgenic mice demonstrated that induction of ODC activity is sufficient to recruit bulge stem cells in quiescent skin. Because increased ODC activity not only stimulates proliferation but also increases reactive oxygen species (ROS) generation via subsequent induction of polyamine catabolic oxidases, we used an inhibitor of polyamine catabolic oxidase activity, MDL72527, to investigate whether ROS generation by polyamine catabolic oxidases contributes to skin tumorigenesis in DMBA-initiated ODC-ER transgenic skin. Newborn ODC-ER transgenic mice and their normal littermates were initiated with a single topical dose of DMBA. To assess tumor development originating from dormant bulge stem cells that possess DMBA-initiated mutations, epidermal ODC activity was induced in ODC-ER mice with 4OHT 5 weeks after DMBA initiation followed by MDL72527 treatment. MDL72527 treatment resulted in a shorter tumor latency time, increased tumor burden, increased conversion to carcinomas, and lower tumor levels of p53. Thus, elevated epidermal ODC activity promotes tumorigenesis by stimulating the recruitment of bulge stem cells but not via ROS generation by polyamine catabolic oxidases.
Collapse
Affiliation(s)
- Candace S Hayes
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA
| | | | | | | |
Collapse
|
132
|
Brohem CA, de Carvalho CM, Radoski CL, Santi FC, Baptista MC, Swinka BB, de A. Urban C, de Araujo LRR, Graf RM, Feferman IHS, Lorencini M. Comparison between fibroblasts and mesenchymal stem cells derived from dermal and adipose tissue. Int J Cosmet Sci 2013; 35:448-57. [DOI: 10.1111/ics.12064] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/28/2013] [Indexed: 12/11/2022]
Affiliation(s)
- C. A. Brohem
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | | | - C. L. Radoski
- Department of Biotechnology; Positivo University; Curitiba; Paraná; Brazil
| | - F. C. Santi
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - M. C. Baptista
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - B. B. Swinka
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - C. de A. Urban
- Department of Biotechnology; Positivo University; Curitiba; Paraná; Brazil
| | | | - R. M. Graf
- Department of Plastic Surgery; Federal University of Paraná; Curitiba; Paraná; Brazil
| | - I. H. S. Feferman
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| | - M. Lorencini
- Department of Research and Development; Grupo Boticário; Biomolecular Research Laboratory; São José dos Pinhais; Paraná; Brazil
| |
Collapse
|
133
|
Schneider RP, Garrobo I, Foronda M, Palacios JA, Marión RM, Flores I, Ortega S, Blasco MA. TRF1 is a stem cell marker and is essential for the generation of induced pluripotent stem cells. Nat Commun 2013; 4:1946. [DOI: 10.1038/ncomms2946] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 04/30/2013] [Indexed: 12/14/2022] Open
|
134
|
Michael S, Lambert PF, Strati K. The HPV16 oncogenes cause aberrant stem cell mobilization. Virology 2013; 443:218-25. [PMID: 23664148 DOI: 10.1016/j.virol.2013.04.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 03/13/2013] [Accepted: 04/11/2013] [Indexed: 12/17/2022]
Abstract
Human Papilloma Virus related epithelial cancers have been speculated to derive from virus-infected tissue stem cells. Stem cells also are thought to provide a reservoir of latently infected cells that can persist for long periods. In this study we have examined the effects of HPV16 E6 and E7 oncogenes on multipotent epithelial stem cells, using in vivo systems. Our results show that expression of HPV16 oncogenes reduces the number of bulge label-retaining cells within hair follicles at telogen suggesting aberrant mobilization, a result supported by increased mobilization upon acute anagen induction. Importantly the loss of relative quiescence, a hallmark feature of stem cells, occurs in the absence of a reduction in other stem cell markers. This points to an atypical stem cell compartment in the context of E6 and E7 expression. We hypothesize that this aberrant compartment may have important roles in the viral life cycle and/or ensuing carcinogenesis.
Collapse
Affiliation(s)
- Stella Michael
- Department of Biological Sciences, University of Cyprus, 1 University Ave., Nicosia 2109, Cyprus
| | | | | |
Collapse
|
135
|
Wong CE, Yu JS, Quigley DA, To MD, Jen KY, Huang PY, Del Rosario R, Balmain A. Inflammation and Hras signaling control epithelial-mesenchymal transition during skin tumor progression. Genes Dev 2013; 27:670-82. [PMID: 23512660 PMCID: PMC3613613 DOI: 10.1101/gad.210427.112] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/22/2013] [Indexed: 12/19/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is thought to be an important, possibly essential, component of the process of tumor dissemination and metastasis. About 20%-30% of Hras mutant mouse skin carcinomas induced by chemical initiation/promotion protocols have undergone EMT. Reduced exposure to TPA-induced chronic inflammation causes a dramatic reduction in classical papillomas and squamous cell carcinomas (SCCs), but the mice still develop highly invasive carcinomas with EMT properties, reduced levels of Hras and Egfr signaling, and frequent Ink4/Arf deletions. Deletion of Hras from the mouse germline also leads to a strong reduction in squamous tumor development, but tumors now acquire activating Kras mutations and exhibit more aggressive metastatic properties. We propose that invasive carcinomas can arise by different genetic and biological routes dependent on exposure to chronic inflammation and possibly from different target cell populations within the skin. Our data have implications for the use of inhibitors of inflammation or of Ras/Egfr pathway signaling for prevention or treatment of invasive cancers.
Collapse
Affiliation(s)
- Christine E. Wong
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Jennifer S. Yu
- Department of Radiation Oncology
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - David A. Quigley
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Minh D. To
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Kuang-Yu Jen
- Department of Pathology, University of California at San Francisco, San Francisco, California 94143, USA
| | - Phillips Y. Huang
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Reyno Del Rosario
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
136
|
Takeda N, Jain R, Leboeuf MR, Padmanabhan A, Wang Q, Li L, Lu MM, Millar SE, Epstein JA. Hopx expression defines a subset of multipotent hair follicle stem cells and a progenitor population primed to give rise to K6+ niche cells. Development 2013; 140:1655-64. [PMID: 23487314 DOI: 10.1242/dev.093005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The mammalian hair follicle relies on adult resident stem cells and their progeny to fuel and maintain hair growth throughout the life of an organism. The cyclical and initially synchronous nature of hair growth makes the hair follicle an ideal system with which to define homeostatic mechanisms of an adult stem cell population. Recently, we demonstrated that Hopx is a specific marker of intestinal stem cells. Here, we show that Hopx specifically labels long-lived hair follicle stem cells residing in the telogen basal bulge. Hopx(+) cells contribute to all lineages of the mature hair follicle and to the interfollicular epidermis upon epidermal wounding. Unexpectedly, our analysis identifies a previously unappreciated progenitor population that resides in the lower hair bulb of anagen-phase follicles and expresses Hopx. These cells co-express Lgr5, do not express Shh and escape catagen-induced apoptosis. They ultimately differentiate into the cytokeratin 6-positive (K6) inner bulge cells in telogen, which regulate the quiescence of adjacent hair follicle stem cells. Although previous studies have suggested that K6(+) cells arise from Lgr5-expressing lower outer root sheath cells in anagen, our studies indicate an alternative origin, and a novel role for Hopx-expressing lower hair bulb progenitor cells in contributing to stem cell homeostasis.
Collapse
Affiliation(s)
- Norifumi Takeda
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Yang Y, Li Y, Wang K, Wang Y, Yin W, Li L. P38/NF-κB/snail pathway is involved in caffeic acid-induced inhibition of cancer stem cells-like properties and migratory capacity in malignant human keratinocyte. PLoS One 2013; 8:e58915. [PMID: 23516577 PMCID: PMC3596354 DOI: 10.1371/journal.pone.0058915] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 02/08/2013] [Indexed: 12/28/2022] Open
Abstract
Background Skin cancer is the most common cancer throughout the world. The epithelial-mesenchymal transition (EMT) and the acquisition of cancer stem cells (CSCs)-like properties emerge as critical steps in the metastasis of human skin cancers. Caffeic acid (CaA) exerts anticarcinogenic effects. However, the effects of CaA on the migratory capability and on the CSCs-like properties of skin cancer cells, and the molecular mechanisms underlying it are not fully understood. Methods Malignant HaCaT cells were treated by CaA. Transwell assay was performed to determine that CaA attenuated the migratory capability; Spheroid formation assay was performed to confirm that CaA decreased the CSCs-like phenotype; Treated malignant HaCaT cells were molecularly characterized by RT-PCR, Western blots, Southwestern blot, and immunoprecipitation. Results In CaA-treated malignant human keratinocyte (malignant HaCaT cells), inhibition of the migratory capability and CSCs-like phenotype were observed. CaA up-regulated the phosphorylation of p38, and down-regulated the activation of nuclear factor κB (NF-κB)/snail signal pathway. Indeed, p38 decreased the DNA-binding activity of NF-κB to the promoter of snail gene, which resulted in the transcriptional inactivation of snail. Blockage of p38 attenuated the CaA-induced inhibition of migratory capability and CSCs-like phenotype in malignant HaCaT cells. Conclusions CaA attenuates the migratory capability and CSCs-like Properties of malignant human keratinocyte, in which, p38-mediated down-regulation of NF-κB/snail signal pathway is involved.
Collapse
Affiliation(s)
- Ye Yang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yuan Li
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Kebo Wang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yu Wang
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Wenqin Yin
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
| | - Lei Li
- Department of Hygiene Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing, People’s Republic of China
- The Key Laboratory of Modern Toxicology, Ministry of Education, Nanjing Medical University, Nanjing, People’s Republic of China
- * E-mail:
| |
Collapse
|
138
|
Competitive balance of intrabulge BMP/Wnt signaling reveals a robust gene network ruling stem cell homeostasis and cyclic activation. Proc Natl Acad Sci U S A 2013; 110:1351-6. [PMID: 23292934 DOI: 10.1073/pnas.1121312110] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hair follicles facilitate the study of stem cell behavior because stem cells in progressive activation stages, ordered within the follicle architecture, are capable of cyclic regeneration. To study the gene network governing the homeostasis of hair bulge stem cells, we developed a Keratin 15-driven genetic model to directly perturb molecular signaling in the stem cells. We visualize the behavior of these modified stem cells, evaluating their hair-regenerating ability and profile their molecular expression. Bone morphogenetic protein (BMP)-inactivated stem cells exhibit molecular profiles resembling those of hair germs, yet still possess multipotentiality in vivo. These cells also exhibit up-regulation of Wnt7a, Wnt7b, and Wnt16 ligands and Frizzled (Fzd) 10 receptor. We demonstrate direct transcriptional modulation of the Wnt7a promoter. These results highlight a previously unknown intra-stem cell antagonistic competition, between BMP and Wnt signaling, to balance stem cell activity. Reduced BMP signaling and increased Wnt signaling tilts each stem cell toward a hair germ fate and, vice versa, based on a continuous scale dependent on the ratio of BMP/Wnt activity. This work reveals one more hierarchical layer regulating stem cell homeostasis beneath the stem cell-dermal papilla-based epithelial-mesenchymal interaction layer and the hair follicle-intradermal adipocyte-based tissue interaction layer. Although hierarchical layers are all based on BMP/Wnt signaling, the multilayered control ensures that all information is taken into consideration and allows hair stem cells to sum up the total activators/inhibitors involved in making the decision of activation.
Collapse
|
139
|
Nakrieko KA, Irvine TS, Dagnino L. Isolation of hair follicle bulge stem cells from YFP-expressing reporter mice. Methods Mol Biol 2013; 989:21-32. [PMID: 23483384 DOI: 10.1007/978-1-62703-330-5_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
In this article we provide a method to isolate hair follicle stem cells that have undergone targeted gene inactivation. The mice from which these cells are isolated are bred into a Rosa26-yellow fluorescent protein (YFP) reporter background, which results in YFP expression in the targeted stem cell population. These cells are isolated and purified by fluorescence-activated cell sorting, using epidermal stem cell-specific markers in conjunction with YFP fluorescence. The purified cells can be used for gene expression studies, clonogenic experiments, and biological assays, such as viability and capacity for directional migration.
Collapse
Affiliation(s)
- Kerry-Ann Nakrieko
- Department of Physiology and Pharmacology, Child Health Research Institute, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
140
|
Antonini D, Sibilio A, Dentice M, Missero C. An Intimate Relationship between Thyroid Hormone and Skin: Regulation of Gene Expression. Front Endocrinol (Lausanne) 2013; 4:104. [PMID: 23986743 PMCID: PMC3749490 DOI: 10.3389/fendo.2013.00104] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/05/2013] [Indexed: 12/23/2022] Open
Abstract
Skin is the largest organ of the human body and plays a key role in protecting the individual from external insults. The barrier function of the skin is performed primarily by the epidermis, a self-renewing stratified squamous epithelium composed of cells that undergo a well-characterized and finely tuned process of terminal differentiation. By binding to their receptors thyroid hormones (TH) regulate epidermal cell proliferation, differentiation, and homeostasis. Thyroid dysfunction has multiple classical manifestations at skin level. Several TH-responsive genes, as well as genes critical for TH metabolism and action, are expressed at epidermal level. The role of TH in skin is still controversial, although it is generally recognized that TH signaling is central for skin physiology and homeostasis. Here we review the data on the epidermis and its function in relation to TH metabolism and regulation of gene expression. An understanding of the cellular and molecular basis of TH action in epidermal cells may lead to the identification of putative therapeutical targets for treatment of skin disorders.
Collapse
Affiliation(s)
| | - Annarita Sibilio
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Monica Dentice
- Department of Clinical Medicine Surgery, University of Naples Federico II, Napoli, Italy
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Fondazione IRCCS SDN, Napoli, Italy
- *Correspondence: Caterina Missero, CEINGE Biotecnologie Avanzate, via Gaetano Salvatore 486, Napoli 80145, Italy e-mail:
| |
Collapse
|
141
|
Petersson M, Frances D, Niemann C. Lineage tracing of hair follicle stem cells in epidermal whole mounts. Methods Mol Biol 2013; 989:45-60. [PMID: 23483386 DOI: 10.1007/978-1-62703-330-5_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Lineage tracing of tissue stem cells represents a powerful tool to address fundamental questions of deve-lopment, differentiation and cellular renewal in a natural tissue environment. The Cre/lox site-specific recombination system is increasingly used to genetically label specific cell populations to perform cell lineage tracing or fate mapping experiments in sophisticated mouse models. Here we describe a method of labeling and subsequent tracking stem cells of the hair follicle bulge region in mouse skin. Hair follicle stem cells are specifically labeled by expressing the Cre recombinase under control of keratin15 (K15) regulatory sequences and by crossing the Cre-containing animals with Cre-sensitive Rosa26R (R26R) reporter mice. To achieve a temporal control of recombinase activity in stem cells, Cre is fused to a modified estrogen receptor (CreER(G)T2). In the K15CreER(G)T2/R26R mouse model, hair follicle stem cells (HFSCs) are specifically labeled after Cre activation upon treatment of mice with tamoxifen. By analyzing the skin tissue at different time points following genetic labeling, important information on stem cell behavior and contribution of labeled stem cells to epidermal structures during tissue homeostasis and hair follicle regeneration are obtained. Combining the lineage tracing approach with the whole mount technique allows examining large areas of the epidermis containing many hair follicles and sebaceous glands and reveals the complex three-dimensional relationship of labeled stem cell clones within the tissue.
Collapse
Affiliation(s)
- Monika Petersson
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | | | | |
Collapse
|
142
|
Niimori D, Kawano R, Felemban A, Niimori-Kita K, Tanaka H, Ihn H, Ohta K. Tsukushi controls the hair cycle by regulating TGF-β1 signaling. Dev Biol 2012; 372:81-7. [DOI: 10.1016/j.ydbio.2012.08.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 08/27/2012] [Accepted: 08/30/2012] [Indexed: 12/13/2022]
|
143
|
Abstract
Tissue renewal is an ongoing process in the epithelium of the skin. We have begun to examine the genetic mechanisms that control stem/progenitor cell activation in the postnatal epidermis. The conserved Hippo pathway regulates stem cell turnover in arthropods through to vertebrates. Here we show that its downstream effector, yes-associated protein (YAP), is active in the stem/progenitor cells of the postnatal epidermis. Overexpression of a C-terminally truncated YAP mutant in the basal epidermis of transgenic mice caused marked expansion of epidermal stem/progenitor cell populations. Our data suggest that the C-terminus of YAP controls the balance between stem/progenitor cell proliferation and differentiation in the postnatal interfollicular epidermis. We conclude that YAP functions as a molecular switch of stem/progenitor cell activation in the epidermis. Moreover, our results highlight YAP as a possible therapeutic target for diseases such as skin cancer, psoriasis, and epidermolysis bullosa.
Collapse
|
144
|
Stem cell expansion during carcinogenesis in stem cell-depleted conditional telomeric repeat factor 2 null mutant mice. Oncogene 2012. [PMID: 23178498 PMCID: PMC3910501 DOI: 10.1038/onc.2012.555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To examine the role of TRF2 in epithelial tumorigenesis, we characterized conditional loss of TRF2 expression in the basal layer of mouse epidermis. These mice exhibit some characteristics of dyskeratosis congenita, a human stem cell depletion syndrome caused by telomere dysfunction. The epidermis in conditional TRF2 null mice exhibited DNA damage response and apoptosis which correlated with stem cell depletion. The stem cell population in conditional TRF2 null epidermis exhibited shorter telomeres than those in control mice. Squamous cell carcinomas induced in conditional TRF2 null mice developed with increased latency and slower growth due to reduced numbers of proliferating cells as the result of increased apoptosis. TRF2 null epidermal stem cells were found in both primary and metastatic tumors. Despite the low grade phenotype of the conditional TRF2 null primary tumors, the number of metastatic lesions was similar to control cancers. Basal cells from TRF2 null tumors demonstrated extreme telomere shortening and dramatically increased numbers of telomeric signals by fluorescence in situ hybridization due to increased genomic instability and aneuploidy in these cancers. DNA damage response signals were detected at telomeres in TRF2 null tumor cells from these mice. The increased genomic instability in these tumors correlated with 8 fold expansion of the transformed stem cell population compared to that in control cancers. We concluded that genomic instability resulting from loss of TRF2 expression provides biological advantages to the cancer stem cell population.
Collapse
|
145
|
Plikus MV, Gay DL, Treffeisen E, Wang A, Supapannachart RJ, Cotsarelis G. Epithelial stem cells and implications for wound repair. Semin Cell Dev Biol 2012; 23:946-53. [PMID: 23085626 DOI: 10.1016/j.semcdb.2012.10.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 12/19/2022]
Abstract
Activation of epithelial stem cells and efficient recruitment of their proliferating progeny plays a critical role in cutaneous wound healing. The reepithelialized wound epidermis has a mosaic composition consisting of progeny that can be traced back both to epidermal and several types of hair follicle stem cells. The contribution of hair follicle stem cells to wound epidermis is particularly intriguing as it involves lineage identity change from follicular to epidermal. Studies from our laboratory show that hair follicle-fated bulge stem cells commit only transient amplifying epidermal progeny that participate in the initial wound re-epithelialization, but eventually are outcompeted by other epidermal clones and largely disappear after a few months. Conversely, recently described stem cell populations residing in the isthmus portion of hair follicle contribute long-lasting progeny toward wound epidermis and, arguably, give rise to new interfollicular epidermal stem cells. The role of epithelial stem cells during wound healing is not limited to regenerating stratified epidermis. By studying regenerative response in large cutaneous wounds, our laboratory uncovered that epithelial cells in the center of the wound can acquire greater morphogenetic plasticity and, together with the underlying wound dermis, can engage in an embryonic-like process of hair follicle neogenesis. Future studies should uncover the cellular and signaling basis of this remarkable adult wound regeneration phenomenon.
Collapse
Affiliation(s)
- Maksim V Plikus
- Department of Dermatology, Kligman Laboratories, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
146
|
Nakamura M, Schneider MR, Schmidt-Ullrich R, Paus R. Mutant laboratory mice with abnormalities in hair follicle morphogenesis, cycling, and/or structure: an update. J Dermatol Sci 2012; 69:6-29. [PMID: 23165165 DOI: 10.1016/j.jdermsci.2012.10.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 12/17/2022]
Abstract
Human hair disorders comprise a number of different types of alopecia, atrichia, hypotrichosis, distinct hair shaft disorders as well as hirsutism and hypertrichosis. Their causes vary from genodermatoses (e.g. hypotrichoses) via immunological disorders (e.g. alopecia areata, autoimmune cicatrical alopecias) to hormone-dependent abnormalities (e.g. androgenetic alopecia). A large number of spontaneous mouse mutants and genetically engineered mice develop abnormalities in hair follicle morphogenesis, cycling, and/or hair shaft formation, whose analysis has proven invaluable to define the molecular regulation of hair growth, ranging from hair follicle development, and cycling to hair shaft formation and stem cell biology. Also, the accumulating reports on hair phenotypes of mouse strains provide important pointers to better understand the molecular mechanisms underlying human hair growth disorders. Since numerous new mouse mutants with a hair phenotype have been reported since the publication of our earlier review on this matter a decade ago, we present here an updated, tabulated mini-review. The updated annotated tables list a wide selection of mouse mutants with hair growth abnormalities, classified into four categories: Mutations that affect hair follicle (1) morphogenesis, (2) cycling, (3) structure, and (4) mutations that induce extrafollicular events (for example immune system defects) resulting in secondary hair growth abnormalities. This synthesis is intended to provide a useful source of reference when studying the molecular controls of hair follicle growth and differentiation, and whenever the hair phenotypes of a newly generated mouse mutant need to be compared with existing ones.
Collapse
Affiliation(s)
- Motonobu Nakamura
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | | | | | | |
Collapse
|
147
|
Jiang R, Li Y, Xu Y, Zhou Y, Pang Y, Shen L, Zhao Y, Zhang J, Zhou J, Wang X, Liu Q. EMT and CSC-like properties mediated by the IKKβ/IκBα/RelA signal pathway via the transcriptional regulator, Snail, are involved in the arsenite-induced neoplastic transformation of human keratinocytes. Arch Toxicol 2012; 87:991-1000. [PMID: 23069812 DOI: 10.1007/s00204-012-0933-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/28/2012] [Indexed: 12/27/2022]
Abstract
Exposure of humans to inorganic arsenic can cause skin cancer. The epithelial-mesenchymal transition (EMT) and acquisition of cancer stem cell (CSC)-like properties are essential steps in the initiation of human skin cancers; however, the mechanisms of action remain obscure. We have found that, during the neoplastic transformation induced by a low concentration (1.0 μM) of arsenite in human keratinocyte HaCaT cells, the cells undergo an EMT and then acquire a malignant CSC-like phenotype. With longer times for transformation of HaCaT cells, there were increased activations of IκB kinase β (IKKβ), inhibitor nuclear factor-kappa B alpha (IκBα), and nuclear factor κB (NF-κB) RelA and increases in the level of Snail. Further, during the transformation of HaCaT cells, the activation of NF-κB RelA up-regulated Snail levels. Inhibition of NF-κB RelA blocked the arsenite-induced EMT, acquisition of a CSC-like phenotype, and neoplastic transformation. These observations show that EMT, along with acquisition of a CSC-like phenotype mediated by IKKβ/IκBα/RelA signal pathway via Snail, contributes to a low concentration of arsenite-induced tumorigenesis.
Collapse
Affiliation(s)
- Rongrong Jiang
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 210029, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Terskikh VV, Vasiliev AV, Vorotelyak EA. Label retaining cells and cutaneous stem cells. Stem Cell Rev Rep 2012; 8:414-25. [PMID: 21744048 DOI: 10.1007/s12015-011-9299-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This is a comprehensive review on label retaining cells (LRC) in epidermal development and homeostasis. The precise in vivo identification and location of epidermal stem cells is a crucial issue in cutaneous biology. We discuss here the following problems: (1) Identification and location of LRC in the interfollicular epithelium and hair follicle; (2) The proliferative potential of LRC and their role in cutaneous homeostasis (3); LRC phenomenon and the Immortal Strand Hypothesis, which suggests an alternative mechanism for retention of genetic information; (4) Significance of LRC studies for development of stem cell concept. Now, it seems evident that LRC are a frequent feature of stem cell niches and revealing highly dormant LRC may be used for identification of stem cell niches in different tissues. LRC were used for screening specific markers of epidermal stem cells. Within a given tissue stem cells have different proliferative characteristics. There are more frequently cycling stem cells which function primarily in homeostasis, while LRC form a reserve of dormant, may be ultimate, stem cells, which are set aside for regeneration of injury or unforeseen need. The authors suggest that LRC dormancy described in Mammalia has much in common with developmental quiescence found in some other animals. For example in C. elegans reproductive system, vulval precursor cells have developmentally programmed cell-cycle arrest in the first larval stage, and then undergo an extended period of quiescence before resuming proliferation. Another example of developmental quiescence is the diapause, a widespread phenomenon exhibited by animals ranging from nematodes to mammals, often occurring at genetically predetermined life history stage.
Collapse
Affiliation(s)
- Vasily V Terskikh
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
149
|
Sennett R, Rendl M. Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling. Semin Cell Dev Biol 2012; 23:917-27. [PMID: 22960356 DOI: 10.1016/j.semcdb.2012.08.011] [Citation(s) in RCA: 270] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/27/2012] [Accepted: 08/24/2012] [Indexed: 12/17/2022]
Abstract
Embryonic hair follicle induction and formation are regulated by mesenchymal-epithelial interactions between specialized dermal cells and epidermal stem cells that switch to a hair fate. Similarly, during postnatal hair growth, communication between mesenchymal dermal papilla cells and surrounding epithelial matrix cells coordinates hair shaft production. Adult hair follicle regeneration in the hair cycle again is thought to be controlled by activating signals originating from the mesenchymal compartment and acting on hair follicle stem cells. Although many signaling pathways are implicated in hair follicle formation and growth, the precise nature, timing, and intersection of these inductive and regulatory signals remains elusive. The goal of this review is to summarize our current understanding and to discuss recent new insights into mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling.
Collapse
Affiliation(s)
- Rachel Sennett
- Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
150
|
Myung PS, Takeo M, Ito M, Atit RP. Epithelial Wnt ligand secretion is required for adult hair follicle growth and regeneration. J Invest Dermatol 2012; 133:31-41. [PMID: 22810306 PMCID: PMC3479363 DOI: 10.1038/jid.2012.230] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
β-Catenin, a key transducer molecule of Wnt signaling, is required for adult hair follicle growth and regeneration. However, the cellular source of Wnt ligands required for Wnt/β-catenin activation during anagen induction is unknown. In this study, we genetically deleted Wntless (Wls), a gene required for Wnt ligand secretion by Wnt-producing cells, specifically in the hair follicle epithelium during telogen phase. We show that epithelial Wnt ligands are required for anagen, as loss of Wls in the follicular epithelium resulted in a profound hair cycle arrest. Both the follicular epithelium and dermal papilla showed markedly decreased Wnt/β-catenin signaling during anagen induction compared with control hair follicles. Surprisingly, hair follicle stem cells that are responsible for hair regeneration maintained expression of stem cell markers but exhibited significantly reduced proliferation. Finally, we demonstrate that epidermal Wnt ligands are critical for adult wound-induced de novo hair formation. Collectively, these data show that Wnt ligands secreted by the hair follicle epithelium are required for adult hair follicle regeneration and provide new insight into potential cellular targets for the treatment of hair disorders such as alopecia.
Collapse
Affiliation(s)
- Peggy S Myung
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | |
Collapse
|