1
|
Yusupova M, Ankawa R, Yosefzon Y, Meiri D, Bachelet I, Fuchs Y. Apoptotic dysregulation mediates stem cell competition and tissue regeneration. Nat Commun 2023; 14:7547. [PMID: 37985759 PMCID: PMC10662150 DOI: 10.1038/s41467-023-41684-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 09/14/2023] [Indexed: 11/22/2023] Open
Abstract
Since adult stem cells are responsible for replenishing tissues throughout life, it is vital to understand how failure to undergo apoptosis can dictate stem cell behavior both intrinsically and non-autonomously. Here, we report that depletion of pro-apoptotic Bax protein bestows hair follicle stem cells with the capacity to eliminate viable neighboring cells by sequestration of TNFα in their membrane. This in turn induces apoptosis in "loser" cells in a contact-dependent manner. Examining the underlying mechanism, we find that Bax loss-of-function competitive phenotype is mediated by the intrinsic activation of NFκB. Notably, winner stem cells differentially respond to TNFα, owing to their elevated expression of TNFR2. Finally, we report that in vivo depletion of Bax results in an increased stem cell pool, accelerating wound-repair and de novo hair follicle regeneration. Collectively, we establish a mechanism of mammalian cell competition, which can have broad therapeutic implications for tissue regeneration and tumorigenesis.
Collapse
Affiliation(s)
- Marianna Yusupova
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Roi Ankawa
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
- Augmanity, Rehovot, Israel
| | - Yahav Yosefzon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - David Meiri
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Yaron Fuchs
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
- Augmanity, Rehovot, Israel.
| |
Collapse
|
2
|
Abstract
Diseases affecting the hair follicle are common in domestic animals, but despite the importance of an intact skin barrier and a fully functional hair coat, knowledge about the detailed morphological features and the diversity of these complex mini-organs are often limited, although mandatory to evaluate skin biopsies with a history of alopecia. The factors that regulate the innate hair follicle formation and the postnatal hair cycle are still not completely understood in rodents, only rudimentarily known in humans, and are poorly understood in our companion animals. This review aims to summarize the current knowledge about hair follicle and hair shaft anatomy, the arrangement of hair follicles, hair follicle morphogenesis in the embryo, and the lifelong regeneration during the postnatal hair cycle in domestic animals. The role of follicular stem cells and the need for a multitude of interacting signaling events during hair follicle morphogenesis and regeneration is unquestioned. Because of the lack of state of the art methods that can be applied in rodents but are not feasible in companion animals, most of the information in this review is based on rodent studies. However, the few data from domestic animals that are available will be discussed, and it can be assumed that at least the principal molecular mechanisms are similar in rodents and other species.
Collapse
|
3
|
Keister BD, Mesa KR, Blagoev KB. Apoptotic cells may drive cell death in hair follicles during their regression cycle. Oncotarget 2023; 14:893-899. [PMID: 37861373 PMCID: PMC10588663 DOI: 10.18632/oncotarget.28529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Intravital microscopy in live mice has shown that the elimination of epithelial cells during hair follicle regression involves supra-basal cell differentiation and basal cell apoptosis through synergistic action of TGF-β (transforming growth factor) and mesenchymal-epithelial interactions. In this process the basal epithelial cells are not internally committed to death and the mesenchymal dermal papilla (DP) plays essential role in death induction. Because the DP cells are not necessary for completion of the cycle but only for its initiation it is still an open question what is the mechanism leading to the propagation of apoptosis towards the regenerative stem cell population. Here, we use a quantitative analysis of the length of hair follicles during their regression cycle. The data are consistent with a propagation mechanism driven by apoptotic cells inducing apoptosis in their neighboring cells. The observation that the apoptosis slows down as the apoptotic front approaches the stem cells at the end of the follicle is consistent with a gradient of a pro-survival signal sent by these stem cells. An experiment that can falsify this mechanism is proposed.
Collapse
Affiliation(s)
- Bradley D. Keister
- National Science Foundation, Physics Division, Alexandria, VA 22230, USA
| | - Kailin R. Mesa
- The Jane Coffin Childs Memorial Fund for Medical Research, New Haven, CT 06520, USA
- Department of Genetics and Dermatology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Krastan B. Blagoev
- National Science Foundation, Physics Division, Alexandria, VA 22230, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR3664, Laboratoire Dynamique du Noyau, Paris 75005, France
| |
Collapse
|
4
|
Chovatiya G, Li KN, Li J, Ghuwalewala S, Tumbar T. Alk1 acts in non-endothelial VE-cadherin + perineurial cells to maintain nerve branching during hair homeostasis. Nat Commun 2023; 14:5623. [PMID: 37699906 PMCID: PMC10497554 DOI: 10.1038/s41467-023-40761-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/09/2023] [Indexed: 09/14/2023] Open
Abstract
Vascular endothelial (VE)-cadherin is a well-recognized endothelial cell marker. One of its interacting partners, the TGF-β receptor Alk1, is essential in endothelial cells for adult skin vasculature remodeling during hair homeostasis. Using single-cell transcriptomics, lineage tracing and gene targeting in mice, we characterize the cellular and molecular dynamics of skin VE-cadherin+ cells during hair homeostasis. We describe dynamic changes of VE-cadherin+ endothelial cells specific to blood and lymphatic vessels and uncover an atypical VE-cadherin+ cell population. The latter is not a predicted adult endovascular progenitor, but rather a non-endothelial mesenchymal perineurial cell type, which forms nerve encapsulating tubular structures that undergo remodeling during hair homeostasis. Alk1 acts in the VE-cadherin+ perineurial cells to maintain proper homeostatic nerve branching by enforcing basement membrane and extracellular matrix molecular signatures. Our work implicates the VE-cadherin/Alk1 duo, classically known as endothelial-vascular specific, in perineurial-nerve homeostasis. This has broad implications in vascular and nerve disease.
Collapse
Affiliation(s)
- Gopal Chovatiya
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Kefei Nina Li
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Jonathan Li
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Sangeeta Ghuwalewala
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Tudorita Tumbar
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
5
|
Chovatiya G, Li KN, Ghuwalewala S, Tumbar T. Single-cell transcriptomics of adult skin VE-cadherin expressing lineages during hair cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533784. [PMID: 36993228 PMCID: PMC10055414 DOI: 10.1101/2023.03.22.533784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Adult skin homeostasis involves global reorganization of dermal lineages at different stages of the mouse hair growth cycle. Vascular endothelial cadherin (VE-cadherin encoded by Cdh5 ) expressing cells from blood and lymphatic vasculature structures are known to remodel during the adult hair cycle. Here we employ single-cell RNA-sequencing (scRNA-seq) 10x-genomics analysis of FACS-sorted VE-cadherin expressing cells marked via Cdh5-CreER genetic labeling at resting (telogen) and growth (anagen) stage of hair cycle. Our comparative analysis between the two stages uncovers a persistent Ki67 + proliferative EC population and documents changes in EC population distribution and gene expression. Global gene expression changes in all the analyzed populations revealed bioenergetic metabolic changes that may drive vascular remodeling during HF growth phase, alongside a few highly restricted cluster-specific gene expression differences. This study uncovers active cellular and molecular dynamics of adult skin endothelial lineages during hair cycle that may have broad implications in adult tissue regeneration and for understanding vascular disease.
Collapse
|
6
|
A reductionist approach to determine the effect of cell-cell contact on human epidermal stem cell differentiation. Acta Biomater 2022; 150:265-276. [PMID: 35926780 PMCID: PMC9810539 DOI: 10.1016/j.actbio.2022.07.054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 01/07/2023]
Abstract
The balance between stem cell renewal and differentiation is determined by the interplay between intrinsic cellular controls and extrinsic factors presented by the microenvironment, or 'niche'. Previous studies on cultured human epidermis have utilised suspension culture and restricted cell spreading to investigate regulation of differentiation in single keratinocytes. However, keratinocytes are typically adherent to neighbouring cells in vivo. We therefore developed experimental models to investigate the combined effects of cell-ECM adhesion and cell-cell contact. We utilized lipid-modified oligonucleotides to form clusters of keratinocytes which were subsequently placed in suspension to induce terminal differentiation. In this experimental model cell-cell contact had no effect on suspension-induced differentiation of keratinocytes. We next developed a high-throughput platform for robust geometrical confinement of keratinocytes to hexagonal ECM-coated islands permitting direct cell-cell contact between single cells. As in the case of circular islands, differentiation was stimulated on the smallest single hexagonal islands. However, the percentage of involucrin-positive cells on small bowtie islands was significantly lower than on single islands, demonstrating that cell-cell contact reduced differentiation in response to decreased substrate adhesion. None of the small bowtie islands contained two involucrin-positive cells. Rather, if one cell was involucrin-positive the other was involucrin-negative. This suggests that there is intrinsic asymmetry in the effect of cell-cell contact in decreasing differentiation. Thus, our reductionist approaches provide new insights into the effect of the niche on keratinocyte differentiation. STATEMENT OF SIGNIFICANCE: Stem cell behaviour is regulated by a combination of external signals, including the nature of the adhesive substrate and cell-cell interactions. An understanding of how different signals are integrated creates the possibility of developing new biomaterials to promote tissue regeneration and broaden our understanding of skin diseases such as eczema and psoriasis, in which stem cell proliferation and differentiation are perturbed. In this study we have applied two methods to engineer intercellular adhesion of human epidermal stem cells, one involving lipid-modified DNA and the other involving hexagonal micropatterns. We show that the effect of cell-cell adhesion depends on cell-substrate adhesion and uncover evidence that two cells in equivalent environments can nevertheless behave differently.
Collapse
|
7
|
Sasaki GH. Clinical Use of Extracellular Vesicles in the Management of Male and Female Pattern Hair Loss: A Preliminary Retrospective IRB Safety and Efficacy Study. Aesthet Surg J Open Forum 2022; 4:ojac045. [PMID: 35923863 PMCID: PMC9342625 DOI: 10.1093/asjof/ojac045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
Background
Pattern hair loss is a common disorder in female and male subjects that may benefit from the use of cell-free XoFlo (Direct Biologics, LLC, Austin, TX) therapy.
Objectives
To assess the safety, efficacy and satisfaction of a single extracellular vesicle (EV) treatment over 6 months.
Methods
A retrospective open-label study among 22 female and 9 male subjects who demonstrated early stages of alopecia or were in remission from prior medical and surgical treatments. The amount of undiluted or diluted volumes of EV solution used was determined by extent and degree of alopecia. Global photography, SGAIS and IGAIS questionnaires, and trichoscan measurements were compared at baseline and six months in three response categories.
Results
Frequent growth responses were observed: older aged females and younger aged males, shorter history of alopecia; earlier stages of hair loss; larger and undiluted volumes of XoFlo; prior positive responses to medical and surgical treatments; and absence or control of disease factors affecting hair. The benefit of micro-needling to therapy was indeterminate. Global photography, trichoscan for density, follicle diameter, terminal: vellus ratio, and SGAIS/IGAIS satisfaction questionnaires at baseline and six months were useful in assessing clinical efficacy. No significant adverse reactions were observed.
Conclusions
Intradermal injections with varying doses of EVs were safe and effective among indicated alopecic female and male subjects. Findings suggest that the presence of positive factors, absence of conditions known to negatively affect hair growth, and administration of larger volumes of XoFlo may have a significant influence on the use of this new cell-free therapy. FDA-approved biologic, multi-centered IRB/ Investigational New Drug (IND) trials are clearly required to determine its future in the management of hair loss.
Collapse
|
8
|
Kocagöz Y, Demirler MC, Eski SE, Güler K, Dokuzluoglu Z, Fuss SH. Disparate progenitor cell populations contribute to maintenance and repair neurogenesis in the zebrafish olfactory epithelium. Cell Tissue Res 2022; 388:331-358. [PMID: 35266039 DOI: 10.1007/s00441-022-03597-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 02/02/2022] [Indexed: 12/25/2022]
Abstract
Olfactory sensory neurons (OSNs) undergo constant turnover under physiological conditions but also regenerate efficiently following tissue injury. Maintenance and repair neurogenesis in the olfactory epithelium (OE) have been attributed to the selective activity of globose (GBCs) and horizontal basal cells (HBCs), respectively. In zebrafish, cells with GBC-like properties are localized to the peripheral margins of the sensory OE and contribute to OSN neurogenesis in the intact OE, while cells that resemble HBCs at the morphological and molecular level are more uniformly distributed. However, the contribution of these cells to the restoration of the injured OE has not been demonstrated. Here, we provide a detailed cellular and molecular analysis of the tissue response to injury and show that a dual progenitor cell system also exists in zebrafish. Zebrafish HBCs respond to the structural damage of the OE and generate a transient population of proliferative neurogenic progenitors that restores OSNs. In contrast, selective ablation of OSNs by axotomy triggers neurogenic GBC proliferation, suggesting that distinct signaling events activate GBC and HBC responses. Molecular analysis of differentially expressed genes in lesioned and regenerating OEs points toward an involvement of the canonical Wnt/β-catenin pathway. Activation of Wnt signaling appears to be sufficient to stimulate mitotic activity, while inhibition significantly reduces, but does not fully eliminate, HBC responses. Zebrafish HBCs are surprisingly active even under physiological conditions with a strong bias toward the zones of constitutive OSN neurogenesis, suggestive of a direct lineage relationship between progenitor cell subtypes.
Collapse
Affiliation(s)
- Yigit Kocagöz
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey
| | - Mehmet Can Demirler
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey
| | - Sema Elif Eski
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey
- Institute of Interdisciplinary Research in Human and Molecular Biology, Free University of Brussels, Campus Erasme, 1070, Brussels, Belgium
| | - Kardelen Güler
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey
| | - Zeynep Dokuzluoglu
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey
| | - Stefan H Fuss
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Kuzey Park 319, 34342, Bebek - Istanbul, Turkey.
| |
Collapse
|
9
|
Li H, Ziemer M, Stojanovic I, Saksida T, Maksimovic-Ivanic D, Mijatovic S, Djmura G, Gajic D, Koprivica I, Krajnovic T, Draca D, Simon JC, Lethaus B, Savkovic V. Mesenchymal Stem Cells From Mouse Hair Follicles Reduce Hypertrophic Scarring in a Murine Wound Healing Model. Stem Cell Rev Rep 2022; 18:2028-2044. [PMID: 35080748 PMCID: PMC9391240 DOI: 10.1007/s12015-021-10288-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
Wound healing of acute full-thickness injuries and chronic non-healing ulcers leads to delayed wound closure, prolonged recovery period and hypertrophic scarring, generating a demand for an autologous cell therapy and a relevant pre-clinical research models for wound healing. In this study, an immunocompetent model for wound healing was employed using a syngeneic murine cell line of mesenchymal stem cells cultured from the mouse whisker hair follicle outer root sheath (named moMSCORS). moMSCORS were isolated using an air-liquid interface method, expanded in vitro and characterized according to the MSC definition criteria - cell viability, in vitro proliferation, MSC phenotype and multi-lineage differentiations. Moreover, upon applying moMSCORS in an in vivo full-thickness wound model in the syngeneic C57BL/6 mice, the treated wounds displayed different morphology to that of the untreated wound beds. Quantitative evaluation of angiogenesis, granulation and wound closure involving clinical scoring and software-based quantification indicated a lower degree of inflammation in the treated wounds. Histological staining of treated wounds by the means of H&E, Alcian Blue, PicroSirius Red and αSMA immune labelling showed lower cellularity, less collagen filaments as well as thinner dermal and epidermal layers compared with the untreated wounds, indicating a general reduction of hypertrophic scars. The decreased inflammation, accelerated wound closure and non-hypertrophic scarring, which were facilitated by moMSCORS, hereby address a common problem of hypertrophic scars and non-physiological tissue properties upon wound closure, and additionally offer an in vivo model for the autologous cell-based wound healing.
Collapse
Affiliation(s)
- Hanluo Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, Hubei University of Technology, Wuhan, 430068, Hubei Province, China.,Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany
| | - Mirjana Ziemer
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Ivana Stojanovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Maksimovic-Ivanic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Sanja Mijatovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Goran Djmura
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Dragica Gajic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivan Koprivica
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Krajnovic
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dijana Draca
- Institute for Biological Research "Sinisa Stankovic" (IBISS) - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jan-Christoph Simon
- Clinic for Dermatology, Venereology and Allergology, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Bernd Lethaus
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany
| | - Vuk Savkovic
- Department of Cranial Maxillofacial Plastic Surgery, University Clinic Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
10
|
Aleemardani M, Trikić MZ, Green NH, Claeyssens F. The Importance of Mimicking Dermal-Epidermal Junction for Skin Tissue Engineering: A Review. Bioengineering (Basel) 2021; 8:bioengineering8110148. [PMID: 34821714 PMCID: PMC8614934 DOI: 10.3390/bioengineering8110148] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022] Open
Abstract
There is a distinct boundary between the dermis and epidermis in the human skin called the basement membrane, a dense collagen network that creates undulations of the dermal-epidermal junction (DEJ). The DEJ plays multiple roles in skin homeostasis and function, namely, enhancing the adhesion and physical interlock of the layers, creating niches for epidermal stem cells, regulating the cellular microenvironment, and providing a physical boundary layer between fibroblasts and keratinocytes. However, the primary role of the DEJ has been determined as skin integrity; there are still aspects of it that are poorly investigated. Tissue engineering (TE) has evolved promising skin regeneration strategies and already developed TE scaffolds for clinical use. However, the currently available skin TE equivalents neglect to replicate the DEJ anatomical structures. The emergent ability to produce increasingly complex scaffolds for skin TE will enable the development of closer physical and physiological mimics to natural skin; it also allows researchers to study the DEJ effect on cell function. Few studies have created patterned substrates that could mimic the human DEJ to explore their significance. Here, we first review the DEJ roles and then critically discuss the TE strategies to create the DEJ undulating structure and their effects. New approaches in this field could be instrumental for improving bioengineered skin substitutes, creating 3D engineered skin, identifying pathological mechanisms, and producing and screening drugs.
Collapse
Affiliation(s)
- Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
| | - Michael Zivojin Trikić
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
| | - Nicola Helen Green
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield S1 3JD, UK
| | - Frederik Claeyssens
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
- Correspondence:
| |
Collapse
|
11
|
Ankawa R, Goldberger N, Yosefzon Y, Koren E, Yusupova M, Rosner D, Feldman A, Baror-Sebban S, Buganim Y, Simon DJ, Tessier-Lavigne M, Fuchs Y. Apoptotic cells represent a dynamic stem cell niche governing proliferation and tissue regeneration. Dev Cell 2021; 56:1900-1916.e5. [PMID: 34197726 DOI: 10.1016/j.devcel.2021.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 12/14/2020] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
Stem cells (SCs) play a key role in homeostasis and repair. While many studies have focused on SC self-renewal and differentiation, little is known regarding the molecular mechanism regulating SC elimination and compensation upon loss. Here, we report that Caspase-9 deletion in hair follicle SCs (HFSCs) attenuates the apoptotic cascade, resulting in significant temporal delays. Surprisingly, Casp9-deficient HFSCs accumulate high levels of cleaved caspase-3 and are improperly cleared due to an essential caspase-3/caspase-9 feedforward loop. These SCs are retained in an apoptotic-engaged state, serving as mitogenic signaling centers by continuously releasing Wnt3 and instructing proliferation. Investigating the underlying mechanism, we reveal a caspase-3/Dusp8/p38 module responsible for Wnt3 induction, which operates in both normal and Casp9-deleted HFSCs. Notably, Casp9-deleted mice display accelerated wound repair and de novo hair follicle regeneration. Taken together, we demonstrate that apoptotic cells represent a dynamic SC niche, from which emanating signals drive SC proliferation and tissue regeneration.
Collapse
Affiliation(s)
- Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nitzan Goldberger
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yahav Yosefzon
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Marianna Yusupova
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daniel Rosner
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alona Feldman
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shulamit Baror-Sebban
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, the Hebrew University of Jerusalem, Hadassah Medical School, Jerusalem, Israel
| | - David J Simon
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
12
|
Li KN, Tumbar T. Hair follicle stem cells as a skin-organizing signaling center during adult homeostasis. EMBO J 2021; 40:e107135. [PMID: 33880808 PMCID: PMC8167365 DOI: 10.15252/embj.2020107135] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cells are the essential source of building blocks for tissue homeostasis and regeneration. Their behavior is dictated by both cell-intrinsic cues and extrinsic cues from the microenvironment, known as the stem cell niche. Interestingly, recent work began to demonstrate that hair follicle stem cells (HFSCs) are not only passive recipients of signals from the surroundings, but also actively send out signals to modulate the organization and function of their own niches. Here, we discuss recent findings, and briefly refer to the old, on the interaction of HFSCs and their niches with the emphasis on the outwards signals from HFSCs toward their niches. We also highlight recent technology advancements that further promote our understanding of HFSC niches. Taken together, the HFSCs emerge as a skin-organizing center rich in signaling output for niche remodeling during various stages of adult skin homeostasis. The intricate crosstalk between HFSCs and their niches adds important insight to skin biology that will inform clinical and bioengineering fields aiming to build complete and functional 3D organotypic cultures for skin replacement therapies.
Collapse
Affiliation(s)
- Kefei Nina Li
- Molecular Biology and GeneticsCornell UniversityIthacaNYUSA
| | | |
Collapse
|
13
|
Martino PA, Heitman N, Rendl M. The dermal sheath: An emerging component of the hair follicle stem cell niche. Exp Dermatol 2021; 30:512-521. [PMID: 33006790 PMCID: PMC8016715 DOI: 10.1111/exd.14204] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/15/2020] [Accepted: 09/20/2020] [Indexed: 12/17/2022]
Abstract
Hair follicles cyclically regenerate throughout adult mammalian life, owing to a resident population of epithelial hair follicle stem cells. Stem cell (SC) activity drives bouts of follicle growth, which are periodically interrupted by follicle regression and rest. These phases and the transitions between them are tightly spatiotemporally coordinated by signalling crosstalk between stem/progenitor cells and the various cell types of the microenvironment, or niche. The dermal papilla (DP) is a cluster of specialized mesenchymal cells that have long been recognized for important niche roles in regulating hair follicle SC activation, as well as progenitor proliferation and differentiation during follicle growth. In addition to the DP, the mesenchyme of the murine pelage follicle is also comprised of a follicle-lining smooth muscle known as the dermal sheath (DS), which has been far less studied than the DP yet may be equally specialized and important for hair cycling. In this review, we define the murine pelage DS in comparison with human DS and discuss recent work that highlights the emergent importance of the DS in the hair follicle SC niche. Last, we examine potential therapeutic applications for the DS in hair regeneration and wound healing.
Collapse
Affiliation(s)
- Pieter A. Martino
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
| | - Nicholas Heitman
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Atran Building AB7-10C, Box 1020; 1428 Madison Ave, New York, NY 10029, USA
| |
Collapse
|
14
|
Differentiated Daughter Cells Regulate Stem Cell Proliferation and Fate through Intra-tissue Tension. Cell Stem Cell 2020; 28:436-452.e5. [PMID: 33264636 DOI: 10.1016/j.stem.2020.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
Basal stem cells fuel development, homeostasis, and regeneration of the epidermis. The proliferation and fate decisions of these cells are highly regulated by their microenvironment, including the basement membrane and underlying mesenchymal cells. Basal progenitors give rise to differentiated progeny that generate the epidermal barrier. Here, we present data that differentiated progeny also regulate the proliferation, differentiation, and migration of basal progenitor cells. Using two distinct mouse lines, we found that increasing contractility of differentiated cells resulted in non-cell-autonomous hyperproliferation of stem cells and prevented their commitment to a hair follicle lineage. This increased contractility also impaired movement of basal progenitors during hair placode morphogenesis and diminished migration of melanoblasts. These data suggest that intra-tissue tension regulates stem cell proliferation, fate decisions, and migration and that differentiated epidermal keratinocytes are a component of the stem cell niche that regulates development and homeostasis of the skin.
Collapse
|
15
|
Chen X, Yang R, Wang J, Ruan S, Lin Z, Xin Q, Yang R, Xie J. Porcine acellular dermal matrix accelerates wound healing through miR-124-3p.1 and miR-139-5p. Cytotherapy 2020; 22:494-502. [PMID: 32571650 DOI: 10.1016/j.jcyt.2020.04.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Cutaneous wound management is a major health problem and imposes a huge economic burden worldwide. Previous studies have demonstrated that wound healing is a highly coordinated process including epithelialization, angiogenesis, remodeling and scarring. This progression requires self-renewal, preservation and repair properties of stem cells. However, our understanding of the detailed internal regulatory mechanism following injury and the means to accelerate wound healing are limited. METHODS Our previous research revealed that porcine acellular dermal matrix (ADM) effectively promotes wound healing and scar formation through epidermal stem cells (ESCs), and this process is relevant to the alteration of internal miRNA levels. In this study, we investigated the regulatory function of porcine ADM treatment on miRNAs in ESCs. RESULTS We report that the treatment of porcine ADM reduced the levels of miR-124-3p.1 and miR-139-5p in wounds. MiR-124-3p.1 and miR-139-5p inhibited the expression of JAG1 and Notch1, respectively, by directly targeting miRNAs in ESCs. CONCLUSIONS This work demonstrates that porcine ADM induced down-regulation of miR-124-3p.1/139-5p in wounds and up-regulation of JAG1/Notch1 in ESCs, thus enhancing cutaneous wound healing.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Ronghua Yang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Jingru Wang
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Shubin Ruan
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Zepeng Lin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Qi Xin
- Department of Burn Surgery, the First People's Hospital of Foshan, Foshan, China
| | - Ridong Yang
- Department of Dermatology, Guangzhou Dermatology and Prevention Institute, Guangzhou, China.
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
16
|
Horsley V. Lifting Each Other Up: Epidermal Stem Cells in Tissue Homeostasis. Dev Cell 2020; 51:296-298. [PMID: 31689385 DOI: 10.1016/j.devcel.2019.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Multiple stem cells maintain and repair tissues, yet how they communicate is not well understood. In this issue of Developmental Cell, Veniaminova et al. (2019) report that each sebaceous gland is maintained by local stem cells and that Notch signaling regulates multiple aspects of their function, revealing tissue homeostasis mechanisms.
Collapse
Affiliation(s)
- Valerie Horsley
- Department of Molecular, Cellular, and Developmental Biology and Department of Dermatology, Yale University, 219 Prospect Street, New Haven, CT 06520, USA.
| |
Collapse
|
17
|
Moon H, White AC, Borowsky AD. New insights into the functions of Cox-2 in skin and esophageal malignancies. Exp Mol Med 2020; 52:538-547. [PMID: 32235869 PMCID: PMC7210257 DOI: 10.1038/s12276-020-0412-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 02/08/2023] Open
Abstract
Understanding the cellular and molecular mechanisms of tumor initiation and progression for each cancer type is central to making improvements in both prevention and therapy. Identifying the cancer cells of origin and the necessary and sufficient mechanisms of transformation and progression provide opportunities for improved specific clinical interventions. In the last few decades, advanced genetic manipulation techniques have facilitated rapid progress in defining the etiologies of cancers and their cells of origin. Recent studies driven by various groups have provided experimental evidence indicating the cellular origins for each type of skin and esophageal cancer and have identified underlying mechanisms that stem/progenitor cells use to initiate tumor development. Specifically, cyclooxygenase-2 (Cox-2) is associated with tumor initiation and progression in many cancer types. Recent studies provide data demonstrating the roles of Cox-2 in skin and esophageal malignancies, especially in squamous cell carcinomas (SCCs) occurring in both sites. Here, we review experimental evidence aiming to define the origins of skin and esophageal cancers and discuss how Cox-2 contributes to tumorigenesis and differentiation.
Collapse
Affiliation(s)
- Hyeongsun Moon
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA.
| | - Andrew C White
- Department of Biological Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Alexander D Borowsky
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA
| |
Collapse
|
18
|
Kaur K, Kaur R, Bala I. Therapeutics of premature hair graying: A long journey ahead. J Cosmet Dermatol 2019; 18:1206-1214. [PMID: 31115162 DOI: 10.1111/jocd.13000] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 04/09/2019] [Accepted: 04/26/2019] [Indexed: 01/24/2023]
Abstract
Premature graying of hair has major psychosocial and socioeconomic repercussion, as it is considered as a sign of hastily progressing old age, ill health and often leads to loss of self-esteem. Hair is said to gray prematurely when it happens before the age of 20 years in Caucasians, 25 years in Asians, and 30 years in Africans. The hair color chiefly depends on melanin pigment, and fabrication of this pigment takes place in melanosomes through the process of melanogenesis. This complex biochemical pathway (melanogenesis) is further dependent on tyrosinase which acts as fuel.The normal human scalp is subjected to various factors categorized as intrinsic and extrinsic leading to graying of hair. Intrinsic factors comprise of variants responsible for changes at genetic level while extrinsic factors include air pollution, ultraviolet radiation, smoking, and nutrition. It has been proposed that direct or indirect effect of all these factors results in the generation of reactive oxygen species (ROS), thus leading to further damage. Though research has expanded in last few years in terms of microscopic, biochemical (hormonal, enzymatic), and molecular changes happening within hair follicle/shaft, still the exact mechanism leading to premature graying of hair is not well understood. Probable solutions toward this quandary are diet, herbal remedies, and temporary hair colorants. Ironically, the latter one being the most common has various side effects such as allergic reactions, inflammation, and hair loss. The aim of this paper was to review the manifestation and probable future interventions in preventing premature hair graying.
Collapse
Affiliation(s)
- Kiranjeet Kaur
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| | - Rajveer Kaur
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| | - Indu Bala
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
19
|
Wu L, Belyaeva OV, Adams MK, Klyuyeva AV, Lee SA, Goggans KR, Kesterson RA, Popov KM, Kedishvili NY. Mice lacking the epidermal retinol dehydrogenases SDR16C5 and SDR16C6 display accelerated hair growth and enlarged meibomian glands. J Biol Chem 2019; 294:17060-17074. [PMID: 31562240 DOI: 10.1074/jbc.ra119.010835] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/24/2019] [Indexed: 12/18/2022] Open
Abstract
Retinol dehydrogenases catalyze the rate-limiting step in the biosynthesis of retinoic acid, a bioactive lipid molecule that regulates the expression of hundreds of genes by binding to nuclear transcription factors, the retinoic acid receptors. Several enzymes exhibit retinol dehydrogenase activities in vitro; however, their physiological relevance for retinoic acid biosynthesis in vivo remains unclear. Here, we present evidence that two murine epidermal retinol dehydrogenases, short-chain dehydrogenase/reductase family 16C member 5 (SDR16C5) and SDR16C6, contribute to retinoic acid biosynthesis in living cells and are also essential for the oxidation of retinol to retinaldehyde in vivo Mice with targeted knockout of the more catalytically active SDR16C6 enzyme have no obvious phenotype, possibly due to functional redundancy, because Sdr16c5 and Sdr16c6 exhibit an overlapping expression pattern during later developmental stages and in adulthood. Mice that lack both enzymes are viable and fertile but display accelerated hair growth after shaving and also enlarged meibomian glands, consistent with a nearly 80% reduction in the retinol dehydrogenase activities of skin membrane fractions from the Sdr16c5/Sdr16c6 double-knockout mice. The up-regulation of hair-follicle stem cell genes is consistent with reduced retinoic acid signaling in the skin of the double-knockout mice. These results indicate that the retinol dehydrogenase activities of murine SDR16C5 and SDR16C6 enzymes are not critical for survival but are responsible for most of the retinol dehydrogenase activity in skin, essential for the regulation of the hair-follicle cycle, and required for the maintenance of both sebaceous and meibomian glands.
Collapse
Affiliation(s)
- Lizhi Wu
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Olga V Belyaeva
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Mark K Adams
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Alla V Klyuyeva
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Seung-Ah Lee
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Kelli R Goggans
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Robert A Kesterson
- Department of Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Kirill M Popov
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| | - Natalia Y Kedishvili
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama 35294
| |
Collapse
|
20
|
Li KN, Jain P, He CH, Eun FC, Kang S, Tumbar T. Skin vasculature and hair follicle cross-talking associated with stem cell activation and tissue homeostasis. eLife 2019; 8:e45977. [PMID: 31343406 PMCID: PMC6684267 DOI: 10.7554/elife.45977] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/25/2019] [Indexed: 12/21/2022] Open
Abstract
Skin vasculature cross-talking with hair follicle stem cells (HFSCs) is poorly understood. Skin vasculature undergoes dramatic remodeling during adult mouse hair cycle. Specifically, a horizontal plexus under the secondary hair germ (HPuHG) transiently neighbors the HFSC activation zone during the quiescence phase (telogen). Increased density of HPuHG can be induced by reciprocal mutations in the epithelium (Runx1) and endothelium (Alk1) in adult mice, and is accompanied by prolonged HFSC quiescence and by delayed entry and progression into the hair growth phase (anagen). Suggestively, skin vasculature produces BMP4, a well-established HFSC quiescence-inducing factor, thus contributing to a proliferation-inhibitory environment near the HFSC. Conversely, the HFSC activator Runx1 regulates secreted proteins with previously demonstrated roles in vasculature remodeling. We suggest a working model in which coordinated remodeling and molecular cross-talking of the adult epithelial and endothelial skin compartments modulate timing of HFSC activation from quiescence for proper tissue homeostasis of adult skin.
Collapse
Affiliation(s)
- Kefei Nina Li
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Prachi Jain
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Catherine Hua He
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Flora Chae Eun
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Sangjo Kang
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| | - Tudorita Tumbar
- Molecular Biology and GeneticsCornell UniversityIthacaUnited States
| |
Collapse
|
21
|
Sasaki GH. Review of Human Hair Follicle Biology: Dynamics of Niches and Stem Cell Regulation for Possible Therapeutic Hair Stimulation for Plastic Surgeons. Aesthetic Plast Surg 2019; 43:253-266. [PMID: 30324295 DOI: 10.1007/s00266-018-1248-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Plastic surgeons are frequently asked to manage male- and female-pattern hair loss in their practice. This article discusses the epidemiology, pathophysiology, and current management of androgenetic alopecia and emphasizes more recent knowledge of stem cell niches in hair follicles that drive hair cycling, alopecia, and its treatment. The many treatment programs available for hair loss include newer strategies that involve the usage of growth factors, platelet-rich plasma, and fat to stimulate follicle growth. Future research may clarify novel biomolecular mechanisms that target specific cells that promote hair regeneration.Level of Evidence V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
22
|
Abstract
Regulatory T cells (Tregs) are emerging as an essential stem cell niche component that promotes wound repair in adipose, muscle, and lung tissues. Recently in Cell, Ali et al. (2017) report that skin resident Tregs facilitate the proliferation and differentiation of hair follicle stem cells through Notch signaling.
Collapse
|
23
|
Tian Z, Zhao Q, Biswas S, Deng W. Methods of reactivation and reprogramming of neural stem cells for neural repair. Methods 2017; 133:3-20. [PMID: 28864354 DOI: 10.1016/j.ymeth.2017.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/21/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022] Open
Abstract
Research on the biology of adult neural stem cells (NSCs) and induced NSCs (iNSCs), as well as NSC-based therapies for diseases in central nervous system (CNS) has started to generate the expectation that these cells may be used for treatments in CNS injuries or disorders. Recent technological progresses in both NSCs themselves and their derivatives have brought us closer to therapeutic applications. Adult neurogenesis presents in particular regions in mammal brain, known as neurogenic niches such as the dental gyrus (DG) in hippocampus and the subventricular zone (SVZ), within which adult NSCs usually stay for long periods out of the cell cycle, in G0. The reactivation of quiescent adult NSCs needs orchestrated interactions between the extrinsic stimulis from niches and the intrinsic factors involving transcription factors (TFs), signaling pathway, epigenetics, and metabolism to start an intracellular regulatory program, which promotes the quiescent NSCs exit G0 and reenter cell cycle. Extrinsic and intrinsic mechanisms that regulate adult NSCs are interconnected and feedback on one another. Since endogenous neurogenesis only happens in restricted regions and steadily fails with disease advances, interest has evolved to apply the iNSCs converted from somatic cells to treat CNS disorders, as is also promising and preferable. To overcome the limitation of viral-based reprogramming of iNSCs, bioactive small molecules (SM) have been explored to enhance the efficiency of iNSC reprogramming or even replace TFs, making the iNSCs more amenable to clinical application. Despite intense research efforts to translate the studies of adult and induced NSCs from the bench to bedside, vital troubles remain at several steps in these processes. In this review, we examine the present status, advancement, pitfalls, and potential of the two types of NSC technologies, focusing on each aspects of reactivation of quiescent adult NSC and reprogramming of iNSC from somatic cells, as well as on progresses in cell-based regenerative strategies for neural repair and criteria for successful therapeutic applications.
Collapse
Affiliation(s)
- Zuojun Tian
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China; Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Qiuge Zhao
- Department of Neurology, The Institute of Guangzhou Respiratory Disease, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, PR China
| | - Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA 95817, USA; Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA.
| |
Collapse
|
24
|
Hatzfeld M, Keil R, Magin TM. Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics. Cold Spring Harb Perspect Biol 2017; 9:a029157. [PMID: 28096266 PMCID: PMC5453391 DOI: 10.1101/cshperspect.a029157] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Adherens junctions (AJs) and desmosomes connect the actin and keratin filament networks of adjacent cells into a mechanical unit. Whereas AJs function in mechanosensing and in transducing mechanical forces between the plasma membrane and the actomyosin cytoskeleton, desmosomes and intermediate filaments (IFs) provide mechanical stability required to maintain tissue architecture and integrity when the tissues are exposed to mechanical stress. Desmosomes are essential for stable intercellular cohesion, whereas keratins determine cell mechanics but are not involved in generating tension. Here, we summarize the current knowledge of the role of IFs and desmosomes in tissue mechanics and discuss whether the desmosome-keratin scaffold might be actively involved in mechanosensing and in the conversion of chemical signals into mechanical strength.
Collapse
Affiliation(s)
- Mechthild Hatzfeld
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - René Keil
- Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg, 06114 Halle, Germany
| | - Thomas M Magin
- Institute of Biology, Division of Cell and Developmental Biology and Saxonian Incubator for Clinical Translation (SIKT), University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
25
|
Kostic L, Sedov E, Soteriou D, Yosefzon Y, Fuchs Y. Isolation of Stem Cells and Progenitors from Mouse Epidermis. ACTA ACUST UNITED AC 2017; 41:1C.20.1-1C.20.11. [DOI: 10.1002/cpsc.26] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lana Kostic
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology and Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology Haifa Israel
| | - Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology and Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology Haifa Israel
| | - Despina Soteriou
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology and Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology Haifa Israel
| | - Yahav Yosefzon
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology and Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology Haifa Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology and Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology Haifa Israel
| |
Collapse
|
26
|
Oulhen N, Swartz SZ, Laird J, Mascaro A, Wessel GM. Transient translational quiescence in primordial germ cells. Development 2017; 144:1201-1210. [PMID: 28235822 PMCID: PMC5399625 DOI: 10.1242/dev.144170] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 02/01/2017] [Indexed: 01/07/2023]
Abstract
Stem cells in animals often exhibit a slow cell cycle and/or low transcriptional activity referred to as quiescence. Here, we report that the translational activity in the primordial germ cells (PGCs) of the sea urchin embryo (Strongylocentrotus purpuratus) is quiescent. We measured new protein synthesis with O-propargyl-puromycin and L-homopropargylglycine Click-iT technologies, and determined that these cells synthesize protein at only 6% the level of their adjacent somatic cells. Knockdown of translation of the RNA-binding protein Nanos2 by morpholino antisense oligonucleotides, or knockout of the Nanos2 gene by CRISPR/Cas9 resulted in a significant, but partial, increase (47%) in general translation specifically in the PGCs. We found that the mRNA of the translation factor eEF1A is excluded from the PGCs in a Nanos2-dependent manner, a consequence of a Nanos/Pumilio response element (PRE) in its 3'UTR. In addition to eEF1A, the cytoplasmic pH of the PGCs appears to repress translation and simply increasing the pH also significantly restores translation selectively in the PGCs. We conclude that the PGCs of this sea urchin institute parallel pathways to quiesce translation thoroughly but transiently.
Collapse
Affiliation(s)
- Nathalie Oulhen
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - S Zachary Swartz
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Whitehead Institute for Biomedical Research, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Jessica Laird
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Alexandra Mascaro
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Gary M Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| |
Collapse
|
27
|
|
28
|
Rivera-Gonzalez GC, Shook BA, Andrae J, Holtrup B, Bollag K, Betsholtz C, Rodeheffer MS, Horsley V. Skin Adipocyte Stem Cell Self-Renewal Is Regulated by a PDGFA/AKT-Signaling Axis. Cell Stem Cell 2016; 19:738-751. [PMID: 27746098 DOI: 10.1016/j.stem.2016.09.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/14/2016] [Accepted: 09/11/2016] [Indexed: 12/15/2022]
Abstract
Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.
Collapse
Affiliation(s)
| | - Brett A Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Brandon Holtrup
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Katherine Bollag
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Matthew S Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University, New Haven, CT 06520, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Yang GN, Kopecki Z, Cowin AJ. Role of Actin Cytoskeleton in the Regulation of Epithelial Cutaneous Stem Cells. Stem Cells Dev 2016; 25:749-59. [PMID: 27021878 DOI: 10.1089/scd.2016.0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cutaneous stem cells (CSCs) orchestrate the homeostasis and regeneration of mammalian skin. Epithelial CSCs have been isolated and characterized from the skin and hold great potential for tissue engineering and clinical applications. The actin cytoskeleton is known to regulate cell adhesion and motility through its intricate participation in signal transduction and structural modifications. The dynamics of actin cytoskeleton can directly influence CSCs behaviors including tissue morphogenesis, homeostasis, niche maintenance, activation, and wound repair. Various regulators of the actin cytoskeleton including kinases, actin-remodeling proteins, paracrine signals, and micro-RNAs collaborate and contribute to epithelial CSC proliferation, adhesion, and differentiation. This review brings together the latest mechanistic insights into how the actin cytoskeleton participates in the regulation of epithelial CSCs during development, homeostasis, and wound repair.
Collapse
Affiliation(s)
- Gink N Yang
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| |
Collapse
|
30
|
Soteriou D, Kostic L, Sedov E, Yosefzon Y, Steller H, Fuchs Y. Isolating Hair Follicle Stem Cells and Epidermal Keratinocytes from Dorsal Mouse Skin. J Vis Exp 2016. [PMID: 27168117 DOI: 10.3791/53931] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The hair follicle (HF) is an ideal system for studying the biology and regulation of adult stem cells (SCs). This dynamic mini organ is replenished by distinct pools of SCs, which are located in the permanent portion of the HF, a region known as the bulge. These multipotent bulge SCs were initially identified as slow cycling label retaining cells; however, their isolation has been made feasible after identification of specific cell markers, such as CD34 and keratin 15 (K15). Here, we describe a robust method for isolating bulge SCs and epidermal keratinocytes from mouse HFs utilizing fluorescence activated cell-sorting (FACS) technology. Isolated hair follicle SCs (HFSCs) can be utilized in various in vivo grafting models and are a valuable in vitro model for studying the mechanisms that govern multipotency, quiescence and activation.
Collapse
Affiliation(s)
- Despina Soteriou
- Department of Biology and Lokey Center, Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology
| | - Lana Kostic
- Department of Biology and Lokey Center, Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology
| | - Egor Sedov
- Department of Biology and Lokey Center, Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology
| | - Yahav Yosefzon
- Department of Biology and Lokey Center, Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology
| | - Hermann Steller
- Strang Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University;
| | - Yaron Fuchs
- Department of Biology and Lokey Center, Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology;
| |
Collapse
|
31
|
Shirokova V, Biggs LC, Jussila M, Ohyama T, Groves AK, Mikkola ML. Foxi3 Deficiency Compromises Hair Follicle Stem Cell Specification and Activation. Stem Cells 2016; 34:1896-908. [PMID: 26992132 DOI: 10.1002/stem.2363] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 02/27/2016] [Indexed: 01/16/2023]
Abstract
The hair follicle is an ideal system to study stem cell specification and homeostasis due to its well characterized morphogenesis and stereotypic cycles of stem cell activation upon each hair cycle to produce a new hair shaft. The adult hair follicle stem cell niche consists of two distinct populations, the bulge and the more activation-prone secondary hair germ (HG). Hair follicle stem cells are set aside during early stages of morphogenesis. This process is known to depend on the Sox9 transcription factor, but otherwise the establishment of the hair follicle stem cell niche is poorly understood. Here, we show that that mutation of Foxi3, a Forkhead family transcription factor mutated in several hairless dog breeds, compromises stem cell specification. Further, loss of Foxi3 impedes hair follicle downgrowth and progression of the hair cycle. Genome-wide profiling revealed a number of downstream effectors of Foxi3 including transcription factors with a recognized function in hair follicle stem cells such as Lhx2, Runx1, and Nfatc1, suggesting that the Foxi3 mutant phenotype results from simultaneous downregulation of several stem cell signature genes. We show that Foxi3 displays a highly dynamic expression pattern during hair morphogenesis and cycling, and identify Foxi3 as a novel secondary HG marker. Absence of Foxi3 results in poor hair regeneration upon hair plucking, and a sparse fur phenotype in unperturbed mice that exacerbates with age, caused by impaired secondary HG activation leading to progressive depletion of stem cells. Thus, Foxi3 regulates multiple aspects of hair follicle development and homeostasis. Stem Cells 2016;34:1896-1908.
Collapse
Affiliation(s)
- Vera Shirokova
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Leah C Biggs
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maria Jussila
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Takahiro Ohyama
- Department of Otolaryngology - Head & Neck Surgery and Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew K Groves
- Program in Developmental Biology, Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Marja L Mikkola
- Research Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Welle MM, Wiener DJ. The Hair Follicle: A Comparative Review of Canine Hair Follicle Anatomy and Physiology. Toxicol Pathol 2016; 44:564-74. [PMID: 27000375 DOI: 10.1177/0192623316631843] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The hair follicle (HF) has a wide range of functions including thermoregulation, physical and immunological protection against external insults, sensory perception, social interactions, and camouflage. One of the most characteristic features of HFs is that they self-renew during hair cycle (HC) throughout the entire life of an individual to continuously produce new hair. HC disturbances are common in humans and comparable to some alopecic disorders in dogs. A normal HC is maintained by follicular stem cells (SCs), which are predominately found in an area known as the bulge. Due to similar morphological characteristics of the human and canine bulge area, the particularity of compound HFs in humans and dogs as well as similarities in follicular biomarker expression, the dog might be a promising model to study human HC and SC disorders. In this review, we give an overview of normal follicular anatomy, the HC, and follicular SCs and discuss the possible pathogenetic mechanisms of noninflammatory alopecia.
Collapse
Affiliation(s)
- Monika M Welle
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Dominique J Wiener
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, DermFocus, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
33
|
Ojeh N, Pastar I, Tomic-Canic M, Stojadinovic O. Stem Cells in Skin Regeneration, Wound Healing, and Their Clinical Applications. Int J Mol Sci 2015; 16:25476-501. [PMID: 26512657 PMCID: PMC4632811 DOI: 10.3390/ijms161025476] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 12/18/2022] Open
Abstract
The skin is the largest organ of the body and has an array of functions. Skin compartments, epidermis, and hair follicles house stem cells that are indispensable for skin homeostasis and regeneration. These stem cells also contribute to wound repair, resulting in restoration of tissue integrity and function of damaged tissue. Unsuccessful wound healing processes often lead to non-healing wounds. Chronic wounds are caused by depletion of stem cells and a variety of other cellular and molecular mechanisms, many of which are still poorly understood. Current chronic wound therapies are limited, so the search to develop better therapeutic strategies is ongoing. Adult stem cells are gaining recognition as potential candidates for numerous skin pathologies. In this review, we will discuss epidermal and other stem cells present in the skin, and highlight some of the therapeutic applications of epidermal stem cells and other adult stem cells as tools for cell/scaffold-based therapies for non-healing wounds and other skin disorders. We will also discuss emerging concepts and offer some perspectives on how skin tissue-engineered products can be optimized to provide efficacious therapy in cutaneous repair and regeneration.
Collapse
Affiliation(s)
- Nkemcho Ojeh
- Faculty of Medical Sciences, the University of the West Indies, Cave Hill Campus, P.O. Box 64, Bridgetown BB 11000, St. Michael, Barbados; E-Mail:
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller Medical School, 1600 NW 10th Avenue, RMSB, Room 2023-A, Miami, FL 33136, USA; E-Mails: (I.P.); (M.T.-C.)
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller Medical School, 1600 NW 10th Avenue, RMSB, Room 2023-A, Miami, FL 33136, USA; E-Mails: (I.P.); (M.T.-C.)
| | - Olivera Stojadinovic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller Medical School, 1600 NW 10th Avenue, RMSB, Room 2023-A, Miami, FL 33136, USA; E-Mails: (I.P.); (M.T.-C.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-305-243-7295; Fax: +1-305-243-6191
| |
Collapse
|
34
|
Lacina L, Plzak J, Kodet O, Szabo P, Chovanec M, Dvorankova B, Smetana K. Cancer Microenvironment: What Can We Learn from the Stem Cell Niche. Int J Mol Sci 2015; 16:24094-110. [PMID: 26473842 PMCID: PMC4632740 DOI: 10.3390/ijms161024094] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
Epidermal stem cells (ESCs) are crucial for maintenance and self- renewal of skin epithelium and also for regular hair cycling. Their role in wound healing is also indispensable. ESCs reside in a defined outer root sheath portion of hair follicle—also known as the bulge region. ECS are also found between basal cells of the interfollicular epidermis or mucous membranes. The non-epithelial elements such as mesenchymal stem cell-like elements of dermis or surrounding adipose tissue can also contribute to this niche formation. Cancer stem cells (CSCs) participate in formation of common epithelial malignant diseases such as basal cell or squamous cell carcinoma. In this review article, we focus on the role of cancer microenvironment with emphasis on the effect of cancer-associated fibroblasts (CAFs). This model reflects various biological aspects of interaction between cancer cell and CAFs with multiple parallels to interaction of normal epidermal stem cells and their niche. The complexity of intercellular interactions within tumor stroma is depicted on example of malignant melanoma, where keratinocytes also contribute the microenvironmental landscape during early phase of tumor progression. Interactions seen in normal bulge region can therefore be an important source of information for proper understanding to melanoma. The therapeutic consequences of targeting of microenvironment in anticancer therapy and for improved wound healing are included to article.
Collapse
Affiliation(s)
- Lukas Lacina
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, U Nemocnice 3, 12800 Prague 2, Czech Republic.
- Department of Dermatology and Venereology, 1st Faculty of Medicine and General University Hospital, Charles University, U Nemocnice 2, 12808 Prague 2, Czech Republic.
| | - Jan Plzak
- Department of Otorhinolaryngology and Head and Neck Surgery, 1st Faculty of Medicine and University Hospital Motol, Charles University, V Úvalu 84, 15006 Prague 5, Czech Republic.
| | - Ondrej Kodet
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, U Nemocnice 3, 12800 Prague 2, Czech Republic.
- Department of Dermatology and Venereology, 1st Faculty of Medicine and General University Hospital, Charles University, U Nemocnice 2, 12808 Prague 2, Czech Republic.
| | - Pavol Szabo
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, U Nemocnice 3, 12800 Prague 2, Czech Republic.
| | - Martin Chovanec
- Department of Otorhinolaryngology and Head and Neck Surgery, 1st Faculty of Medicine and University Hospital Motol, Charles University, V Úvalu 84, 15006 Prague 5, Czech Republic.
| | - Barbora Dvorankova
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, U Nemocnice 3, 12800 Prague 2, Czech Republic.
| | - Karel Smetana
- Institute of Anatomy, 1st Faculty of Medicine, Charles University, U Nemocnice 3, 12800 Prague 2, Czech Republic.
| |
Collapse
|
35
|
|
36
|
Barker A. Skin structure. Plast Reconstr Surg 2015. [DOI: 10.1002/9781118655412.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
37
|
Peters F, Vorhagen S, Brodesser S, Jakobshagen K, Brüning JC, Niessen CM, Krönke M. Ceramide synthase 4 regulates stem cell homeostasis and hair follicle cycling. J Invest Dermatol 2015; 135:1501-1509. [PMID: 25705848 DOI: 10.1038/jid.2015.60] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 01/28/2015] [Accepted: 02/08/2015] [Indexed: 12/16/2022]
Abstract
Ceramides are crucial for skin barrier function, but little is known about the regulation of epidermal appendages and whether stem cell populations that control their regeneration depend on specific ceramide species. Here we demonstrate that ceramide synthase 4 (CerS4) is highly expressed in the epidermis of adult mice where it is localized in the interfollicular epidermis and defined populations within the pilosebaceous unit. Inactivation of CerS4 in mice resulted in precocious activation of hair follicle bulge stem cells while expanding the Lrig1(+) junctional zone and sebaceous glands. This was preceded first by a decrease in bone morphogenetic protein (BMP) and a subsequent increase in Wnt signaling. This imbalance in quiescent versus activating signals likely promoted a prolonged anagen-like hair follicle state after the second catagen, which exhausted stem cells over time ultimately resulting in hair loss in aged mice. K14-Cre-mediated deletion of CerS4 revealed a similar phenotype, thus suggesting an epidermis intrinsic function of CerS4 in regulating the regeneration of the pilosebaceous unit. The data indicate that CerS4-directed epidermal ceramide composition is essential to control hair follicle stem and progenitor cell behavior potentially through its regulation of BMP and Wnt signaling.
Collapse
Affiliation(s)
- Franziska Peters
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Susanne Vorhagen
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany; Department of Dermatology, University of Cologne, Cologne, Germany
| | - Susanne Brodesser
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany
| | - Kristin Jakobshagen
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
| | - Jens C Brüning
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Cologne, Germany; Institute for Genetics, University of Cologne, Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine, University of Cologne, Cologne, Germany; Max Planck Institute for Neurological Research, Cologne, Germany
| | - Carien M Niessen
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Cologne, Germany; Department of Dermatology, University of Cologne, Cologne, Germany.
| | - Martin Krönke
- Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Cologne, Germany.
| |
Collapse
|
38
|
Larcher F, Espada J, Díaz-Ley B, Jaén P, Juarranz A, Quintanilla M. New Experimental Models of Skin Homeostasis and Diseases. ACTAS DERMO-SIFILIOGRAFICAS 2015. [DOI: 10.1016/j.adengl.2014.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Abstract
The epidermis and associated appendages of the skin represent a multi-lineage tissue that is maintained by perpetual rounds of renewal. During homeostasis, turnover of epidermal lineages is achieved by input from regionalized keratinocytes stem or progenitor populations with little overlap from neighboring niches. Over the last decade, molecular markers selectively expressed by a number of these stem or progenitor pools have been identified, allowing for the isolation and functional assessment of stem cells and genetic lineage tracing analysis within intact skin. These advancements have led to many fundamental observations about epidermal stem cell function such as the identification of their progeny, their role in maintenance of skin homeostasis, or their contribution to wound healing. In this chapter, we provide a methodology to identify and isolate epidermal stem cells and to assess their functional role in their respective niche. Furthermore, recent evidence has shown that the microenvironment also plays a crucial role in stem cell function. Indeed, epidermal cells are under the influence of surrounding fibroblasts, adipocytes, and sensory neurons that provide extrinsic signals and mechanical cues to the niche and contribute to skin morphogenesis and homeostasis. A better understanding of these microenvironmental cues will help engineer in vitro experimental models with more relevance to in vivo skin biology. New approaches to address and study these environmental cues in vitro will also be addressed.
Collapse
Affiliation(s)
- Yanne S Doucet
- Department of Dermatology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | | |
Collapse
|
40
|
Urbán N, Guillemot F. Neurogenesis in the embryonic and adult brain: same regulators, different roles. Front Cell Neurosci 2014; 8:396. [PMID: 25505873 PMCID: PMC4245909 DOI: 10.3389/fncel.2014.00396] [Citation(s) in RCA: 352] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/05/2014] [Indexed: 12/12/2022] Open
Abstract
Neurogenesis persists in adult mammals in specific brain areas, known as neurogenic niches. Adult neurogenesis is highly dynamic and is modulated by multiple physiological stimuli and pathological states. There is a strong interest in understanding how this process is regulated, particularly since active neuronal production has been demonstrated in both the hippocampus and the subventricular zone (SVZ) of adult humans. The molecular mechanisms that control neurogenesis have been extensively studied during embryonic development. Therefore, we have a broad knowledge of the intrinsic factors and extracellular signaling pathways driving proliferation and differentiation of embryonic neural precursors. Many of these factors also play important roles during adult neurogenesis, but essential differences exist in the biological responses of neural precursors in the embryonic and adult contexts. Because adult neural stem cells (NSCs) are normally found in a quiescent state, regulatory pathways can affect adult neurogenesis in ways that have no clear counterpart during embryogenesis. BMP signaling, for instance, regulates NSC behavior both during embryonic and adult neurogenesis. However, this pathway maintains stem cell proliferation in the embryo, while it promotes quiescence to prevent stem cell exhaustion in the adult brain. In this review, we will compare and contrast the functions of transcription factors (TFs) and other regulatory molecules in the embryonic brain and in adult neurogenic regions of the adult brain in the mouse, with a special focus on the hippocampal niche and on the regulation of the balance between quiescence and activation of adult NSCs in this region.
Collapse
Affiliation(s)
- Noelia Urbán
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| | - François Guillemot
- Department of Molecular Neurobiology, MRC National Institute for Medical Research London, UK
| |
Collapse
|
41
|
A transcriptional mechanism integrating inputs from extracellular signals to activate hippocampal stem cells. Neuron 2014; 83:1085-97. [PMID: 25189209 PMCID: PMC4157576 DOI: 10.1016/j.neuron.2014.08.004] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2014] [Indexed: 12/27/2022]
Abstract
The activity of adult stem cells is regulated by signals emanating from the surrounding tissue. Many niche signals have been identified, but it is unclear how they influence the choice of stem cells to remain quiescent or divide. Here we show that when stem cells of the adult hippocampus receive activating signals, they first induce the expression of the transcription factor Ascl1 and only subsequently exit quiescence. Moreover, lowering Ascl1 expression reduces the proliferation rate of hippocampal stem cells, and inactivating Ascl1 blocks quiescence exit completely, rendering them unresponsive to activating stimuli. Ascl1 promotes the proliferation of hippocampal stem cells by directly regulating the expression of cell-cycle regulatory genes. Ascl1 is similarly required for stem cell activation in the adult subventricular zone. Our results support a model whereby Ascl1 integrates inputs from both stimulatory and inhibitory signals and converts them into a transcriptional program activating adult neural stem cells.
Collapse
|
42
|
Dahlhoff M, de Angelis MH, Wolf E, Schneider MR. Ligand-independent epidermal growth factor receptor hyperactivation increases sebaceous gland size and sebum secretion in mice. Exp Dermatol 2014; 22:667-9. [PMID: 24079739 DOI: 10.1111/exd.12219] [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] [Accepted: 08/05/2013] [Indexed: 11/29/2022]
Abstract
The epidermal growth factor receptor (EGFR) system is an established regulator of the development and homeostasis of the hair follicle and interfollicular epidermis. Here, we evaluated EGFR actions on the sebaceous glands (SGs) by employing Dsk5 mice, a mutant line in which the EGFR is constitutively activated in a ligand-independent manner. Compared to control littermates, Dsk5 mice showed increased sebum levels and enlarged SGs, which contained a higher number of cells and showed stronger proliferation. c-myc transcript levels were increased in Dsk5 skin, suggesting that c-myc mediates the proliferative stimuli of the EGFR in the SG. Analysis of differentiation markers revealed deregulated expression of Scd1 and Scd3, indicating that sebaceous lipogenesis is affected in Dsk5 mice. In conclusion, our study indicates that the EGFR is an important regulator of presebocyte proliferation, contributing to the final cell number, to the size and to the lipid output of SGs.
Collapse
Affiliation(s)
- Maik Dahlhoff
- Gene Center, Institute of Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), LMU Munich, Munich, Germany
| | | | | | | |
Collapse
|
43
|
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: 2.3] [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
|
44
|
Larcher F, Espada J, Díaz-Ley B, Jaén P, Juarranz A, Quintanilla M. New experimental models of skin homeostasis and diseases. ACTAS DERMO-SIFILIOGRAFICAS 2014; 106:17-28. [PMID: 24878038 DOI: 10.1016/j.ad.2014.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 02/25/2014] [Accepted: 03/03/2014] [Indexed: 12/19/2022] Open
Abstract
Homeostasis, whose regulation at the molecular level is still poorly understood, is intimately related to the functions of epidermal stem cells. Five research groups have been brought together to work on new in vitro and in vivo skin models through the SkinModel-CM program, under the auspices of the Spanish Autonomous Community of Madrid. This project aims to analyze the functions of DNA methyltransferase 1, endoglin, and podoplanin in epidermal stem cell activity, homeostasis, and skin cancer. These new models include 3-dimensional organotypic cultures, immunodeficient skin-humanized mice, and genetically modified mice. Another aim of the program is to use skin-humanized mice to model dermatoses such as Gorlin syndrome and xeroderma pigmentosum in order to optimize new protocols for photodynamic therapy.
Collapse
Affiliation(s)
- F Larcher
- Unidad de Medicina Regenerativa, Departamento de Investigación Básica, División de Biomedicina Epitelial, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) y Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, España
| | - J Espada
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), Madrid, España; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-UAM, Madrid, España
| | - B Díaz-Ley
- Unidad de Dermatología, Hospital Ramón y Cajal, Madrid, España
| | - P Jaén
- Unidad de Dermatología, Hospital Ramón y Cajal, Madrid, España
| | - A Juarranz
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid (UAM), Madrid, España.
| | - M Quintanilla
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC)-UAM, Madrid, España
| |
Collapse
|
45
|
Cirio MC, de Groh ED, de Caestecker MP, Davidson AJ, Hukriede NA. Kidney regeneration: common themes from the embryo to the adult. Pediatr Nephrol 2014; 29:553-64. [PMID: 24005792 PMCID: PMC3944192 DOI: 10.1007/s00467-013-2597-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 02/07/2023]
Abstract
The vertebrate kidney has an inherent ability to regenerate following acute damage. Successful regeneration of the injured kidney requires the rapid replacement of damaged tubular epithelial cells and reconstitution of normal tubular function. Identifying the cells that participate in the regeneration process as well as the molecular mechanisms involved may reveal therapeutic targets for the treatment of kidney disease. Renal regeneration is associated with the expression of genetic pathways that are necessary for kidney organogenesis, suggesting that the regenerating tubular epithelium may be "reprogrammed" to a less-differentiated, progenitor state. This review will highlight data from various vertebrate models supporting the hypothesis that nephrogenic genes are reactivated as part of the process of kidney regeneration following acute kidney injury (AKI). Emphasis will be placed on the reactivation of developmental pathways and how our understanding of the resulting regeneration process may be enhanced by lessons learned in the embryonic kidney.
Collapse
Affiliation(s)
- M. Cecilia Cirio
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Eric D. de Groh
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Mark P. de Caestecker
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Neil A. Hukriede
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
46
|
Aldehyde dehydrogenase 1 expression in basal cell carcinoma, actinic keratosis and Bowen's disease. Mol Clin Oncol 2014; 1:621-624. [PMID: 24649218 PMCID: PMC3916212 DOI: 10.3892/mco.2013.106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 04/04/2013] [Indexed: 01/16/2023] Open
Abstract
Aldehyde dehydrogenase (ALDH) is an enzyme responsible for oxidizing aldehydes to carbonic acids. ALDH1 is an isoform that is thought to be a stem cell marker as it is highly expressed in the stem cells of various organs. However, its expression in basal cell carcinoma (BCC), actinic keratosis (AK) and Bowen’s disease (BD) of the skin has not yet been analyzed. Twenty-five consecutive operative cases each of BCC, AK and BD, as well as 10 normal skin tissues were assessed for ALDH1 expression by immunohistochemistry. In normal skin, ALDH1 expression was observed in the suprabasal cells of the follicular infundibulum, inner cells of the outer root sheath and sebocytes. BCC cases (88%) showed no or focal-positive immunoreactivity for ALDH1. Focal immunopositivity for ALDH1 was observed in 44% of AK cases, while the remaining cases were ALDH1-negative. By contrast, diffuse positive immunoreactivity for ALDH1 was observed in 64% of BD cases. Differential expression patterns of ALDH1 in AK and BD may reflect the distinct cells of origin of these two conditions. Moreover, a low ALDH1 expression in BCC may also reflect the possible origin of BCC, the basal cells of the outer root sheath.
Collapse
|
47
|
Waghmare SK, Tumbar T. Adult hair follicle stem cells do not retain the older DNA strands in vivo during normal tissue homeostasis. Chromosome Res 2014; 21:203-12. [PMID: 23681654 DOI: 10.1007/s10577-013-9355-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tissue stem cells have been proposed to segregate the chromosomes asymmetrically (in a non-random manner), thereby retaining preferentially the older "immortal" DNA strands bearing the stemness characteristics into one daughter cell, whereas the newly synthesized strands are segregated to the other daughter cell that will commit to differentiation. Moreover, this non-random segregation would protect the stem cell genome from accumulating multiple mutations during repeated DNA replication. This long-standing hypothesis remains an active subject of study due to conflicting results for some systems and lack of consistency among different tissue stem cell populations. In this review, we will focus on work done in the hair follicle, which is one of the best-understood vertebrate tissue stem cell system to date. In cell culture analysis of paired cultured keratinocytes derived from hair follicle, stem cells suggested a non-random segregation of chromosome with respect to the older DNA strand. In vivo, the hair follicle stem cells appear to self-renew and differentiate at different phases of their homeostatic cycle. The fate decisions occur in quiescence when some stem cells migrate out of their niche and commit to differentiation without self-renewal. The stem cells left behind in the niche self-renew symmetrically and randomly segregate the chromosomes at each division, making more stem cells. This model seems to apply to at least a few other vertebrate tissue stem cells in vivo.
Collapse
Affiliation(s)
- Sanjeev K Waghmare
- Advanced Centre for Treatment, Research and Education in Cancer ACTREC, Tata Memorial Centre, Navi Mumbai, 410210, India.
| | | |
Collapse
|
48
|
Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta Gen Subj 2014; 1840:2506-19. [PMID: 24418517 PMCID: PMC4081568 DOI: 10.1016/j.bbagen.2014.01.010] [Citation(s) in RCA: 897] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 02/08/2023]
Abstract
Background Extracellular matrix (ECM) is a dynamic and complex environment characterized by biophysical, mechanical and biochemical properties specific for each tissue and able to regulate cell behavior. Stem cells have a key role in the maintenance and regeneration of tissues and they are located in a specific microenvironment, defined as niche. Scope of review We overview the progresses that have been made in elucidating stem cell niches and discuss the mechanisms by which ECM affects stem cell behavior. We also summarize the current tools and experimental models for studying ECM–stem cell interactions. Major conclusions ECM represents an essential player in stem cell niche, since it can directly or indirectly modulate the maintenance, proliferation, self-renewal and differentiation of stem cells. Several ECM molecules play regulatory functions for different types of stem cells, and based on its molecular composition the ECM can be deposited and finely tuned for providing the most appropriate niche for stem cells in the various tissues. Engineered biomaterials able to mimic the in vivo characteristics of stem cell niche provide suitable in vitro tools for dissecting the different roles exerted by the ECM and its molecular components on stem cell behavior. General significance ECM is a key component of stem cell niches and is involved in various aspects of stem cell behavior, thus having a major impact on tissue homeostasis and regeneration under physiological and pathological conditions. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties. Stem cells have a key role in the maintenance and regeneration of tissues. The extracellular matrix is a critical regulator of stem cell function. Stem cells reside in a dynamic and specialized microenvironment denoted as niche. The extracellular matrix represents an essential component of stem cell niches. Bioengineered niches can be used for investigating stem cell–matrix interactions.
Collapse
Affiliation(s)
- Francesca Gattazzo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Anna Urciuolo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
49
|
Carrasco E, Calvo MI, Espada J. DNA labeling in vivo: quantification of epidermal stem cell chromatin content in whole mouse hair follicles using Fiji image processing software. Methods Mol Biol 2014; 1094:79-88. [PMID: 24162981 DOI: 10.1007/978-1-62703-706-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
DNA labeling in vivo using nucleoside analogues is a current experimental approach to determine cell proliferation rates in cell cultures and tissues. It has also been successfully used to localize adult stem cell niches through the identification of nucleoside label-retaining cells (LRC) in long-term experiments. A major hindrance of this methodology relies on the selection of adequate procedures to quantify the nucleoside analogue content from image data files. Here we propose a simple procedure using Fiji image processing software to accurately calculate nucleoside analogue retaining chromatin/total chromatin (LRC/DAPI) signal ratios in the well-known mouse hair follicle stem cell niche.
Collapse
Affiliation(s)
- Elisa Carrasco
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | |
Collapse
|
50
|
Beyond expectations: novel insights into epidermal keratin function and regulation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 311:265-306. [PMID: 24952920 DOI: 10.1016/b978-0-12-800179-0.00007-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The epidermis is a stratified epithelium that relies on its cytoskeleton and cell junctions to protect the body against mechanical injury, dehydration, and infections. Keratin intermediate filament proteins are involved in many of these functions by forming cell-specific cytoskeletal scaffolds crucial for the maintenance of cell and tissue integrity. In response to various stresses, the expression and organization of keratins are altered at transcriptional and posttranslational levels to restore tissue homeostasis. Failure to restore tissue homeostasis in the presence of keratin gene mutations results in acute and chronic skin disorders for which currently no rational therapies are available. Here, we review the recent progress on the role of keratins in cytoarchitecture, adhesion, signaling, and inflammation. By focusing on epidermal keratins, we illustrate the contribution of keratin isotypes to differentiated epithelial functions.
Collapse
|