Volume 15, Issue 6, Pages (May 2016)

Slides:



Advertisements
Similar presentations
Volume 8, Issue 2, Pages (February 2011)
Advertisements

Volume 8, Issue 5, Pages (May 2011)
Keratinocyte Stem Cells but Not Melanocyte Stem Cells Are the Primary Target for Radiation-Induced Hair Graying  Hitomi Aoki, Akira Hara, Tsutomu Motohashi,
Wnt/β-Catenin and Kit Signaling Sequentially Regulate Melanocyte Stem Cell Differentiation in UVB-Induced Epidermal Pigmentation  Takaaki Yamada, Seiji.
Activated Kras Alters Epidermal Homeostasis of Mouse Skin, Resulting in Redundant Skin and Defective Hair Cycling  Anandaroop Mukhopadhyay, Suguna R.
C-Myc activation in transgenic mouse epidermis results in mobilization of stem cells and differentiation of their progeny  Isabel Arnold, Fiona M Watt 
Gregory D. Rak, Lisa C. Osborne, Mark C. Siracusa, Brian S
Chih-Chiang Chen, Philip J. Murray, Ting Xin Jiang, Maksim V
Volume 13, Issue 4, Pages (October 2013)
Volume 105, Issue 4, Pages (May 2001)
Volume 15, Issue 8, Pages (May 2016)
Melanoblasts' Proper Location and Timed Differentiation Depend on Notch/RBP-J Signaling in Postnatal Hair Follicles  Geneviève Aubin-Houzelstein, Johanna.
Adenovirus-Mediated Wnt10b Overexpression Induces Hair Follicle Regeneration  Yu-Hong Li, Kun Zhang, Ke Yang, Ji-Xing Ye, Yi-Zhan Xing, Hai-Ying Guo, Fang.
Volume 29, Issue 1, Pages (April 2014)
Ectodysplasin A Pathway Contributes to Human and Murine Skin Repair
Kai Kretzschmar, Denny L. Cottle, Pawel J. Schweiger, Fiona M. Watt 
Volume 131, Issue 4, Pages (October 2006)
Volume 9, Issue 5, Pages (November 2017)
Protective Effect of Kit Signaling for Melanocyte Stem Cells against Radiation-Induced Genotoxic Stress  Hitomi Aoki, Akira Hara, Tsutomu Motohashi, Takahiro.
Volume 128, Issue 3, Pages (March 2005)
Molecular Therapy - Methods & Clinical Development
Approach for the Derivation of Melanocytes from Induced Pluripotent Stem Cells  Tamihiro Kawakami, Tatsuro Okano, Sora Takeuchi, Kayoko Osumi, Yoshinao.
Makoto Takeo, Christopher S. Hale, Mayumi Ito 
Dedicated Epithelial Recipient Cells Determine Pigmentation Patterns
Atypical Protein Kinase C Isoform, aPKCλ, Is Essential for Maintaining Hair Follicle Stem Cell Quiescence  Shin-Ichi Osada, Naoko Minematsu, Fumino Oda,
P21-Activated Kinase 4 Critically Regulates Melanogenesis via Activation of the CREB/MITF and β-Catenin/MITF Pathways  Cheong-Yong Yun, Soon-Tae You,
Impaired Skin Regeneration and Remodeling after Cutaneous Injury and Chemically Induced Hyperplasia in Taps-Transgenic Mice  Maike Hildenbrand, Verena.
Nicole Amberg, Martin Holcmann, Gabriel Stulnig, Maria Sibilia 
Skin-Specific Deletion of Mis18α Impedes Proliferation and Stratification of Epidermal Keratinocytes  Koog Chan Park, Minkyoung Lee, Yoon Jeon, Raok Jeon,
Peggy S. Myung, Makoto Takeo, Mayumi Ito, Radhika P. Atit 
Hosein Kouros-Mehr, Euan M. Slorach, Mark D. Sternlicht, Zena Werb 
Volume 134, Issue 2, Pages e3 (February 2008)
Roles of GasderminA3 in Catagen–Telogen Transition During Hair Cycling
Integrin β6-Deficient Mice Show Enhanced Keratinocyte Proliferation and Retarded Hair Follicle Regression after Depilation  Yanshuang Xie, Kevin J. McElwee,
Volume 22, Issue 4, Pages (January 2018)
Transcription Factor CTIP2 Maintains Hair Follicle Stem Cell Pool and Contributes to Altered Expression of LHX2 and NFATC1  Shreya Bhattacharya, Heather.
Mohammad Rashel, Ninche Alston, Soosan Ghazizadeh 
Volume 8, Issue 2, Pages (February 2011)
Roxanne Toivanen, Adithi Mohan, Michael M. Shen  Stem Cell Reports 
Fuz Controls the Morphogenesis and Differentiation of Hair Follicles through the Formation of Primary Cilia  Daisy Dai, Huiping Zhu, Bogdan Wlodarczyk,
Volume 10, Issue 5, Pages (May 2012)
Volume 3, Issue 2, Pages (February 2013)
The Neurofibromatosis Type 1 (Nf1) Tumor Suppressor is a Modifier of Carcinogen- Induced Pigmentation and Papilloma Formation in C57BL/6 Mice  Radhika.
Hitomi Aoki, Hiroyuki Tomita, Akira Hara, Takahiro Kunisada 
Epithelial Stem Cells: A Folliculocentric View
Slc1a3-CreER as a Targeting Tool for the K6+ Epithelial Stem Cell Niche and its Precursors during Mouse Hair Follicle Cycle  Aiko Sada, Prachi Jain, Sherry.
Secreted Frizzled-Related Protein 2 (sFRP2) Functions as a Melanogenic Stimulator; the Role of sFRP2 in UV-Induced Hyperpigmentary Disorders  Misun Kim,
Abdul Q. Sheikh, Janet K. Lighthouse, Daniel M. Greif  Cell Reports 
The Vitamin D Receptor Is Required for Mouse Hair Cycle Progression but not for Maintenance of the Epidermal Stem Cell Compartment  Héctor G. Pálmer,
Molecular Mechanisms Regulating Hair Follicle Development
Volume 25, Issue 11, Pages (November 2017)
SOX2 Is a Marker for Stem-like Tumor Cells in Bladder Cancer
Differential Effects of Neurofibromin Gene Dosage on Melanocyte Development  Mugdha Deo, Jenny Li-Ying Huang, Helmut Fuchs, Martin Hrabe de Angelis, Catherine.
Fate of Prominin-1 Expressing Dermal Papilla Cells during Homeostasis, Wound Healing and Wnt Activation  Grace S. Kaushal, Emanuel Rognoni, Beate M. Lichtenberger,
Transient Expression of WNT2 Promotes Somatic Cell Reprogramming by Inducing β- Catenin Nuclear Accumulation  Mizuki Kimura, May Nakajima-Koyama, Joonseong.
The Suppressor of Cytokine Signaling (SOCS)-3 Determines Keratinocyte Proliferative and Migratory Potential during Skin Repair  Andreas Linke, Itamar.
Volume 1, Issue 1, Pages (June 2013)
Epidermal Stem Cells in the Isthmus/Infundibulum Influence Hair Shaft Differentiation: Evidence from Targeted DLX3 Deletion  Jin-Chul Kim, Olivier Duverger,
Andreya Sharov, Desmond J. Tobin, Tatyana Y
Makoto Senoo, Filipa Pinto, Christopher P. Crum, Frank McKeon  Cell 
Volume 15, Issue 3, Pages (April 2016)
Delineating Immune-Mediated Mechanisms Underlying Hair Follicle Destruction in the Mouse Mutant Defolliculated  Fiona Ruge, Aikaterini Glavini, Awen M.
An Extended Epidermal Response Heals Cutaneous Wounds in the Absence of a Hair Follicle Stem Cell Contribution  Abigail K. Langton, Sarah E. Herrick,
Jaana Mannik, Kamil Alzayady, Soosan Ghazizadeh 
Jack Heath, Abigail K. Langton, Nigel L. Hammond, Paul A
Thrombospondin-1 Plays a Critical Role in the Induction of Hair Follicle Involution and Vascular Regression During the Catagen Phase  Kiichiro Yano, Michael.
Volume 18, Issue 4, Pages (April 2010)
B-Raf and C-Raf Are Required for Melanocyte Stem Cell Self-Maintenance
Loss of Keratin 10 Leads to Mitogen-activated Protein Kinase (MAPK) Activation, Increased Keratinocyte Turnover, and Decreased Tumor Formation in Mice 
P21-Activated Kinase 4 Critically Regulates Melanogenesis via Activation of the CREB/MITF and β-Catenin/MITF Pathways  Cheong-Yong Yun, Soon-Tae You,
Presentation transcript:

Volume 15, Issue 6, Pages 1291-1302 (May 2016) EdnrB Governs Regenerative Response of Melanocyte Stem Cells by Crosstalk with Wnt Signaling  Makoto Takeo, Wendy Lee, Piul Rabbani, Qi Sun, Hai Hu, Chae Ho Lim, Prashiela Manga, Mayumi Ito  Cell Reports  Volume 15, Issue 6, Pages 1291-1302 (May 2016) DOI: 10.1016/j.celrep.2016.04.006 Copyright © 2016 The Authors Terms and Conditions

Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 1 Loss of EdnrB Leads to Loss of Pigment, Reduced Proliferation, and Failure to Maintain McSCs (A) Gross appearance of control (left) and EdnrB cKO (right) mice at second telogen. (B) Bright-field whole-mount picture of hair shaft from control (left) and EdnrB cKO (right) mice. (C) Quantification of percentage of gray hair at each telogen. (D–F and H–J) Immunohistochemical analysis of bulb melanocytes in skin sections at anagen IV with Tomato and indicated melanocyte differentiation markers from Tyr-CreER;Rosa-stop-Tomato (control; D–F) and EdnrB cKO;Rosa-stop-Tomato mice (H–J). (G and K) Bright-field image of bulb from control (G) and EdnrB cKO mouse (K). (L) Quantification of the percentage of Tomato-positive cells that are also positive for Dct, MITF, and S100, respectively. (M and O) Immunohistochemistry for BrdU on control (M) and EdnrB cKO mice (O). (N and P) Anagen VI skin sections stained with Dct and cleaved caspase 3 (left panel) in hair follicle bulb in control (N) and EdnrB cKO mice (P). Single caspase 3 staining is shown in right panel. (Q and R) Quantification of the percentage of BrdU+/Tomato+ cells (Q) and cleaved caspase 3+/Dct+ cells (R). (S) Whole-mount image of X-gal-stained single hair follicles from Dct-LacZ (control; top) and unpigmented (middle) and pigmented hair follicles (bottom) from EdnrB cKO; Dct-LacZ mice. (T) Distribution and average (blue line) of absolute number of Dct-LacZ+ melanocytes per hair follicle. Dashed lines indicate border between hair follicle and dermis. Arrowheads indicate double positive cells for indicated markers. Data are presented as the mean ± SD. ∗p < 0.001; ∗∗p < 0.01; ∗∗∗p < 0.02; ∗∗∗∗p < 0.05. The scale bar represents 50 μm in (D)–(F) and (H)–(J) and 20 μm in (M), (N), and (S). See also Figure S1. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 2 Overexpression of Edn1 Promotes McSCs Differentiation and Proliferation during Anagen (A) Whole-mount X-gal staining from K14-rtTA; TetO-Edn1-LacZ mice 1 week after Dox treatment during telogen. Right panel is higher magnification of picture shown in left panel. (B and C) Whole-mount image of telogen (B) or anagen (C) hair follicle from control (left) and K14-rtTA; TetO-Edn1-LacZ (right) mice that were induced with Dox during telogen (B) or anagen (C). (D–M and O–X) Immunostaining of skin sections from control (D–H and O–S) and K14-rtTA; TetO-Edn1-LacZ (I–M and T–X) mice for Tyr and Dct (D–M) and Ki67 and Dct (O–X) in the McSC niche and bulb compartment of the hair follicle at indicated hair cycle stages. (N and Y) Quantification of the percentage of Tyr-positive cells (N) and Ki67-positive cells (Y) in Dct-positive cells. Dashed lines indicate border between hair follicle and dermis. Arrowheads indicate pigmentation in (C) and double positive cells for indicated markers in immunohistochemical analysis. Data are presented as the mean ± SD. ∗p < 0.002; ∗∗p < 0.05. The scale bar represents 100 μm in (A) and 20 μm in (B), (C), and (E). See also Figures S2 and S3. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 3 Overexpression of Edn1 Promotes Upward Migration of McSCs and Generation of Epidermal Melanocytes following Wounding (A–D) Whole-mount image of X-gal-stained wound area of Dct-LacZ (control; A and B) and Tyr-CreER; EdnrBfl/fl; Dct-lacZ (C and D) at indicated days after re-epithelialization. (E–J) Double immunohistochemical staining of Dct and Ki67 in the bulge (E and H), upper hair follicle (F and I), and inter-follicular epidermis (G and J) in control (E–G) and K14-rtTA; TetO-Edn1-LacZ (Edn1; H–J) mice. (K–N) Whole-mount analyses of wound site (K and L) and de novo hair follicles (M and N) within wound site from control (K and M) and Edn1 mice (L and N) at 8 days after re-epithelialization. (O–R) Quantification of the number of Dct-LacZ+ cells in wound site (O), the percentage of Ki67+/Dct+ cells (P), the number of pigmented cells in wound site (Q), and the percentage of pigmented de novo hair (R), respectively. Dashed lines indicate periphery of wound site in (A) and (D) and boundary between epidermis and dermis in (E)–(J). Arrowheads show Dct-LacZ+ cells in wound area in (A)–(D) and Ki67+/Dct+ cells (H)–(J). IFE, inter-follicular epidermis; UF, upper follicle. Data are presented as the mean ± SD. ∗p < 0.01; ∗∗p < 0.02; ∗∗∗p < 0.05. The scale bar represents 1 mm in (A), 50 μm in (E), 200 μm in (K) and (L), and 100 μm in (M) and (N). See also Figures S4–S6. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 4 Edn1 Promotes Generation of Epidermal Melanocytes in Human Scalp Tissue (A) Schematic figure of human scalp explant culture experiment. (B–F) Immunohistochemical analysis with K14 and MITF of sections of human scalp before ablation (B), after ablation of skin prior to culture (C), 8 days in culture in the absence (D) and presence of Edn1 (E), and presence of both Edn1 and BQ788 (F). (G and H) Quantification of the number of MITF+ cells in human scalp before culture (G) and after 8 days in culture (H). White dashed line indicates the border between inter-follicular epidermis and dermis. Red dashed line indicates the border between hair follicle epidermis and dermis. Arrowheads indicate MITF+ cells. Data are presented as the mean ± SD. ∗p < 0.05; ∗∗p < 0.005. The scale bar represents 100 μm. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 5 Edn1 Synergizes with Wnt Pathway to Enhance Proliferation and Migration of McSCs (A–H) Immunostaining of skin sections from control (A and C–E) and K14-rtTA; TetO-Edn1-LacZ (Edn1; B and F–H) mice for Dct, β-catenin, and Ki67 in early anagen hair follicle. Higher magnification of single immunofluorescent staining of boxed area in (A) and (B) are shown in (C)–(E) and (F)–(H), respectively. (I) Quantification of the percentage of Ki67+ melanocytes. (J–Q) Immunostaining of skin sections from wounded area of control (J and L–N) and Edn1 mice (K and O–Q) for Dct, β-catenin, and Ki67. Higher magnification of single immunofluorescent staining of boxed is shown in (L)–(Q). (R) Quantification of the percentage of Ki67+ melanocytes. (S) Experimental design of in vitro cell proliferation assay using melan-A mouse melanocyte line with knockdown of β-catenin prior to addition of exogenous Edn1 ligand. (T) Quantification of number of melan-A cells at 3 days in culture. Data are presented as average number of cells ± SD. Dashed lines indicate border between epidermis and dermis. Data are presented as the mean ± SD. ∗p < 0.002; ∗∗p < 0.05. The scale bar represents 20 μm. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 6 Loss of β-cat Function Suppresses Edn1-Mediated Effects on McSC Proliferation, Differentiation, and Upward Migration (A) Experimental scheme for treatment of Tyr-CreER; β-cateninfl/fl; K14-rtTA; TetO-Edn1-LacZ (β-cat cKO;Edn1) mice and control K14-rtTA; TetO-Edn1-LacZ (Edn1) mice. (B–E) Gross appearance of Edn1 (B and D) and β-cat cKO; Edn1 mice (C and E) at second (B and C) and third telogen (D and E). (F–K) Immunohistochemistry for indicated markers (F, G, I, and J) and bright-field image (H and K) of bulge/sHG region in skin sections from Edn1 mice (F–H) and β-cat cKO; Edn1 mice (I–K) at anagen II. (L–Q) Bright-field image (L–N) and Dct immunostaining of whole-mount wound site (O–Q) from Tyr-CreER; β-cateninfl/fl (β-cat cKO; L and O), Edn1 (M and P), and β-cat cKO;Edn1 mice (N and Q). (R and S) Quantification of the percentage of Dct+ cells positive for Ki67, Tyr, and pigmentation (R) and the number of Dct+ cells in wounded site (S). Dashed lines indicate border between hair follicle and dermis. Arrowheads indicate double positive cells for indicated markers (F and G) and pigmented cells (H). Data are presented as the mean ± SD. ∗p < 0.05; ∗∗p < 0.02; ∗∗∗p < 0.001. The scale bar represents 1 cm in (B)–(E), 10 μm in (F), and 200 μm in (L). Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions

Figure 7 Edn1 Overexpression Can Overcome the Deficiency of Mc1R in the Generation of Epidermal Melanocytes (A) Experimental scheme. (B–E) Bright-field image (B and D) and Dct immunostaining of whole-mount wound site (C and E) from Tyr-CreER; Mc1Rfl/fl (Mc1R cKO; B and C) and Tyr-CreER; Mc1Rfl/fl; K14-rtTA; TetO-Edn1-LacZ mice (Mc1R cKO; Edn1 Tg; D and E). (F) Quantification of Dct+ epidermal melanocytes in wound site. Asterisks indicate autofluorescence. Data are presented as the mean ± SD. ∗p < 0.05. The scale bar represents 100 μm. See also Figure S7. Cell Reports 2016 15, 1291-1302DOI: (10.1016/j.celrep.2016.04.006) Copyright © 2016 The Authors Terms and Conditions