1
|
Yamaguchi HL, Yamaguchi Y, Peeva E. Hair regrowth in alopecia areata and re-pigmentation in vitiligo in response to treatment: Commonalities and differences. J Eur Acad Dermatol Venereol 2024. [PMID: 39258892 DOI: 10.1111/jdv.20311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/02/2024] [Indexed: 09/12/2024]
Abstract
Both alopecia areata (AA) and vitiligo share common pathogenesis involving, interferon-γ (IFN-γ) and interleukin-15 (IL-15) signalling pathways that activate cytotoxic CD8+ T lymphocytes. These shared mechanisms may explain why both diseases respond to currently available treatments (e.g. topical/systemic corticosteroid) and emerging treatment modalities. As compared with the speed of re-pigmentation in vitiligo lesions, the regeneration of pigmented terminal hair follicles in AA lesions appears fast in response to treatments targeting the inhibition of the Janus kinases (JAKs) and other kinases. We summarize the commonalities and differences between AA and vitiligo focusing on the treatment modalities, followed by recent findings associated with hair follicle stem cells (HFSC) in hair bulge (HBg) and melanocyte stem cells (McSC) in HBg and hair germ (HGm). We then discuss how HFSC and HGm-McSC are involved in the initiation of anagen phase, followed by pigmented terminal hair regrowth in the recovering AA lesions in association with immunology. We also discuss how HBg-McSC contribute to the migration of fully dendritic mature melanocytes into interfollicular epidermis and the equal distribution of melanin in recovering vitiligo lesions. Finally, we present four hypotheses to elucidate the delayed distribution of melanin by mature melanocytes in depigmented vitiligo lesions from the aspects of stem cell biology, as compared with quick hair recovery in AA: (1) McSC are less abundant than HFSC. (2) McSC require a long travel, whereas HFSC reside close to hair regeneration trigger point. (3) Keratinocyte scaffold to accept melanin is not well preserved, whereas scaffold for hair regrowth is well preserved. (4) Inhibitors targeting JAKs and other kinases have less direct effects on melanocyte proliferation and differentiation in vitiligo than hair regrowth in AA. Our review provides an overview of treatment modalities and bridges the gap between scientific advancement and clinical practice in AA and vitiligo management.
Collapse
Affiliation(s)
- Hiroki L Yamaguchi
- Inflammation & Immunology Research Unit, Pfizer, Cambridge, Massachusetts, USA
| | - Yuji Yamaguchi
- Inflammation & Immunology Research Unit, Pfizer, Collegeville, Pennsylvania, USA
| | - Elena Peeva
- Inflammation & Immunology Research Unit, Pfizer, Cambridge, Massachusetts, USA
| |
Collapse
|
2
|
Wang Y, Ge H, Chen P, Wang Y. Wnt/β-catenin signaling in corneal epithelium development, homeostasis, and pathobiology. Exp Eye Res 2024; 246:110022. [PMID: 39117134 DOI: 10.1016/j.exer.2024.110022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/07/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
The corneal epithelium is located on the most anterior surface of the eyeball and protects against external stimuli. The development of the corneal epithelium and the maintenance of corneal homeostasis are essential for the maintenance of visual acuity. It has been discovered recently via the in-depth investigation of ocular surface illnesses that the Wnt/β-catenin signaling pathway is necessary for the growth and stratification of corneal epithelial cells as well as the control of endothelial cell stability. In addition, the Wnt/β-catenin signaling pathway is directly linked to the development of common corneal illnesses such as keratoconus, fungal keratitis, and corneal neovascularization. This review mainly summarizes the role of the Wnt/β-catenin signaling pathway in the development, homeostasis, and pathobiology of cornea, hoping to provide new insights into the study of corneal epithelium and the treatment of related diseases.
Collapse
Affiliation(s)
- Yihui Wang
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Huanhuan Ge
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China
| | - Peng Chen
- School of Basic Medicine, Qingdao University, Qingdao 266071, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Ye Wang
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong 266042, China.
| |
Collapse
|
3
|
Zhang J, Liu Y, Chang J, Zhang R, Liu Z, Liang J, Wang D, Feng J, Zhao W, Xiao H. Shh Gene Regulates the Proliferation and Apoptosis of Dermal Papilla Cells to Affect Its Differential Expression in Secondary Hair Follicle Growth Cycle of Cashmere Goats. Animals (Basel) 2024; 14:2049. [PMID: 39061511 PMCID: PMC11273991 DOI: 10.3390/ani14142049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Sonic hedgehog (Shh) is a component of the Hedgehog signaling pathway, playing an important role in regulating cell proliferation, differentiation, apoptosis, and the repair of damaged organisms. To further clarify the expression pattern of Shh gene in the secondary hair follicle growth cycle of cashmere goats and its mechanism of action on secondary hair follicle papilla cells, and improve cashmere quality, in this study, we took Inner Mongolia Albas white cashmere goats as the research objects and collected skin samples at different growth stages to obtain secondary hair follicles, detected Shh and its gene expression by RT-qPCR, Western blot, immunohistochemistry, and other techniques, while we also cultured DPCs in vitro. Shh gene overexpression and interference vectors were constructed, and the effects of Shh gene on the proliferation and apoptosis of DPCs were studied through cell transfection technology. The results showed that there are significant differences in Shh and its gene expression in the secondary hair follicle growth cycle skins of cashmere goats, with the highest expression level in anagen, followed by catagen, and the lowest expression level in telogen. Shh was mainly expressed in the inner root sheath, outer root sheath, and secondary hair follicle papilla. After the overexpression of Shh gene, the proliferation and vitality of the hair papilla cells were enhanced compared to the interference group. After Shh gene interference, the apoptosis rate of the cells increased, indicating that Shh gene can regulate downstream Ptch, Smo, and Gli2 gene expression to promote the proliferation of DPCs, and thus form its expression pattern in the secondary hair follicle growth cycle of cashmere goats.
Collapse
Affiliation(s)
- Junjie Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Yujing Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Jiale Chang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Ru Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Zhaomin Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Jiayue Liang
- Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dong Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Juan Feng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Wei Zhao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| | - Hongmei Xiao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010010, China
- Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010010, China
| |
Collapse
|
4
|
Hashimoto D, Fujimoto K, Nakata M, Suzuki T, Kumegawa S, Ueda Y, Suzuki K, Asamura S, Yamada G. Developmental and functional roles of androgen and interactive signals for external genitalia and erectile tissues. Reprod Med Biol 2024; 23:e12611. [PMID: 39372370 PMCID: PMC11456227 DOI: 10.1002/rmb2.12611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Background Recent progress in molecular and signal analyses revealed essential functions of cellular signals including androgen and related growth factors such as Wnt regulators for external genitalia (ExG) development and its pathogenesis. Accumulated data showed their fundamental functions also for erectile tissue (corporal body) development and its abnormalities. The current review focuses on such signals from developmental and functional viewpoints. Methods Experimental strategies including histological and molecular signal analyses with conditional mutant mice for androgen and Wnt signals have been extensively utilized. Main findings Essential roles of androgen for the development of male-type ExG and urethral formation are shown. Wnt signals are associated with androgen for male-type ExG organogenesis. Androgen plays essential roles in the development of erectile tissue, the corporal body and it also regulates the duration time of erection. Wnt and other signals are essential for the regulation of mesenchymal cells of erectile tissue as shown by its conditional mutant mouse analyses. Stress signals, continuous erection, and the potential of lymphatic characteristics of the erectile vessels with sinusoids are also shown. Conclusion Reiterated involvement of androgen, Wnt, and other regulatory factors is stated for the development and pathogenesis of ExG and erectile tissues.
Collapse
Affiliation(s)
- Daiki Hashimoto
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kota Fujimoto
- Department of UrologyUrological Science Institute, Yonsei University College of MedicineSeoulSouth Korea
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Masanori Nakata
- Department of Physiology, Faculty of MedicineWakayama Medical UniversityWakayamaJapan
| | - Takuya Suzuki
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Shinji Kumegawa
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Yuko Ueda
- Department of UrologyOsaka Women's and Children's HospitalOsakaJapan
| | - Kentaro Suzuki
- Faculty of Life and Environmental SciencesUniversity of YamanashiYamanashiJapan
| | - Shinichi Asamura
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| | - Gen Yamada
- Department of Plastic and Reconstructive SurgeryWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
5
|
Li J, Tian G, Wang X, Tang H, Liu Y, Guo H, Wang C, Chen Y, Yang Y. Effects of short photoperiod on cashmere growth, hormone concentrations and hair follicle development-related gene expression in cashmere goats. JOURNAL OF APPLIED ANIMAL RESEARCH 2023. [DOI: 10.1080/09712119.2022.2153853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Junda Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Guangjie Tian
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Xingtao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Hongyu Tang
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Yuyang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Hongran Guo
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Chunxin Wang
- Jilin Academy of Agriculture Sciences, Gongzhuling, People’s Republic of China
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, People’s Republic of China
| |
Collapse
|
6
|
Muangsanguan A, Linsaenkart P, Chaitep T, Sangta J, Sommano SR, Sringarm K, Arjin C, Rachtanapun P, Jantanasakulwong K, Phimolsiripol Y, Castagnini JM, Ruksiriwanich W. Hair Growth Promotion and Anti-Hair Loss Effects of By-Products Arabica Coffee Pulp Extracts Using Supercritical Fluid Extraction. Foods 2023; 12:4116. [PMID: 38002174 PMCID: PMC10670875 DOI: 10.3390/foods12224116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/06/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Coffee has been a common ingredient in many traditional hair loss remedies, but limited scientific evidence supports its use, particularly in coffee pulp. Androgenetic alopecia (AGA) is caused by androgens, inflammation, and oxidative stress. In the present study, supercritical fluid extraction (SFE) was used under various conditions to obtain six coffee pulp extracts. The SFE-4 extract, using 50% (v/v) ethanol as a co-solvent at conditions of 100 °C and 500 bars for 30 min, exhibited the highest phenolic, flavonoid, and caffeine contents. Additionally, the SFE-4 extract increased the migration and cell proliferation of HFDPCs (human hair follicle dermal papilla cells), which control hair cycle regulation, and had scavenging effects on ABTS and DPPH radicals. Additionally, the SFE-4 extract showed potassium ion channel opener activity in HFDPCs, as well as a stimulation effect on the enzyme matrix metalloproteinase-2 (MMP-2) (28.53 ± 1.08% of control), which may be related to the vascular endothelial growth factor (VEGF) gene upregulation. In human prostate cancer cells (DU-145) and HFDPC cells, the SFE-4 extract significantly decreased the expression of SRD5A1, SRD5A2, and SRD5A3, an essential pathway involved in AGA. Hair growth factor genes in the Wnt/-catenin (CTNNB1) and Sonic Hedgehog (SHH, SMO, and GLI1) pathways could be significantly activated by the SFE-4 extract. These results imply that employing SFE in coffee pulp extraction could help AGA treatment by preventing hair loss and promoting hair growth pathways. This would help small coffee producers gain economic empowerment and ensure the long-term sustainability of agricultural waste utilization.
Collapse
Affiliation(s)
- Anurak Muangsanguan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.M.); (P.L.); (T.C.)
- Master of Science Program in Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pichchapa Linsaenkart
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.M.); (P.L.); (T.C.)
| | - Tanakarn Chaitep
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.M.); (P.L.); (T.C.)
| | - Jiraporn Sangta
- Interdisciplinary Program in Biotechnology, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sarana Rose Sommano
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand; (S.R.S.); (K.S.)
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
- Department of Plant and Soil Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Korawan Sringarm
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand; (S.R.S.); (K.S.)
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Chaiwat Arjin
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Pornchai Rachtanapun
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Kittisak Jantanasakulwong
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Yuthana Phimolsiripol
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Juan M. Castagnini
- Research Group in Innovative Technologies for Sustainable Food (ALISOST), Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy, Universitat de València, Avenida Vicent Andrés Estellés s/n, 46100 Burjassot, Spain;
| | - Warintorn Ruksiriwanich
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (A.M.); (P.L.); (T.C.)
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand; (S.R.S.); (K.S.)
- Center of Excellence in Agro Bio-Circular-Green Industry (Agro BCG), Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand; (P.R.); (K.J.); (Y.P.)
| |
Collapse
|
7
|
Ku YC, Omer Sulaiman H, Anderson SR, Abtahi AR. The Potential Role of Exosomes in Aesthetic Plastic Surgery: A Review of Current Literature. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2023; 11:e5051. [PMID: 37313480 PMCID: PMC10259637 DOI: 10.1097/gox.0000000000005051] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/13/2023] [Indexed: 06/15/2023]
Abstract
Despite an increasing surge of exosome use throughout the aesthetic arena, a paucity of published exosome-based literature exists. Exosomes are membrane-bound extracellular vesicles derived from various cell types, exerting effects via intercellular communication and regulation of several signaling pathways. The purpose of this review was to summarize published articles elucidating mechanisms and potential applications, report available products and clinical techniques, and prompt further investigation of this emerging treatment within the plastic surgery community. Methods A literature review was performed using PubMed with keywords exosomes, secretomes, extracellular vesicles, plastic surgery, skin rejuvenation, scar revision, hair growth, body contouring, and breast augmentation. Publications from 2010 to 2021 were analyzed for relevance and level of evidence. A Google search identified exosome distributors, where manufacturing/procurement details, price, efficacy, and clinical indications for use were obtained by direct contact and summarized in table format. Results Exosomes are currently derived from bone marrow, placental, adipose, and umbilical cord tissue. Laboratory-based exosome studies demonstrate enhanced outcomes in skin rejuvenation, scar revision, hair restoration, and fat graft survival on the macro and micro levels. Clinical studies are limited to anecdotal results. Prices vary considerably from $60 to nearly $5000 based on company, source tissue, and exosome concentration. No exosome-based products are currently Food and Drug Administration-approved. Conclusions Administered alone or as an adjunct, current reports show promise in several areas of aesthetic plastic surgery. However, ongoing investigation is warranted to further delineate concentration, application, safety profile, and overall outcome efficacy.
Collapse
Affiliation(s)
- Ying C. Ku
- From the Department of Surgery, Campbell University School of Osteopathic Medicine, Buies Creek, NC
| | - Hafsa Omer Sulaiman
- Department of Surgery, Hull University Hospitals, Castle Hill Hospital, Cottingham, UK
| | - Spencer R. Anderson
- Division of Plastic and Reconstructive Surgery, Wright State University Boonshoft School of Medicine, Dayton, OH
| | - Ali R. Abtahi
- Division of Plastic and Reconstructive Surgery, University of Iowa Hospitals and Clinics, Iowa City, IA
| |
Collapse
|
8
|
Wang S, Hu T, He M, Gu Y, Cao X, Yuan Z, Lv X, Getachew T, Quan K, Sun W. Defining ovine dermal papilla cell markers and identifying key signaling pathways regulating its intrinsic properties. Front Vet Sci 2023; 10:1127501. [PMID: 36923053 PMCID: PMC10009177 DOI: 10.3389/fvets.2023.1127501] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Dermal papilla cell (DPC), one of the key cell types during hair follicle development and regeneration, specifies hair size, shape and cycling. It is also an important in vitro screening model for hair growth. Although some characteristics of DPCs, such as agglutinative growth and marker genes, have been studied in mice and humans, the intrinsic properties of ovine DPCs and the regulatory mechanism of the intrinsic properties during continued culture in vitro remained unknown. In this study, based on our previous single-cell transcriptome sequencing on sheep lambskin, we verified SOX18 and PDGFRA as the novel marker genes of ovine DPCs through immunofluorescence staining on skin sections and cultured DPCs. Using continued cell culture and alkaline phosphatase staining, we found that different from mice and humans, ovine DPCs exhibit particularly robust and stable aggregation with unbated alkaline phosphatase activity till 30 passages during continued culture in vitro. Also, we found that the expression of some marker genes and the activity of Wnt/β-catenin signaling differ between early passaged DPCs and multiple passaged DPCs. Further, using Wnt/β-catenin agonist and antagonist, we demonstrated that Wnt/β-catenin signaling could regulate cell aggregation and alkaline phosphatase activity of ovine DPCs through regulating FGF and IGF signaling. This study provides the basis for isolating ovine DPCs and defines their intrinsic properties, which contribute to improving wool performance and medicine of hair regeneration.
Collapse
Affiliation(s)
- Shanhe Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Tingyan Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Mingliang He
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yifei Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xiukai Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Zehu Yuan
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Xiaoyang Lv
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China
| | - Tesfaye Getachew
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa, Ethiopia
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China.,International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou, China.,"Innovative China" "Belt and Road" International Agricultural Technology Innovation Institute for Evaluation, Protection, and Improvement on Sheep Genetic Resource, Yangzhou, China
| |
Collapse
|
9
|
The Molecular Mechanism of Natural Products Activating Wnt/β-Catenin Signaling Pathway for Improving Hair Loss. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111856. [PMID: 36430990 PMCID: PMC9693075 DOI: 10.3390/life12111856] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Hair loss, or alopecia, is a dermatological disorder that causes psychological stress and poor quality of life. Drug-based therapeutics such as finasteride and minoxidil have been clinically used to treat hair loss, but they have limitations due to their several side effects in patients. To solve this problem, there has been meaningful progress in elucidating the molecular mechanisms of hair growth and finding novel targets to develop therapeutics to treat it. Among various signaling pathways, Wnt/β-catenin plays an essential role in hair follicle development, the hair cycle, and regeneration. Thus, much research has demonstrated that various natural products worldwide promote hair growth by stimulating Wnt/β-catenin signaling. This review discusses the functional role of the Wnt/β-catenin pathway and its related signaling molecules. We also review the molecular mechanism of the natural products or compounds that activate Wnt/β-catenin signaling and provide insights into developing therapeutics or cosmeceuticals that treat hair loss.
Collapse
|
10
|
Wan J, Dai H, Zhang X, Liu S, Lin Y, Somani AK, Xie J, Han J. Distinct transcriptomic landscapes of cutaneous basal cell carcinomas and squamous cell carcinomas. Genes Dis 2021; 8:181-192. [PMID: 33997165 PMCID: PMC8099692 DOI: 10.1016/j.gendis.2019.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/06/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
The majority of non-melanoma skin cancer (NMSC) is cutaneous basal cell carcinoma (BCC) or squamous cell carcinoma (SCC), which are also called keratinocyte carcinomas, as both of them originate from keratinocytes. The incidence of keratinocyte carcinomas is over 5 million per year in the US, three-fold higher than the total incidence of all other types of cancer combined. While there are several reports on gene expression profiling of BCC and SCC, there are significant variations in the reported gene expression changes in different studies. One reason is that tumor-adjacent normal skin specimens were not included in many studies as matched controls. Furthermore, while numerous studies of skin stem cells in mouse models have been reported, their relevance to human skin cancer remains unknown. In this report, we analyzed gene expression profiles of paired specimens of keratinocyte carcinomas with their matched normal skin tissues as the control. Among several novel findings, we discovered a significant number of zinc finger encoding genes up-regulated in human BCC. In BCC, a novel link was found between hedgehog signaling, Wnt signaling, and the cilium. While the SCC cancer-stem-cell gene signature is shared between human and mouse SCCs, the hair follicle stem-cell signature of mice was not highly represented in human SCC. Differential gene expression (DEG) in human BCC shares gene signature with both bulge and epidermal stem cells. We have also determined that human BCCs and SCCs have distinct gene expression patterns, and some of them are not fully reflected in current mouse models.
Collapse
Affiliation(s)
- Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- School of Informatics and Computing, Indiana University – Purdue University at Indianapolis, Indianapolis, IN, 46202, USA
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300000, PR China
| | - Xiaoli Zhang
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yuan Lin
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Ally-Khan Somani
- Dermatologic Surgery & Cutaneous Oncology Division, Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingwu Xie
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jiali Han
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, 46202, USA
| |
Collapse
|
11
|
Weng T, Wu P, Zhang W, Zheng Y, Li Q, Jin R, Chen H, You C, Guo S, Han C, Wang X. Regeneration of skin appendages and nerves: current status and further challenges. J Transl Med 2020; 18:53. [PMID: 32014004 PMCID: PMC6996190 DOI: 10.1186/s12967-020-02248-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Tissue-engineered skin (TES), as an analogue of native skin, is promising for wound repair and regeneration. However, a major drawback of TES products is a lack of skin appendages and nerves to enhance skin healing, structural integrity and skin vitality. Skin appendages and nerves are important constituents for fully functional skin. To date, many studies have yielded remarkable results in the field of skin appendages reconstruction and nerve regeneration. However, patients often complain about a loss of skin sensation and even cutaneous chronic pain. Restoration of pain, temperature, and touch perceptions should now be a major challenge to solve in order to improve patients’ quality of life. Current strategies to create skin appendages and sensory nerve regeneration are mainly based on different types of seeding cells, scaffold materials, bioactive factors and involved signaling pathways. This article provides a comprehensive overview of different strategies for, and advances in, skin appendages and sensory nerve regeneration, which is an important issue in the field of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Pan Wu
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Wei Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Yurong Zheng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qiong Li
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Haojiao Chen
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Chuangang You
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Songxue Guo
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
12
|
Ohyama M. Use of human intra-tissue stem/progenitor cells and induced pluripotent stem cells for hair follicle regeneration. Inflamm Regen 2019; 39:4. [PMID: 30834027 PMCID: PMC6388497 DOI: 10.1186/s41232-019-0093-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/23/2019] [Indexed: 01/07/2023] Open
Abstract
Background The hair follicle (HF) is a unique miniorgan, which self-renews for a lifetime. Stem cell populations of multiple lineages reside within human HF and enable its regeneration. In addition to resident HF stem/progenitor cells (HFSPCs), the cells with similar biological properties can be induced from human-induced pluripotent stem cells (hiPSCs). As approaches to regenerate HF by combining HF-derived cells have been established in rodents and a huge demand exists to treat hair loss diseases, attempts have been made to bioengineer human HF using HFSPCs or hiPSCs. Main body of the abstract The aim of this review is to comprehensively summarize the strategies to regenerate human HF using HFSPCs or hiPSCs. HF morphogenesis and regeneration are enabled by well-orchestrated epithelial-mesenchymal interactions (EMIs). In rodents, various combinations of keratinocytes with mesenchymal (dermal) cells with trichogenic capacity, which were transplanted into in vivo environment, have successfully generated HF structures. The regeneration efficiency was higher, when epithelial or dermal HFSPCs were adopted. The success in HF formation most likely depended on high receptivity to trichogenic dermal signals and/or potent hair inductive capacity of HFSPCs. In theory, the use of epithelial HFSPCs in the bulge area and dermal papilla cells, their precursor cells in the dermal sheath, or trichogenic neonatal dermal cells should elicit intense EMI sufficient for HF formation. However, technical hurdles, represented by the limitation in starting materials and the loss of intrinsic properties during in vitro expansion, hamper the stable reconstitution of human HFs with this approach. Several strategies, including the amelioration of culture condition or compartmentalization of cells to strengthen EMI, can be conceived to overcome this obstacle. Obviously, use of hiPSCs can resolve the shortage of the materials once reliable protocols to induce wanted HFSPC subsets have been developed, which is in progress. Taking advantage of their pluripotency, hiPSCs may facilitate previously unthinkable approaches to regenerate human HFs, for instance, via bioengineering of 3D integumentary organ system, which can also be applied for the treatment of other diseases. Short conclusion Further development of methodologies to reproduce bona fide EMI in HF formation is indispensable. However, human HFSPCs and hiPSCs hold promise as materials for human HF regeneration.
Collapse
Affiliation(s)
- Manabu Ohyama
- Department of Dermatology, Kyorin University Faculty of Medicine, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611 Japan
| |
Collapse
|
13
|
Wang P, Ma H, Li Y, Chen D, Li X, Gao X. Retracted Article: FOXC1 silencing promotes A549 cell apoptosis through inhibiting the PI3K/AKT/hedgehog/Gli2 signaling pathway. RSC Adv 2018; 8:33786-33793. [PMID: 35548824 PMCID: PMC9086723 DOI: 10.1039/c8ra06041j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/17/2018] [Indexed: 11/30/2022] Open
Abstract
Lung cancer begins in the lung and is a leading cause of premature death. Forkhead box C1 (FOXC1) has been reported to play an important role in different types of cancer, and evidence suggests that FOXC1 is highly expressed in non-small cell lung cancer (NSCLC) patients. However, the function and molecular mechanism of FOXC1 in the NSCLC cell line A549 is still unclear. In the present study, we indicate that FOXC1 is expressed in the NSCLC cell lines A549, H460, and SK-MES-1 at a high level compared with control human bronchial epithelial (HBE) cells. FOXC1 silencing promotes A549 cell apoptosis, whereas it inhibits cell survival. The levels of anti-apoptosis protein Bcl-2 decreased and the expression of pro-apoptosis protein Bax increased in FOXC1 silenced cells. Further studies show that FOXC1 knockdown inhibits the PI3K/AKT/hedgehog/Gli2 pathway. Overexpressed AKT or Gli2 reversed the effects of FOXC1 silencing on A549 cell survival and apoptosis. Taken together, our results conclude that FOXC1 silencing reduced the survival of cancer cells and promoted their apoptosis, and that the PI3K/AKT/hedgehog/Gli2 pathway plays an important role in the functioning of FOXC1 silencing.
Collapse
Affiliation(s)
- Pei Wang
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| | - Hongbing Ma
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| | - Yong Li
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| | - Dong Chen
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| | - Xiaohui Li
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| | - Xiang Gao
- Department of Cardiothoracic Surgery, Huaihe Hospital of Henan University Baobei Road No. 8 Kaifeng 475000 China +86-0371-23906599 +86-0371-23906599
| |
Collapse
|
14
|
Biggs LC, Mäkelä OJ, Myllymäki SM, Das Roy R, Närhi K, Pispa J, Mustonen T, Mikkola ML. Hair follicle dermal condensation forms via Fgf20 primed cell cycle exit, cell motility, and aggregation. eLife 2018; 7:36468. [PMID: 30063206 PMCID: PMC6107334 DOI: 10.7554/elife.36468] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal condensation is a critical step in organogenesis, yet the underlying molecular and cellular mechanisms remain poorly understood. The hair follicle dermal condensate is the precursor to the permanent mesenchymal unit of the hair follicle, the dermal papilla, which regulates hair cycling throughout life and bears hair inductive potential. Dermal condensate morphogenesis depends on epithelial Fibroblast Growth Factor 20 (Fgf20). Here, we combine mouse models with 3D and 4D microscopy to demonstrate that dermal condensates form de novo and via directional migration. We identify cell cycle exit and cell shape changes as early hallmarks of dermal condensate morphogenesis and find that Fgf20 primes these cellular behaviors and enhances cell motility and condensation. RNAseq profiling of immediate Fgf20 targets revealed induction of a subset of dermal condensate marker genes. Collectively, these data indicate that dermal condensation occurs via directed cell movement and that Fgf20 orchestrates the early cellular and molecular events. All mammal hair springs from hair follicles under the skin. These follicles sit in the dermis, beneath the outermost skin layer, the epidermis. In the embryo, hair follicles develop from unspecialized cells in two tissues, the epithelium and the mesenchyme, which will later develop into the dermis and epidermis, respectively. As development progresses, the cells of these tissues begin to cluster, and signals passing back and forth between the epithelium and mesenchyme instruct the cells what to do. In the mesenchyme, cells called fibroblasts squeeze up against their neighbors, forming patches called dermal condensates. These mature into so-called dermal papillae, which supply specific molecules called growth factors that regulate hair formation throughout lifetime. Fibroblasts in the developing skin respond to a signal from the epithelium called fibroblast growth factor 20 (Fgf20), but we do not yet understand its effects. It is possible that Fgf20 tells the cells to divide, forming clusters of daughter cells around their current location. Or, it could be that Fgf20 tells the cells to move, encouraging them to travel towards one another to form groups. To address this question, Biggs, Mäkelä et al. examined developing mouse skin grown in the laboratory. They traced cells marked with fluorescent tags to analyze their behavior as the condensates formed. This revealed that the Fgf20 signal acts as a rallying call, triggering fibroblast movement. The cells changed shape and moved towards one another, rather than dividing to create their own clusters. In fact, they switched off their own cell cycle as the condensates formed, halting their ability to divide. A technique called RNA sequencing revealed that Fgf20 also promotes the use of genes known to be active in dermal condensates. Dermal papillae control hair growth, and transplanting them under the skin can form new hair follicles. However, these cells lose this ability when grown in the laboratory. Understanding how they develop could be beneficial for future hair growth therapy. Further work could also address fundamental questions in embryology. Condensates of cells from the mesenchyme also precede the formation of limbs, bones, muscles and organs. Extending this work could help us to understand this critical developmental step.
Collapse
Affiliation(s)
- Leah C Biggs
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Otto Jm Mäkelä
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Satu-Marja Myllymäki
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Rishi Das Roy
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Katja Närhi
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Johanna Pispa
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Tuija Mustonen
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Marja L Mikkola
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Zhou L, Wang H, Jing J, Yu L, Wu X, Lu Z. Regulation of hair follicle development by exosomes derived from dermal papilla cells. Biochem Biophys Res Commun 2018; 500:325-332. [PMID: 29654758 DOI: 10.1016/j.bbrc.2018.04.067] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 04/10/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Dermal papilla cells (DPCs) play a critical role in the regulation of hair follicle (HF) growth, formation, and cycling. DPCs are thought to regulate HF growth through a paracrine mechanism, in which exosomes may play a critical role. METHODS DPC-Exos were cutaneously injected into HFs at different HF cycle stages and the effects were evaluated by histological and immunohistochemical analyses. The effects of DPC-Exos on proliferation, migration, and cell cycle status of outer root sheath cells (ORSCs) were evaluated. After treatment of DPC-Exos, changes in mRNA and protein levels of β-catenin and Sonic hedgehog (Shh) in ORSCs were detected. RESULTS DPC-Exos were approximately 105 nm in diameter and expressed tumor susceptibility gene 101, cluster of differentiation (CD)9, and CD63. Injection of DPC-Exos accelerated the onset of HF anagen and delayed catagen in mice. Immunohistochemical analyses revealed that β-catenin and Shh levels were upregulated in the skin. In vitro, DPC-Exo treatment enhanced ORSC proliferation and migration, and stimulated the expression of β-catenin and Shh. CONCLUSION DPC-Exos contribute to the regulation of HF growth and development, and provide a potential avenue for the treatment of hair loss.
Collapse
Affiliation(s)
- Lijuan Zhou
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Wang
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Jing
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lijuan Yu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xianjie Wu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongfa Lu
- Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
Roles of the Hedgehog Signaling Pathway in Epidermal and Hair Follicle Development, Homeostasis, and Cancer. J Dev Biol 2017; 5:jdb5040012. [PMID: 29615568 PMCID: PMC5831796 DOI: 10.3390/jdb5040012] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/15/2017] [Accepted: 11/18/2017] [Indexed: 12/15/2022] Open
Abstract
The epidermis is the outermost layer of the skin and provides a protective barrier against environmental insults. It is a rapidly-renewing tissue undergoing constant regeneration, maintained by several types of stem cells. The Hedgehog (HH) signaling pathway is one of the fundamental signaling pathways that contributes to epidermal development, homeostasis, and repair, as well as to hair follicle development and follicle bulge stem cell maintenance. The HH pathway interacts with other signal transduction pathways, including those activated by Wnt, bone morphogenetic protein, platelet-derived growth factor, Notch, and ectodysplasin. Furthermore, aberrant activation of HH signaling is associated with various tumors, including basal cell carcinoma. Therefore, an understanding of the regulatory mechanisms of the HH signaling pathway is important for elucidating fundamental mechanisms underlying both organogenesis and carcinogenesis. In this review, we discuss the role of the HH signaling pathway in the development and homeostasis epidermis and hair follicles, and in basal cell carcinoma formation, providing an update of current knowledge in this field.
Collapse
|
17
|
Veltri A, Lang C, Lien WH. Concise Review: Wnt Signaling Pathways in Skin Development and Epidermal Stem Cells. Stem Cells 2017; 36:22-35. [DOI: 10.1002/stem.2723] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/23/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Anthony Veltri
- de Duve Institute, Faculty of Medicine, Université Catholique de Louvain; Brussels Belgium
| | - Christopher Lang
- de Duve Institute, Faculty of Medicine, Université Catholique de Louvain; Brussels Belgium
| | - Wen-Hui Lien
- de Duve Institute, Faculty of Medicine, Université Catholique de Louvain; Brussels Belgium
| |
Collapse
|
18
|
Hair follicle stem cell proliferation, Akt and Wnt signaling activation in TPA-induced hair regeneration. Histochem Cell Biol 2017; 147:749-758. [DOI: 10.1007/s00418-017-1540-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2017] [Indexed: 01/21/2023]
|
19
|
Choe Y, Huynh T, Pleasure SJ. Epithelial cells supply Sonic Hedgehog to the perinatal dentate gyrus via transport by platelets. eLife 2015; 4. [PMID: 26457609 PMCID: PMC4600762 DOI: 10.7554/elife.07834] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 09/12/2015] [Indexed: 12/14/2022] Open
Abstract
Dentate neural stem cells produce neurons throughout life in mammals. Sonic hedgehog (Shh) is critical for maintenance of these cells; however, the perinatal source of Shh is enigmatic. In the present study, we examined the role of Shh expressed by hair follicles (HFs) that expand perinatally in temporal concordance with the proliferation of Shh-responding dentate stem cells. Specific inhibition of Shh from HFs or from epithelial sources in general hindered development of Shh-responding dentate stem cells. We also found that the blood–brain barrier (BBB) of the perinatal dentate gyrus (DG) is leaky with stem cells in the dentate exposed to blood-born factors. In attempting to identify how Shh might be transported in blood, we found that platelets contain epithelial Shh, provide Shh to the perinatal DG and that inhibition of platelet generation reduced hedgehog-responsive dentate stem cells. DOI:http://dx.doi.org/10.7554/eLife.07834.001 Although most of the neurons in the brain have been made by the time we are born, new neurons develop throughout life in part of the brain called the hippocampus. These neurons are thought to help with learning and forming memories. Conditions such as depression and Alzheimer's disease have been linked to not being able to produce enough new neurons. The neurons develop from a pool of stem cells in part of the hippocampus. A protein called Sonic Hedgehog (Shh) helps to ensure there are enough stem cells and control when they develop into new neurons. The brain cells that produce Shh in adult mice do not appear until a week after birth, by which point the stem cells are already present and generating neurons. This has led scientists to question where these cells get Shh from around the time of birth. One idea is that cells outside of the brain contribute the Shh such as hair follicles—the structures that hairs grow out of—in the scalp. Hair follicles produce Shh, develop at around the same time as the brain stem cells, and are known to regulate the development of other nearby stem cells. So, Choe et al. conducted a series of experiments in genetically engineered newborn mice and found that the brain stem cells multiply at around the same time that the hair follicles start to produce Shh. Furthermore, reducing the amount of Shh produced by the hair follicles hampered the growth of these stem cells and caused fewer neurons to develop from the stem cell pool. These results raised the question of how Shh gets from the hair follicles to the stem cell pool in the developing brain. In adult animals, a barrier exists between the brain and the blood supply to protect the brain from infection. However, parts of this barrier are still leaky before birth, which might allow blood cells to carry Shh to the brain. Cloe et al. found that platelets—the blood cells responsible for clotting—are able to carry Shh to the brain stem cell pool. Further experiments showed that preventing platelets from forming caused fewer stem cells to develop. The suggestion that Shh from the epithelium—the tissue layer that hair follicles are found in—is able to signal to the brain during a specific window of time raises several questions that require further study. Does epithelial Shh also signal to other organs during embryonic or postnatal development? Does injury to the nervous system that increases the permeability of the blood–brain barrier lead to the delivery of Shh to the brain via the circulation in adult animals? DOI:http://dx.doi.org/10.7554/eLife.07834.002
Collapse
Affiliation(s)
- Youngshik Choe
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Trung Huynh
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Samuel J Pleasure
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Program in Neuroscience, University of California, San Francisco, San Francisco, United States.,Program in Developmental Stem Cell Biology, University of California, San Francisco, San Francisco, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
20
|
Carroll LS, Capecchi MR. Hoxc8 initiates an ectopic mammary program by regulating Fgf10 and Tbx3 expression and Wnt/β-catenin signaling. Development 2015; 142:4056-67. [PMID: 26459221 DOI: 10.1242/dev.128298] [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] [Received: 07/09/2015] [Accepted: 10/01/2015] [Indexed: 01/22/2023]
Abstract
The role of Hox genes in the formation of cutaneous accessory organs such as hair follicles and mammary glands has proved elusive, a likely consequence of overlapping function and expression among various homeobox factors. Lineage and immunohistochemical analysis of Hoxc8 in mice revealed that this midthoracic Hox gene has transient but strong regional expression in ventrolateral surface ectoderm at E10.5, much earlier than previously reported. Targeted mice were generated to conditionally misexpress Hoxc8 from the Rosa locus using select Cre drivers, which significantly expanded the domain of thoracic identity in mutant embryos. Accompanying this expansion was the induction of paired zones of ectopic mammary development in the cervical region, which generated between three and five pairs of mammary placodes anterior to the first wild-type mammary rudiment. These rudiments expressed the mammary placode markers Wnt10b and Tbx3 and were labeled by antibodies to the mammary mesenchyme markers ERα and androgen receptor. Somitic Fgf10 expression, which is required for normal mammary line formation, was upregulated in mutant cervical somites, and conditional ablation of ectodermal Tbx3 expression eliminated all normally positioned and ectopic mammary placodes. We present evidence that Hoxc8 participates in regulating the initiation stages of mammary placode morphogenesis, and suggest that this and other Hox genes are likely to have important roles during regional specification and initiation of these and other cutaneous accessory organs.
Collapse
Affiliation(s)
- Lara S Carroll
- Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA
| | - Mario R Capecchi
- Department of Human Genetics and Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
21
|
Nakamura M, Fukunaga-Kalabis M, Yamaguchi Y, Furuhashi T, Nishida E, Kato H, Mizuno T, Sugiura M, Morita A. Site-specific migration of human fetal melanocytes in volar skin. J Dermatol Sci 2015; 78:143-8. [PMID: 25818865 DOI: 10.1016/j.jdermsci.2015.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 03/03/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Melanocytes originate from the neural crest and migrate ventrally from the dorsal neural tube during embryogenesis. How human melanocytes locate at their suitable positions during embryogenesis, however, is unclear. Although a growing body of evidence indicates that melanocytes, melanoblasts, and melanocyte stem cells are closely related to hair follicles, little is known about volar skin. OBJECTIVE The aim of this study was to observe skin development during human fetal period and clarify the site-specific migration process of human fetal sole melanocytes. METHODS We obtained 4-mm punch biopsies from the scalp, back, abdomen, and right sole of 36 aborted fetuses (gestational age 12-21 weeks). We compared the migration process between hairly areas and volar areas by immunohistochemical staining. RESULTS Immunohistochemical examination revealed that gp100 (HMB-45) sensitively detects human melanocytes in embryogenesis. Melanocytes were present at the epidermal base, where hair placodes/buds form at 12-15 weeks gestation. Fetal melanocytes in hair follicles are supplied from the epidermis. In volar skin, melanocytes originally localize only in the acrosyringium, where they migrate deeper into with gland development at 16-18 weeks gestation. Palmoplantar melanocyte migration and maturation processes differ considerably from those of the other hairy skin sites. CONCLUSION Eccrine sweat glands seem to have a central role in the palmoplantar melanocyte migration process, similar to the role of hair follicles in hairy sites.
Collapse
Affiliation(s)
- Motoki Nakamura
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | | | - Yuji Yamaguchi
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takuya Furuhashi
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Emi Nishida
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | - Mayumi Sugiura
- Department of Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
22
|
The disruption of a novel limb cis-regulatory element of SHH is associated with autosomal dominant preaxial polydactyly-hypertrichosis. Eur J Hum Genet 2015; 24:37-43. [PMID: 25782671 DOI: 10.1038/ejhg.2015.53] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 02/12/2015] [Accepted: 02/19/2015] [Indexed: 12/16/2022] Open
Abstract
The expression gradient of the morphogen Sonic Hedgehog (SHH) is crucial in establishing the number and the identity of the digits during anteroposterior patterning of the limb. Its anterior ectopic expression is responsible for preaxial polydactyly (PPD). Most of these malformations are due to the gain-of-function of the Zone of Polarizing Activity Regulatory Sequence, the only limb-specific enhancer of SHH known to date. We report a family affected with a novel condition associating PPD and hypertrichosis of the upper back, following an autosomal dominant mode of inheritance. This phenotype is consistent with deregulation of SHH expression during limb and follicle development. In affected members, we identified a 2 kb deletion located ~240 kb upstream from the SHH promoter. The deleted sequence is capable of repressing the transcriptional activity of the SHH promoter in vitro, consistent with a silencer activity. We hypothesize that the deletion of this silencer could be responsible for SHH deregulation during development, leading to a PPD-hypertrichosis phenotype.
Collapse
|
23
|
BMP-FGF signaling axis mediates Wnt-induced epidermal stratification in developing mammalian skin. PLoS Genet 2014; 10:e1004687. [PMID: 25329657 PMCID: PMC4199507 DOI: 10.1371/journal.pgen.1004687] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/19/2014] [Indexed: 01/08/2023] Open
Abstract
Epidermal stratification of the mammalian skin requires proliferative basal progenitors to generate intermediate cells that separate from the basal layer and are replaced by post-mitotic cells. Although Wnt signaling has been implicated in this developmental process, the mechanism underlying Wnt-mediated regulation of basal progenitors remains elusive. Here we show that Wnt secreted from proliferative basal cells is not required for their differentiation. However, epidermal production of Wnts is essential for the formation of the spinous layer through modulation of a BMP-FGF signaling cascade in the dermis. The spinous layer defects caused by disruption of Wnt secretion can be restored by transgenically expressed Bmp4. Non-cell autonomous BMP4 promotes activation of FGF7 and FGF10 signaling, leading to an increase in proliferative basal cell population. Our findings identify an essential BMP-FGF signaling axis in the dermis that responds to the epidermal Wnts and feedbacks to regulate basal progenitors during epidermal stratification. Epidermis, a thin layer of stratified epithelium forming the outmost surface of the skin, provides the crucial function to protect animals from environmental insults, such as bacterial pathogens and water loss. This barrier function is established in embryogenesis, during which single layered epithelial cells differentiate into distinct layers of keratinocytes. Many human genetic diseases are featured with epidermal disruption, affecting at least one in five patients. Skin regeneration and future therapeutics require a thorough understanding of the molecular mechanisms underlying epidermal stratification. Wnt ligands have been implicated in hair follicle induction during skin development and self-renewal of stem cells in the interfollicular epidermis of adult skin; however, little is known about the mechanism of how Wnt signaling controls epidermal stratification during embryogenesis. In this study, by using a genetic mouse model to disrupt Wnt production in skin development, we found that signaling of epidermal Wnt in the dermis initiate mesenchymal responses by activating a Bone Morphogenetic Protein (BMP) and Fibroblast growth factor (FGF) signaling cascade, and this activation is required for feedback regulations in the embryonic epidermis to control stratification. Our findings identify a genetic hierarchy of signaling essential for epidermal-mesenchymal interactions, and promote our understanding of mammalian skin development.
Collapse
|
24
|
Miyagawa S, Harada M, Matsumaru D, Tanaka K, Inoue C, Nakahara C, Haraguchi R, Matsushita S, Suzuki K, Nakagata N, Ng RCL, Akita K, Lui VCH, Yamada G. Disruption of the temporally regulated cloaca endodermal β-catenin signaling causes anorectal malformations. Cell Death Differ 2014; 21:990-7. [PMID: 24632946 PMCID: PMC4013517 DOI: 10.1038/cdd.2014.21] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 12/30/2013] [Accepted: 01/09/2014] [Indexed: 12/22/2022] Open
Abstract
The cloaca is temporally formed and eventually divided by the urorectal septum (URS) during urogenital and anorectal organ development. Although congenital malformations, such as anorectal malformations (ARMs), are frequently observed during this process, the underlying pathogenic mechanisms remain unclear. β-Catenin is a critical component of canonical Wnt signaling and is essential for the regulation of cell differentiation and morphogenesis during embryogenesis. The expression of β-catenin is observed in endodermal epithelia, including URS epithelia. We modulated the β-catenin gene conditionally in endodermal epithelia by utilizing tamoxifen-inducible Cre driver line (ShhCreERT2). Both β-catenin loss- and gain-of-function (LOF and GOF) mutants displayed abnormal clefts in the perineal region and hypoplastic elongation of the URS. The mutants also displayed reduced cell proliferation in the URS mesenchyme. In addition, the β-catenin GOF mutants displayed reduced apoptosis and subsequently increased apoptosis in the URS epithelium. This instability possibly resulted in reduced expression levels of differentiation markers, such as keratin 1 and filaggrin, in the perineal epithelia. The expression of bone morphogenetic protein (Bmp) genes, such as Bmp4 and Bmp7, was also ectopically induced in the epithelia of the URS in the β-catenin GOF mutants. The expression of the Msx2 gene and phosphorylated-Smad1/5/8, possible readouts of Bmp signaling, was also increased in the mutants. Moreover, we introduced an additional mutation for a Bmp receptor gene: BmprIA. The ShhCreERT2/+; β-cateninflox(ex3)/+; BmprIAflox/− mutants displayed partial restoration of URS elongation compared with the β-catenin GOF mutants. These results indicate that some ARM phenotypes in the β-catenin GOF mutants were caused by abnormal Bmp signaling. The current analysis revealed the close relation of endodermal β-catenin signaling to the ARM phenotypes. These results are considered to shed light on the pathogenic mechanisms of human ARMs.
Collapse
Affiliation(s)
- S Miyagawa
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan [3] Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Aichi, Japan
| | - M Harada
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan [3] Unit of Clinical Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - D Matsumaru
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - K Tanaka
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - C Inoue
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - C Nakahara
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - R Haraguchi
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan [3] Department of Molecular Pathology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - S Matsushita
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - K Suzuki
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - N Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - R C-L Ng
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - K Akita
- Unit of Clinical Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - V C-H Lui
- Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - G Yamada
- 1] Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan [2] Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
25
|
Regulatory effect of β-catenin on proliferation of hair follicle stem cells involves PI3K/Akt pathway. J Appl Biomed 2013. [DOI: 10.2478/v10136-012-0019-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
26
|
Bohr S, Patel SJ, Vasko R, Shen K, Huang G, Yarmush ML, Berthiaume F. Highly upregulated Lhx2 in the Foxn1-/- nude mouse phenotype reflects a dysregulated and expanded epidermal stem cell niche. PLoS One 2013; 8:e64223. [PMID: 23696871 PMCID: PMC3656088 DOI: 10.1371/journal.pone.0064223] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/12/2013] [Indexed: 12/17/2022] Open
Abstract
Hair cycling is a prime example of stem cell dependent tissue regeneration and replenishment, and its regulatory mechanisms remain poorly understood. In the present study, we evaluated the effect of a blockage in terminal keratinocytic lineage differentiation in the Foxn1(-/-) nude phenotype on the epithelial progeny. Most notably we found a constitutive upregulation of LIM homeobox protein 2 (Lhx2), a marker gene of epithelial stem cellness indispensible for hair cycle progression. However, histological evidence along with an erratic, acyclic rise of otherwise suppressed CyclinD1 levels along with several key markers of keratinocyte lineage differentiation indicate a frustrated expansion of epithelial stem cell niches in skin. In addition, CD49f/CD34/CD200-based profiling demonstrated highly significant shifts in subpopulations of epithelial progeny. Intriguingly this appeared to include the expansion of Oct4+ stem cells in dermal fractions of skin isolates in the Foxn1 knock-out opposed to wild type. Overall our findings indicate that the Foxn1(-/-) phenotype has a strong impact on epithelial progeny and thus offers a promising model to study maintenance and regulation of stem cell niches within skin not feasible in other in vitro or in vivo models.
Collapse
Affiliation(s)
- Stefan Bohr
- Center for Engineering in Medicine, Shriners Hospitals for Children and Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | |
Collapse
|
27
|
Mazahery AR, Suzuki K, Nagafuchi A, Miyajima M, Nakagata N, Orvis GD, Behringer RR, Yamada G. Functional analysis of ectodermal β-catenin during external genitalia formation. Congenit Anom (Kyoto) 2013; 53:34-41. [PMID: 23480356 DOI: 10.1111/cga.12001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/10/2012] [Indexed: 02/03/2023]
Abstract
β-catenin is a molecule belonging to the armadillo family of proteins that is a crucial core-component of cellular adherens junctions, and a component of the canonical Wnt-signaling pathway. We attempted to analyze the functional significance of ectodermal-derived β-catenin during the development of the mouse genital tubercle, a mammalian anlage of the external genitalia. For this purpose, the conditional loss of function mouse mutant Wnt7a-Cre;β-cat(f/f) was utilized. Loss of ectodermal β-catenin leads to the formation of urethral cleft during preputial uprising. Although expression of E-cadherin was retained in the genital tubercle ectoderm of mutants, probably through plakoglobin compensatory expression, expression of other crucial adherens junction components such as α-catenin and F-actin in the cell-cell border were distinctly reduced. We also showed that β-catenin is necessary for the expression of its transcriptional downstream target Lef-1 which was localized in the basal layer of the preputial ectoderm, excluding the midventral region at E15.5. Such specialized region was observed to possess cytoplasmic β-catenin expression at this stage. Coincidentally, mitotically active cells were also found in the basal layer of the preputial ectoderm excluding the midventral region. In mutant genital tubercle, cell proliferation in the preputial ectoderm was decreased. Taken together, we suggest that ectodermal β-catenin is necessary not only to maintain adherens junction integrity, but also to regulate cell proliferation possibly through Lef-1 functions. Thus, β-catenin is shown to perform dual functions, initially as an adhesion molecule and later on as a possible transcription factor.
Collapse
Affiliation(s)
- Ahmad Reza Mazahery
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ohyama M, Veraitch O. Strategies to enhance epithelial-mesenchymal interactions for human hair follicle bioengineering. J Dermatol Sci 2013; 70:78-87. [PMID: 23557720 DOI: 10.1016/j.jdermsci.2013.02.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 02/11/2013] [Accepted: 02/13/2013] [Indexed: 12/17/2022]
Abstract
Hair follicle morphogenesis and regeneration depend on intensive but well-orchestrated interactions between epithelial and mesenchymal components. Accordingly, the enhancement of this crosstalk represents a promising approach to achieve successful bioengineering of human hair follicles. The present article summarizes the techniques, both currently available and potentially feasible, to promote epithelial-mesenchymal interactions (EMIs) necessary for human hair follicle regeneration. The strategies include the preparation of epithelial components with high receptivity to trichogenic dermal signals and/or mesenchymal cell populations with potent hair inductive capacity. In this regard, bulge epithelial stem cells, keratinocytes predisposed to hair follicle fate or keratinocyte precursor cells with plasticity may provide favorable epithelial cell populations. Dermal papilla cells sustaining intrinsic hair inductive capacity, putative dermal papilla precursor cells in the dermal sheath/neonatal dermis or trichogenic dermal cells derived from undifferentiated stem/progenitor cells are promising candidates as hair inductive dermal cells. The most established protocol for in vivo hair follicle reconstitution is co-grafting of epithelial and mesenchymal components into immunodeficient mice. In theory, combination of individually optimized cellular components of respective lineages should elicit most intensive EMIs to form hair follicles. Still, EMIs can be further ameliorated by the modulation of non-cell autonomous conditions, including cell compartmentalization to replicate the positional relationship in vivo and humanization of host environment by preparing human stromal bed. These approaches may not always synergistically intensify EMIs, however, step-by-step investigation probing optimal combinations should maximally enhance EMIs to achieve successful human hair follicle bioengineering.
Collapse
Affiliation(s)
- Manabu Ohyama
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan.
| | | |
Collapse
|
29
|
Suzuki K, Yokoyama C, Higashi Y, Daikoku T, Mizoguchi S, Saika S, Yamada G. Wakayama Symposium: Epithelial-Mesenchymal Interaction Regulates Tissue Formation and Characteristics: Insights for Corneal Development. Ocul Surf 2012; 10:217-20. [DOI: 10.1016/j.jtos.2012.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/06/2012] [Accepted: 07/01/2012] [Indexed: 11/16/2022]
|
30
|
β-Catenin signaling regulates Foxa2 expression during endometrial hyperplasia formation. Oncogene 2012; 32:3477-82. [PMID: 22945641 DOI: 10.1038/onc.2012.376] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 06/20/2012] [Accepted: 07/09/2012] [Indexed: 01/10/2023]
Abstract
The Wnt/β-catenin signaling is essential for various organogenesis and is often implicated during tumorigenesis. Dysregulated β-catenin signaling is associated with the formation of endometrial adenocarcinomas (EACs), which is considered as the common form of endometrial cancer in women. In the current study, we investigate the downstream target of Wnt/β-catenin signaling in the uterine epithelia and the mechanism leading to the formation of endometrial hyperplasia. We report that conditional ablation and activation of β-catenin in the uterine epithelia lead to aberrant epithelial structures and endometrial hyperplasia formation, respectively. We demonstrate that β-catenin regulates Foxa2 with its candidate upstream region for the uterine epithelia. Furthermore, knockdown of Foxa2 leads to defects in cell cycle regulation, suggesting a possible function of Foxa2 in the control of cell proliferation. We also observe that β-catenin and Foxa2 expression levels are augmented in the human specimens of complex atypical endometrial hyperplasia, which is considered to have a greater risk of progression to EACs. Thus, our study indicates that β-catenin regulates Foxa2 expression, and this interaction is possibly essential to control cell cycle progression during endometrial hyperplasia formation. Altogether, the augmented expression levels of β-catenin and Foxa2 are essential features during the formation of endometrial hyperplasia.
Collapse
|
31
|
Berthon A, Martinez A, Bertherat J, Val P. Wnt/β-catenin signalling in adrenal physiology and tumour development. Mol Cell Endocrinol 2012; 351:87-95. [PMID: 21930188 DOI: 10.1016/j.mce.2011.09.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 08/16/2011] [Accepted: 09/05/2011] [Indexed: 01/12/2023]
Abstract
Wnt/β-catenin signalling plays essential roles during embryonic development and in adult tissue homeostasis. Canonical signalling through Wnt secreted ligands relies on the control of β-catenin cytoplasmic accumulation and translocation to the nucleus. In this compartment, β-catenin serves as a transcription coactivator for transcription factors such as Lef/Tcf or some nuclear receptors. Constitutive Wnt signalling resulting from inactivation of inhibitors of the pathway or from activating mutations in β-catenin, triggers tumour development in a number of tissues. Analysis of patients' samples and genetically engineered mouse models has shown that Wnt signalling was involved in adrenal development and tumourigenesis. This review will summarise all these recent findings and will focus on some of the mechanisms that may lead to aberrant accumulation of β-catenin in adrenocortical tumours.
Collapse
Affiliation(s)
- Annabel Berthon
- CNRS UMR6247, Génétique Reproduction et Développement, Clermont Université, Aubière, France
| | | | | | | |
Collapse
|
32
|
Cui CY, Kunisada M, Childress V, Michel M, Schlessinger D. Shh is required for Tabby hair follicle development. Cell Cycle 2011; 10:3379-86. [PMID: 21926481 DOI: 10.4161/cc.10.19.17669] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In embryonic Eda mutant ("Tabby") mice, the development of one of the two major types of hair, "primary" hair fails, but other "secondary" hairs develop in normal numbers, though shorter and slightly aberrant. In Tabby mice, Shh is undetectable in skin early on, but is activated during secondary hair formation. We inferred that Shh may be involved in primary hair formation, activated normally by Eda, and also possibly in secondary hair formation, activated by an Eda-independent pathway. Varying the dosage of Shh now supports these inferences. In Shh knockout mice, mice were totally hairless: primary and secondary hair follicle germs were formed, but further progression failed. Consistent with these findings, when Shh loss was restricted to the skin, secondary hair follicle germs were initiated on time in Tabby mice, but their subsequent development (down-growth) failed. An Shh transgene expressed in Tabby skin could not restore induction of primary hair follicles, but restored normal length to the somewhat aberrant secondary hair that was formed and prolonged the anagen phase of hair cycling. Thus, Shh is required for primary and secondary hair down-growth and full secondary hair length, but is not itself sufficient to replace Eda or make fully normal secondary hair.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| | | | | | | | | |
Collapse
|
33
|
Wang X, Tredget EE, Wu Y. Dynamic signals for hair follicle development and regeneration. Stem Cells Dev 2011; 21:7-18. [PMID: 21787229 DOI: 10.1089/scd.2011.0230] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hair follicles form during embryonic development and, after birth, undergo recurrent cycling of growth, regression, and relative quiescence. As a functional mini-organ, the hair follicle develops in an environment with dynamic and alternating changes of diverse molecular signals. Over the past decades, genetically engineered mouse models have been used to study hair follicle morphogenesis and significant advances have been made toward the identification of key signaling pathways and the regulatory genes involved. In contrast, much less is understood in signals regulating hair follicle regeneration. Like hair follicle development, hair follicle regeneration probably relies on populations of stem cells that undergo a highly coordinated and stepwise program of differentiation to produce the completed structure. Here, we review recent advances in the understanding of the molecular signals underlying hair follicle morphogenesis and regeneration, with a focus on the initiation of the primary hair follicle structure placode. Knowledge about hair follicle morphogenesis may help develop novel therapeutic strategies to enhance cutaneous regeneration and improve wound healing.
Collapse
Affiliation(s)
- Xusheng Wang
- Life Science Division, Tsinghua University Graduate School at Shenzhen, China
| | | | | |
Collapse
|
34
|
Luderer HF, Gori F, Demay MB. Lymphoid enhancer-binding factor-1 (LEF1) interacts with the DNA-binding domain of the vitamin D receptor. J Biol Chem 2011; 286:18444-51. [PMID: 21471213 DOI: 10.1074/jbc.m110.188219] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ligand-independent actions of the vitamin D receptor (VDR) are required for normal post-morphogenic hair cycles; however, the molecular mechanisms by which the VDR exerts these actions are not clear. Previous studies demonstrated impaired regulation of the canonical Wnt signaling pathway in primary keratinocytes lacking the VDR. To identify the key effector of canonical Wnt signaling that interacts with the VDR, GST pulldown studies were performed. A novel interaction between the VDR and LEF1 (lymphoid enhancer-binding factor-1) that is independent of β-catenin was identified. This interaction is dependent upon sequences within the N-terminal region of the VDR, a domain required for VDR-DNA interactions and normal hair cycling in mice. Mutation of specific residues within the N-terminal region of the VDR not only abrogated interactions between the VDR and LEF1 but also impaired the ability of the VDR to enhance Wnt signaling in vdr(-/-) primary keratinocytes. Thus, this study demonstrates a novel interaction between the VDR and LEF1 that is mediated by the DNA-binding domain of the VDR and that is required for normal canonical Wnt signaling in keratinocytes.
Collapse
Affiliation(s)
- Hilary F Luderer
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
35
|
Matsumaru D, Haraguchi R, Miyagawa S, Motoyama J, Nakagata N, Meijlink F, Yamada G. Genetic analysis of Hedgehog signaling in ventral body wall development and the onset of omphalocele formation. PLoS One 2011; 6:e16260. [PMID: 21283718 PMCID: PMC3024424 DOI: 10.1371/journal.pone.0016260] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 12/12/2010] [Indexed: 01/03/2023] Open
Abstract
Background An omphalocele is one of the major ventral body wall malformations and
is characterized by abnormally herniated viscera from the body trunk. It has
been frequently found to be associated with other structural malformations,
such as genitourinary malformations and digit abnormalities. In spite of its
clinical importance, the etiology of omphalocele formation is still controversial.
Hedgehog (Hh) signaling is one of the essential growth factor signaling pathways
involved in the formation of the limbs and urogenital system. However, the
relationship between Hh signaling and ventral body wall formation remains
unclear. Methodology/Principal Findings To gain insight into the roles of Hh signaling in ventral body wall formation
and its malformation, we analyzed phenotypes of mouse mutants of Sonic
hedgehog (Shh), GLI-Kruppel family member
3 (Gli3) and Aristaless-like homeobox 4
(Alx4). Introduction of additional Alx4Lst
mutations into the Gli3Xt/Xt background resulted
in various degrees of severe omphalocele and pubic diastasis. In addition,
loss of a single Shh allele restored the omphalocele and
pubic symphysis of Gli3Xt/+; Alx4Lst/Lst
embryos. We also observed ectopic Hh activity in the ventral body wall region
of Gli3Xt/Xt embryos. Moreover, tamoxifen-inducible
gain-of-function experiments to induce ectopic Hh signaling revealed Hh signal
dose-dependent formation of omphaloceles. Conclusions/Significance We suggest that one of the possible causes of omphalocele and pubic diastasis
is ectopically-induced Hh signaling. To our knowledge, this would be the first
demonstration of the involvement of Hh signaling in ventral body wall malformation
and the genetic rescue of omphalocele phenotypes.
Collapse
Affiliation(s)
- Daisuke Matsumaru
- Global COE "Cell Fate Regulation
Research and Education Unit", Department of Organ Formation, Institute of
Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
| | - Ryuma Haraguchi
- Global COE "Cell Fate Regulation
Research and Education Unit", Department of Organ Formation, Institute of
Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
| | - Shinichi Miyagawa
- Global COE "Cell Fate Regulation
Research and Education Unit", Department of Organ Formation, Institute of
Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
| | - Jun Motoyama
- Department of Medical Life Systems,
Doshisha University, Kyoto, Japan
| | - Naomi Nakagata
- Center for Animal Resources and
Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Frits Meijlink
- Hubrecht Institute, KNAW and University
Medical Center, Utrecht, The Netherlands
| | - Gen Yamada
- Global COE "Cell Fate Regulation
Research and Education Unit", Department of Organ Formation, Institute of
Molecular Embryology and Genetics (IMEG), Kumamoto University, Kumamoto, Japan
- * E-mail:
| |
Collapse
|
36
|
Maklad A, Conway M, Hodges C, Hansen LA. Development of innervation to maxillary whiskers in mice. Anat Rec (Hoboken) 2010; 293:1553-67. [PMID: 20648571 DOI: 10.1002/ar.21194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The maxillary vibrissal pad is a unique, richly innervated sensory apparatus. It is highly evolved in the rodent that it constitutes a major source of sensory information to the somatosensory cortex. In this report, indocarbocyanine tracing and immunofluorescence were used to study the embryonic and early neonatal development of innervation to maxillary vibrissal follicles in mice. The first sign of vibrissal follicle innervation occurred at embryonic day 12 (E12), when the lateral nasal and maxillary processes were penetrated by nerve branches with small terminal plexuses assuming the positions of vibrissal follicle primordia. Between E13 and E15, the nerve plexuses at the presumptive follicles grew in size and became more numerous with no signs of specific receptor subtype formation. By E17, the nerve plexuses had grown further in size and the region-specific receptor subtype specification developed. At birth (P0), the superficial vibrissal nerves began to innervate the apical part of the inner conical body, whereas the deep vibrissal nerve gave off the recurrent cavernous branches. At P3, all of the different sets of receptor subtypes had regional distributions, densities and morphologies comparable to those described in adult mice. A 3-day old mouse had all complements of sensory receptors necessary for somatosensory transduction as revealed not only by neuroanatomic tracing but also with immunofluorescence for several markers of neurosensory differentiation. Our data reveal a hitherto unknown time table for the development of peripheral sensory receptors in the vibrissal follicles. This time table parallels that of their central targets in the somatosensory barrel cortex, which develops at P4.
Collapse
Affiliation(s)
- Adel Maklad
- Department of Anatomy, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | |
Collapse
|
37
|
Cui CY, Kunisada M, Piao Y, Childress V, Ko MSH, Schlessinger D. Dkk4 and Eda regulate distinctive developmental mechanisms for subtypes of mouse hair. PLoS One 2010; 5:e10009. [PMID: 20386733 PMCID: PMC2850388 DOI: 10.1371/journal.pone.0010009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 03/08/2010] [Indexed: 12/19/2022] Open
Abstract
The mouse hair coat comprises protective “primary” and thermo-regulatory “secondary” hairs. Primary hair formation is ectodysplasin (Eda) dependent, but it has been puzzling that Tabby (Eda-/y) mice still make secondary hair. We report that Dickkopf 4 (Dkk4), a Wnt antagonist, affects an auxiliary pathway for Eda-independent development of secondary hair. A Dkk4 transgene in wild-type mice had no effect on primary hair, but secondary hairs were severely malformed. Dkk4 action on secondary hair was further demonstrated when the transgene was introduced into Tabby mice: the usual secondary follicle induction was completely blocked. The Dkk4-regulated secondary hair pathway, like the Eda-dependent primary hair pathway, is further mediated by selective activation of Shh. The results thus reveal two complex molecular pathways that distinctly regulate subtype-based morphogenesis of hair follicles, and provide a resolution for the longstanding puzzle of hair formation in Tabby mice lacking Eda.
Collapse
Affiliation(s)
- Chang-Yi Cui
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Makoto Kunisada
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yulan Piao
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Victoria Childress
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Minoru S. H. Ko
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Abstract
Mammalian epidermis is maintained by self-renewal of stem cells and terminal differentiation of their progeny. New data reveal a diversity amongst stem cells that was previously unrecognized. Different stem cell populations have different locations and differ in whether they are quiescent or actively cycling. During normal epidermal homeostasis, each stem cell population feeds a restricted number of differentiated lineages. However, in response to injury or genetic manipulation the different pools of stem cells demonstrate multi-lineage differentiation ability. While it is well established that Wnt signalling promotes hair follicle (HF) differentiation, new observations suggest a role for EGF receptor signalling in promoting differentiation of interfollicular epidermis. NFATc1 maintains quiescence in the HF, while Lrig1 exerts the same function in the junctional zone. The stage is now set for exploring the relationship between the different epidermal stem cell populations and between quiescence and lineage selection.
Collapse
Affiliation(s)
- Fiona M Watt
- Wellcome Trust Centre for Stem Cell Research, Tennis Court Road, Cambridge, UK.
| | | |
Collapse
|
39
|
Araújo R, Fernandes M, Cavaco-Paulo A, Gomes A. Biology of human hair: know your hair to control it. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 125:121-43. [PMID: 21072698 DOI: 10.1007/10_2010_88] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hair can be engineered at different levels--its structure and surface--through modification of its constituent molecules, in particular proteins, but also the hair follicle (HF) can be genetically altered, in particular with the advent of siRNA-based applications. General aspects of hair biology are reviewed, as well as the most recent contributions to understanding hair pigmentation and the regulation of hair development. Focus will also be placed on the techniques developed specifically for delivering compounds of varying chemical nature to the HF, indicating methods for genetic/biochemical modulation of HF components for the treatment of hair diseases. Finally, hair fiber structure and chemical characteristics will be discussed as targets for keratin surface functionalization.
Collapse
Affiliation(s)
- Rita Araújo
- CBMA-Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus of Gualtar, 4710-057, Braga, Portugal
| | | | | | | |
Collapse
|
40
|
Antagonistic crosstalk of Wnt/beta-catenin/Bmp signaling within the Apical Ectodermal Ridge (AER) regulates interdigit formation. Biochem Biophys Res Commun 2009; 391:1653-7. [PMID: 20043884 DOI: 10.1016/j.bbrc.2009.12.109] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 12/17/2009] [Indexed: 12/14/2022]
Abstract
Digit and interdigit (D/ID) development is one of the important research fields in molecular developmental biology. Interdigital cell death (ICD) is a morphogenetic event which has been considered as an essential process for D/ID formation. Although some growth factors including Bmp and Fgf signaling can modulate ICD, growth factor crosstalk regulating ICD is poorly understood. Wnt canonical pathway and Bmp signal crosstalk has been considered as the essential growth factor crosstalk in organogenesis. To elucidate the crosstalk to regulate the D/ID formation, we analyzed conditional mutant mice with limb bud ectoderm expressing constitutively activated beta-catenin signaling. We showed that modulation of Wnt/beta-catenin signal in the limb ectoderm including the AER regulates ID apoptosis. We also demonstrated that Wnt/beta-catenin signaling in the ectoderm can positively regulate Fgf8 possibly antagonizing the epithelial derived Bmp signaling. Human birth defects for digit abnormalities have been known to be affected by multiple parameters. Elucidation of the potential mechanisms underlying such D/ID development is an urgent medical issue to be solved. This work would be one of the first studies showing essential growth factor cascades in the D/ID formation.
Collapse
|
41
|
Van Raamsdonk CD. Hereditary hair loss and the ancient signaling pathways that regulate ectodermal appendage formation. Clin Genet 2009; 76:332-40. [DOI: 10.1111/j.1399-0004.2009.01243.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
42
|
Beites CL, Hollenbeck PLW, Kim J, Lovell-Badge R, Lander AD, Calof AL. Follistatin modulates a BMP autoregulatory loop to control the size and patterning of sensory domains in the developing tongue. Development 2009; 136:2187-97. [PMID: 19474151 DOI: 10.1242/dev.030544] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The regenerative capacity of many placode-derived epithelial structures makes them of interest for understanding the molecular control of epithelial stem cells and their niches. Here, we investigate the interaction between the developing epithelium and its surrounding mesenchyme in one such system, the taste papillae and sensory taste buds of the mouse tongue. We identify follistatin (FST) as a mesenchymal factor that controls size, patterning and gustatory cell differentiation in developing taste papillae. FST limits expansion and differentiation of Sox2-expressing taste progenitor cells and negatively regulates the development of taste papillae in the lingual epithelium: in Fst(-/-) tongue, there is both ectopic development of Sox2-expressing taste progenitors and accelerated differentiation of gustatory cells. Loss of Fst leads to elevated activity and increased expression of epithelial Bmp7; the latter effect is consistent with BMP7 positive autoregulation, a phenomenon we demonstrate directly. We show that FST and BMP7 influence the activity and expression of other signaling systems that play important roles in the development of taste papillae and taste buds. In addition, using computational modeling, we show how aberrations in taste papillae patterning in Fst(-/-) mice could result from disruption of an FST-BMP7 regulatory circuit that normally suppresses noise in a process based on diffusion-driven instability. Because inactivation of Bmp7 rescues many of the defects observed in Fst(-/-) tongue, we conclude that interactions between mesenchyme-derived FST and epithelial BMP7 play a central role in the morphogenesis, innervation and maintenance of taste buds and their stem/progenitor cells.
Collapse
Affiliation(s)
- Crestina L Beites
- Department of Anatomy and Neurobiology and Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | | | | | | | | | | |
Collapse
|