1
|
张 悦, 汤 炜, 田 卫, 于 湄. [Research progress in regulation of hair growth by dermal adipose tissue]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:626-632. [PMID: 38752252 PMCID: PMC11096881 DOI: 10.7507/1002-1892.202402092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/11/2024] [Indexed: 05/18/2024]
Abstract
Objective To summarize the dynamic and synchronized changes between the hair cycle and dermal adipose tissue as well as the impact of dermal adipose tissue on hair growth, and to provide a new research idea for the clinical treatment of hair loss. Methods An extensive review of relevant literature both domestic and international was conducted, analyzing and summarizing the impact of dermal adipose precursor cells, mature dermal adipocytes, and the processes of adipogenesis in dermal adipose tissue on the transition of hair cycle phases. Results Dermal adipose tissue is anatomically adjacent to hair follicles and closely related to the changes in the hair cycle. The proliferation and differentiation of dermal adipose precursor cells promote the transition of hair cycle from telogen to anagen, while mature adipocytes can accelerate the transition from anagen to catagen of the hair cycle by expressing signaling molecules, with adipogenesis in dermal adipose tissue and hair cycle transition signaling coexistence. Conclusion Dermal adipose tissue affects the transition of the hair cycle and regulates hair growth by secreting various signaling molecules. However, the quantity and depth of existing literature are far from sufficient to fully elucidate its prominent role in regulating the hair cycle, and the specific regulatory mechanisms needs to be further studied.
Collapse
Affiliation(s)
- 悦 张
- 四川大学华西口腔医学院 口腔再生医学国家地方联合工程实验室(成都 610041)National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
- 四川大学华西口腔医院口腔颌面创伤整形外科(成都 610041)Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| | - 炜 汤
- 四川大学华西口腔医学院 口腔再生医学国家地方联合工程实验室(成都 610041)National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| | - 卫东 田
- 四川大学华西口腔医学院 口腔再生医学国家地方联合工程实验室(成都 610041)National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
- 四川大学华西口腔医院口腔颌面创伤整形外科(成都 610041)Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| | - 湄 于
- 四川大学华西口腔医学院 口腔再生医学国家地方联合工程实验室(成都 610041)National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| |
Collapse
|
2
|
Marinho PA, Jeong G, Shin SH, Kim SN, Choi H, Lee SH, Park BC, Hong YD, Kim HJ, Park WS. The development of an in vitrohuman hair follicle organoid with a complexity similar to that in vivo. Biomed Mater 2024; 19:025041. [PMID: 38324888 DOI: 10.1088/1748-605x/ad2707] [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: 09/01/2023] [Accepted: 02/07/2024] [Indexed: 02/09/2024]
Abstract
In vitrohair follicle (HF) models are currently limited toex vivoHF organ cultures (HFOCs) or 2D models that are of low availability and do not reproduce the architecture or behavior of the hair, leading to poor screening systems. To resolve this issue, we developed a technology for the construction of a humanin vitrohair construct based on the assemblage of different types of cells present in the hair organ. First, we demonstrated that epithelial cells, when isolatedin vitro, have similar genetic signatures regardless of their dissection site, and their trichogenic potential is dependent on the culture conditions. Then, using cell aggregation techniques, 3D spheres of dermal papilla (DP) were constructed, and subsequently, epithelial cells were added, enabling the production and organization of keratins in hair, similar to what is seenin vivo. These reconstructed tissues resulted in the following hair compartments: K71 (inner root-sheath), K85 (matrix region), K75 (companion layer), and vimentin (DP). Furthermore, the new hair model was able to elongate similarly toex vivoHFOC, resulting in a shaft-like shape several hundred micrometers in length. As expected, when the model was exposed to hair growth enhancers, such as ginseng extract, or inhibitors, such as TGF-B-1, significant effects similar to thosein vivowere observed. Moreover, when transplanted into skin biopsies, the new constructs showed signs of integration and hair bud generation. Owing to its simplicity and scalability, this model fully enables high throughput screening of molecules, which allows understanding of the mechanism by which new actives treat hair loss, finding optimal concentrations, and determining the synergy and antagonism among different raw materials. Therefore, this model could be a starting point for applying regenerative medicine approaches to treat hair loss.
Collapse
Affiliation(s)
| | - Gyusang Jeong
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Seung Hyun Shin
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Su Na Kim
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Hyeongwon Choi
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Sung Hoon Lee
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Byung Cheol Park
- Department of Dermatology, College of Medicine, Dankook University, Cheonan-si, Republic of Korea
| | - Yong Deog Hong
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Hyoung-June Kim
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| | - Won-Seok Park
- AMOREPACIFIC Research and Innovation Center, Yongin-si, Republic of Korea
| |
Collapse
|
3
|
Wang X, Plikus MV. Aged Skin Cells Nurture Stem Cells toward Regeneration. J Invest Dermatol 2024; 144:11-14. [PMID: 37897482 DOI: 10.1016/j.jid.2023.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 10/30/2023]
Affiliation(s)
- Xiaojie Wang
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
4
|
Wu W, Zhou W, Jiang J, Wang M, Zhang J, Yang J, Tang Q, Liu H, Liu D, Xu W, Zhong JL, Yang L, Lei M. Mechanical stimuli-induced CCL2 restores adult mouse cells to regenerate hair follicles. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:94-110. [PMID: 37020681 PMCID: PMC10068016 DOI: 10.1016/j.omtn.2023.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
Aged cells have declined regenerative ability when subjected to environmental insult. Here we elucidate the mechanism by which mechanical stimulus induces hair regeneration at the microenvironmental regulation level using the hair plucking and organoid culture models. We observed that the skin cells harvested from post-plucking day 3 (PPD3) have the best self-organizing ability during skin organoid culture and have the highest hair regeneration upon transplantation. By bulk RNA sequencing (RNA-seq) and single-cell RNA-seq analysis and in situ hybridization, we identified that the chemokine signaling pathway genes including CCL2 are significantly increased in the skin at PPD3 and in skin organoid cultures. Immunostaining shows that the PPD3 skin epithelial cells have increased multipotency, which is verified by the ability to self-organize to form epidermal aggregates during organoid culture. By adding CCL2 recombinant protein to the organoid culture using an environmental reprogramming protocol, we observed the PPD0 adult skin cells, which lose their regenerative ability can self-organize in organoid culture and regenerate hair follicles robustly upon transplantation. Our study demonstrates that CCL2 functions in immune regulation of hair regeneration under mechanical stimulus, and enhances cell multipotency during organoid culture. This provides a therapeutic potential for future clinical application.
Collapse
Affiliation(s)
- Wang Wu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Wei Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jingwei Jiang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mengyue Wang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jinwei Zhang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jing Yang
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Qu Tang
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Huawen Liu
- Three Gorges Hospital, Chongqing University, Chongqing 404000, China
| | - Deming Liu
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Dermatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Wei Xu
- Department of Dermatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Julia Li Zhong
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Li Yang
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Mingxing Lei
- 111 Project Laboratory of Biomechanics and Tissue Repair & Key Laboratory of Biorheological Science and Technology of Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
5
|
Liu Y, Guerrero-Juarez CF, Xiao F, Shettigar NU, Ramos R, Kuan CH, Lin YC, de Jesus Martinez Lomeli L, Park JM, Oh JW, Liu R, Lin SJ, Tartaglia M, Yang RB, Yu Z, Nie Q, Li J, Plikus MV. Hedgehog signaling reprograms hair follicle niche fibroblasts to a hyper-activated state. Dev Cell 2022; 57:1758-1775.e7. [PMID: 35777353 PMCID: PMC9344965 DOI: 10.1016/j.devcel.2022.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 03/10/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
Hair follicle stem cells are regulated by dermal papilla fibroblasts, their principal signaling niche. Overactivation of Hedgehog signaling in the niche dramatically accelerates hair growth and induces follicle multiplication in mice. On single-cell RNA sequencing, dermal papilla fibroblasts increase heterogeneity to include new Wnt5ahigh states. Transcriptionally, mutant fibroblasts activate regulatory networks for Gli1, Alx3, Ebf1, Hoxc8, Sox18, and Zfp239. These networks jointly upregulate secreted factors for multiple hair morphogenesis and hair-growth-related pathways. Among these is non-conventional TGF-β ligand Scube3. We show that in normal mouse skin, Scube3 is expressed only in dermal papillae of growing, but not in resting follicles. SCUBE3 protein microinjection is sufficient to induce new hair growth, and pharmacological TGF-β inhibition rescues mutant hair hyper-activation phenotype. Moreover, dermal-papilla-enriched expression of SCUBE3 and its growth-activating effect are partially conserved in human scalp hair follicles. Thus, Hedgehog regulates mesenchymal niche function in the hair follicle via SCUBE3/TGF-β mechanism.
Collapse
Affiliation(s)
- Yingzi Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Fei Xiao
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Nitish Udupi Shettigar
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Amplifica Holdings Group, Inc., San Diego, CA 92128, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Chen-Hsiang Kuan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Jung Min Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea; Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ji Won Oh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea; Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea; Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea
| | - Ruiqi Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Sung-Jan Lin
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Engineering and Department of Dermatology, National Taiwan University, Taipei, Taiwan
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome 00146, Italy
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
6
|
Dinh K, Wang Q. A probabilistic Boolean model on hair follicle cell fate regulation by TGF-β. Biophys J 2022; 121:2638-2652. [PMID: 35714600 PMCID: PMC9300639 DOI: 10.1016/j.bpj.2022.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
Hair follicles (HFs) are mini skin organs that undergo cyclic growth. Various signals regulate HF cell fate decisions jointly. Recent experimental results suggest that transforming growth factor beta (TGF-β) exhibits a dual role in HF cell fate regulation that can be either anti- or pro-apoptosis. To understand the underlying mechanisms of HF cell fate control, we develop a novel probabilistic Boolean network (pBN) model on the HF epithelial cell gene regulation dynamics. First, the model is derived from literature, then refined using single-cell RNA sequencing data. Using the model, we both explore the mechanisms underlying HF cell fate decisions and make predictions that could potentially guide future experiments: 1) we propose that a threshold-like switch in the TGF-β strength may necessitate the dual roles of TGF-β in either activating apoptosis or cell proliferation, in cooperation with bone morphogenetic protein (BMP) and tumor necrosis factor (TNF) and at different stages of a follicle growth cycle; 2) our model shows concordance with the high-activator-low-inhibitor theory of anagen initiation; 3) we predict that TNF may be more effective in catagen initiation than TGF-β, and they may cooperate in a two-step fashion; 4) finally, predictions of gene knockout and overexpression reveal the roles in HF cell fate regulations of each gene. Attractor and motif analysis from the associated Boolean networks reveal the relations between the topological structure of the gene regulation network and the cell fate regulation mechanism. A discrete spatial model equipped with the pBN illustrates how TGF-β and TNF cooperate in initiating and driving the apoptosis wave during catagen.
Collapse
Affiliation(s)
- Katherine Dinh
- Department of Biology, University of California, Riverside, California
| | - Qixuan Wang
- Department of Mathematics, University of California, Riverside, California; Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, California.
| |
Collapse
|
7
|
da Cruz GK, Martins MIM, Antunes FTT, de Souza AH, Wiilland EDF, Picada JN, Brum LFDS. Evaluation of the efficacy and toxicity of oral and topical pumpkin oil on the hair growth of mice. Acta Histochem 2022; 124:151894. [PMID: 35447441 DOI: 10.1016/j.acthis.2022.151894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/11/2022]
Abstract
This study aimed to evaluate the efficacy and safety of a topical and oral administration of pumpkin seed oil (PSO) on the hair growth of BALB/c male mice. The animals had their dorsal area shaved (2 ×2 cm) and they were divided into 6 experimental groups. They received orally saline (OS), finasteride (F), or PSO (OP) for 14 days; or topically saline (TS), minoxidil (M), or PSO (TP) for 7 days. The euthanasia of all of the mice occurred on the 22nd day, and the histological slides from the skin area were analyzed. Lipoperoxidation in the liver was assessed through the TBARS method and was also evaluated by the antioxidant enzymes (SOD and CAT). The comet assay and the micronucleus tests were performed for genotoxic/mutagenic safety analyses. A significant increase in the number of hair follicles in the TP group was seen (8.8 ± 0.8) but it was disorganized, with loose dermal collagen. Finasteride presented a significant increase in the levels of the TBARS, SOD, and CAT in the liver, and M increased the DNA damage in the blood and the liver tissues. PSO did not induce any significant changes. In addition, PSO did not induce genotoxic or mutagenic effects. In conclusion, the oral PSO for 14 days acted in the proliferation of the hair follicles, without toxicity signals in the liver. DATA AVAILABILITY: The authors confirm that all of the relevant data is included in the article and/or in the supplementary information file.
Collapse
|
8
|
Chen P, Zhang F, Fan Z, Shen T, Liu B, Chen R, Qu Q, Wang J, Miao Y, Hu Z. Nanoscale microenvironment engineering for expanding human hair follicle stem cell and revealing their plasticity. J Nanobiotechnology 2021; 19:94. [PMID: 33789665 PMCID: PMC8010974 DOI: 10.1186/s12951-021-00840-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
Background Periodically regenerated hair follicles provide an excellent research model for studying tissue regeneration and stem cell homeostasis. Periodic activation and differentiation of hair follicle stem cells (HFSCs) fuel cyclical bouts of hair regeneration. HFSCs represent an excellent paradigm for studying tissue regeneration and somatic stem cell homeostasis. However, these crucial studies are hampered by the lack of a culture system able to stably expand human HFSCs and regulate their fate. Results Here, we use layer-by-layer (LbL) self-assembly with gelatin/alginate to construct a nanoscale biomimetic extracellular matrix (ECM) for an HFSC population. The LbL coating provides ECM and mechanical support for individual cells, which helps to maintain the CD200+α6+ HFSC population to a certain extent. Addition of key signal molecules (FGF-7 and VEGF-A) simulates the minimum essential components of the stem cell microenvironment, thereby effectively and stably expanding HFSCs and maintaining the CD200+α6+ HFSC population. Subsequently, BMP2 loaded to the nanocoated layer, as a slow-release signal molecule, activates BMP signaling to regulate HFSCs’ fate in order to obtain a purified CD200+α6+ HFSC population. Conclusion This system can minimize the microenvironment of HFSCs; thus, stably amplifying HFSCs and revealing their plasticity. Our study thus provides a new tool for studies of hair follicle reconstruction and stem cell homeostasis. ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00840-5.
Collapse
Affiliation(s)
- Peng Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Feifei Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Zhexiang Fan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Tianding Shen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Bingcheng Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Ruosi Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Qian Qu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jin Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
9
|
Paus R. Shining a (blue) light on hair follicle chronobiology and photobiomodulation. Exp Dermatol 2021; 30:189-192. [PMID: 33433942 DOI: 10.1111/exd.14271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Ralf Paus
- Dr Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Centre for Dermatology Research, University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester, UK.,Monasterium Laboratory, Münster, Germany
| |
Collapse
|
10
|
Sha K, Chen M, Liu F, Xu S, Wang B, Peng Q, Zhang Y, Xie H, Li J, Deng Z. Platelet factor 4 inhibits human hair follicle growth and promotes androgen receptor expression in human dermal papilla cells. PeerJ 2020; 8:e9867. [PMID: 32953277 PMCID: PMC7476492 DOI: 10.7717/peerj.9867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
Platelet-rich plasma (PRP) has been reported recently as a potential therapeutic approach for alopecia, such as androgenetic alopecia, but the exact mechanisms and effects of specific components of this recipe remain largely unknown. In this study, we identified that platelet factor 4 (PF4), a component of PRP, significantly suppressed human hair follicle growth and restrained the proliferation of human dermal papilla cells (hDPCs). Furthermore, our results showed that PF4 upregulated androgen receptor (AR) in human dermal papilla cells in vitro and via hair follicle organ culture. Among the hair growth-promoting and DP-signature genes investigated, PF4 decreased the expression of Wnt5a, Wnt10b, LEF1, HEY1 and IGF-1, and increased DKK1 expression, but did not affect BMP2 and BMP4 expression. Collectively, Our data demonstrate that PF4 suppresses human hair follicle growth possibly via upregulating androgen receptor signaling and modulating hair growth-associated genes, which provides thought-provoking insights into the application and optimization of PRP in treating hair loss.
Collapse
Affiliation(s)
- Ke Sha
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China.,Xiangya International Academy of Translational Medicine, Central South University, Changsha, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Central South University, Changsha, China
| | - Fangfen Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ben Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Qinqin Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| |
Collapse
|
11
|
Peña‐Jimenez D, Fontenete S, Megias D, Fustero‐Torre C, Graña‐Castro O, Castellana D, Loewe R, Perez‐Moreno M. Lymphatic vessels interact dynamically with the hair follicle stem cell niche during skin regeneration in vivo. EMBO J 2019; 38:e101688. [PMID: 31475747 PMCID: PMC6769427 DOI: 10.15252/embj.2019101688] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/14/2022] Open
Abstract
Lymphatic vessels are essential for skin fluid homeostasis and immune cell trafficking. Whether the lymphatic vasculature is associated with hair follicle regeneration is, however, unknown. Here, using steady and live imaging approaches in mouse skin, we show that lymphatic vessels distribute to the anterior permanent region of individual hair follicles, starting from development through all cycle stages and interconnecting neighboring follicles at the bulge level, in a stem cell-dependent manner. Lymphatic vessels further connect hair follicles in triads and dynamically flow across the skin. At the onset of the physiological stem cell activation, or upon pharmacological or genetic induction of hair follicle growth, lymphatic vessels transiently expand their caliber suggesting an increased tissue drainage capacity. Interestingly, the physiological caliber increase is associated with a distinct gene expression correlated with lymphatic vessel reorganization. Using mouse genetics, we show that lymphatic vessel depletion blocks hair follicle growth. Our findings point toward the lymphatic vasculature being important for hair follicle development, cycling, and organization, and define lymphatic vessels as stem cell niche components, coordinating connections at tissue-level, thus provide insight into their functional contribution to skin regeneration.
Collapse
Affiliation(s)
- Daniel Peña‐Jimenez
- Epithelial Cell Biology GroupCancer Cell Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
| | - Silvia Fontenete
- Epithelial Cell Biology GroupCancer Cell Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
- Section of Cell Biology and PhysiologyDepartment of BiologyUniversity of CopenhagenCopenhagenDenmark
| | - Diego Megias
- Confocal Microscopy Core UnitBiotechnology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
| | - Coral Fustero‐Torre
- Bioinformatics UnitStructural Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
| | - Osvaldo Graña‐Castro
- Bioinformatics UnitStructural Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
| | - Donatello Castellana
- Epithelial Cell Biology GroupCancer Cell Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
- Center for Cooperative Research Biosciences (CIC bioGUNE)Derio BizkaiaSpain
| | - Robert Loewe
- Department of DermatologyMedical University of ViennaViennaAustria
| | - Mirna Perez‐Moreno
- Epithelial Cell Biology GroupCancer Cell Biology ProgrammeSpanish Cancer Research Centre (CNIO)MadridSpain
- Section of Cell Biology and PhysiologyDepartment of BiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
12
|
Lv X, Gao W, Jin C, Wang L, Wang Y, Chen W, Zou S, Huang S, Li Z, Wang J, Sun W. Preliminary study on microR-148a and microR-10a in dermal papilla cells of Hu sheep. BMC Genet 2019; 20:70. [PMID: 31455210 PMCID: PMC6712829 DOI: 10.1186/s12863-019-0770-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/08/2019] [Indexed: 12/14/2022] Open
Abstract
Background Hu sheep, a unique Chinese breed with high reproductive performance, are also well known for their rare white lambskin in China. The quality of lambskin is affected by hair follicles, and dermal papilla cells are an important component of hair follicles that plays a key role in hair follicle growth and development. This study helps elucidate the effect of miR-148a and miR-10a on hair follicle growth and development. Results Based on the results of gene chip and high-throughput sequencing, bone morphogenetic protein 7 (BMP7) was used as a research object. Bioinformatics analysis and the dual-luciferase reporter system indicated that, along with Western blot and quantitative real-time polymerase chain reaction (qRT-PCR) that miR-148a and miR-10a target relationships with BMP7. BMP7 was the target gene both for miR-148a and miR-10a by the dual-luciferase reporter system and Western blot. Hu sheep dermal papilla cells were successfully isolated and purified, and after transfecting miR-148a/miR-10a mimics and inhibitors into dermal papilla cells, a Cell Counting Kit-8 (CCK-8) was used to determine that miR-148a/miR-10a inhibited the proliferation of Hu sheep dermal papilla cells. In addition, after the overexpression of miR-148a, the expression levels of Smad3 (P < 0.05), Smad6 (P < 0.05), Smad4 (P < 0.01), and Smad5 (P < 0.01) were significantly higher than those of the control groups. After the inhibition of miR-148a, the expression levels of Smad3 (P < 0.05), Smad4 (P < 0.05), and TGF-β (P < 0.01) were significantly lower than those of the control groups. After the overexpression of miR-10a, the expression levels of Smad1 (P < 0.01), Smad2 (P < 0.05), Smad4 (P < 0.01), Smad5 (P < 0.01), and TGF-β (P < 0.05) were significantly lower than those of the control groups. After the inhibition of miR-10a, the expression levels of Smad1 (P < 0.01) and Smad2 (P < 0.05) were significantly lower than those of the control groups. Conclusions These results revealed the target relationship between miR-148a, miR-10a and BMP7, and the effect of miR-148a and miR-10a on the proliferation of dermal papilla cells. They will provide the basis for a follow-up study on how miR-148a, and miR-10a mediate BMP7 regulation of hair follicle growth and development.
Collapse
Affiliation(s)
- Xiaoyang Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Wen Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Chengyan Jin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Lihong Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yue Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Weihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Shuangxia Zou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Sainan Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Zhifeng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jinyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China. .,Joint international research laboratory of agriculture and agri - product safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
13
|
Plikus MV, Chuong CM. Understanding skin morphogenesis across developmental, regenerative and evolutionary levels. Exp Dermatol 2019; 28:327-331. [PMID: 30951234 DOI: 10.1111/exd.13932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California.,Center for Complex Biological Systems, University of California, Irvine, Irvine, California.,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California.,Integrative Stem Cell Center, China Medical University, Taichung, Taiwan.,International Wound Repair and Regenerative Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
14
|
Widelitz RB, Lin GW, Lai YC, Mayer JA, Tang PC, Cheng HC, Jiang TX, Chen CF, Chuong CM. Morpho-regulation in diverse chicken feather formation: Integrating branching modules and sex hormone-dependent morpho-regulatory modules. Dev Growth Differ 2018; 61:124-138. [PMID: 30569461 DOI: 10.1111/dgd.12584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
Many animals can change the size, shape, texture and color of their regenerated coats in response to different ages, sexes, or seasonal environmental changes. Here, we propose that the feather core branching morphogenesis module can be regulated by sex hormones or other environmental factors to change feather forms, textures or colors, thus generating a large spectrum of complexity for adaptation. We use sexual dimorphisms of the chicken to explore the role of hormones. A long-standing question is whether the sex-dependent feather morphologies are autonomously controlled by the male or female cell types, or extrinsically controlled and reversible. We have recently identified core feather branching molecular modules which control the anterior-posterior (bone morphogenetic orotein [BMP], Wnt gradient), medio-lateral (Retinoic signaling, Gremlin), and proximo-distal (Sprouty, BMP) patterning of feathers. We hypothesize that morpho-regulation, through quantitative modulation of existing parameters, can act on core branching modules to topologically tune the dimension of each parameter during morphogenesis and regeneration. Here, we explore the involvement of hormones in generating sexual dimorphisms using exogenously delivered hormones. Our strategy is to mimic male androgen levels by applying exogenous dihydrotestosterone and aromatase inhibitors to adult females and to mimic female estradiol levels by injecting exogenous estradiol to adult males. We also examine differentially expressed genes in the feathers of wildtype male and female chickens to identify potential downstream modifiers of feather morphogenesis. The data show male and female feather morphology and their color patterns can be modified extrinsically through molting and resetting the stem cell niche during regeneration.
Collapse
Affiliation(s)
- Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Gee-Way Lin
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Chih Lai
- Department of Pathology, University of Southern California, Los Angeles, California.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Julie A Mayer
- Department of Pathology, University of Southern California, Los Angeles, California.,Biocept Inc., San Diego, California
| | - Pin-Chi Tang
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Hsu-Chen Cheng
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Chih-Feng Chen
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California.,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, Taiwan.,The IEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
15
|
Hair Regeneration under Stress. J Invest Dermatol 2018; 138:1257-1259. [DOI: 10.1016/j.jid.2018.02.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
|
16
|
Luo J, Chen M, Liu Y, Xie H, Yuan J, Zhou Y, Ding J, Deng Z, Li J. Nature-derived lignan compound VB-1 exerts hair growth-promoting effects by augmenting Wnt/β-catenin signaling in human dermal papilla cells. PeerJ 2018; 6:e4737. [PMID: 29761053 PMCID: PMC5947041 DOI: 10.7717/peerj.4737] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/20/2018] [Indexed: 12/18/2022] Open
Abstract
Background Vitexin is a kind of lignan compound which has been shown to possess a variety of pharmacological effects, such as anti-inflammatory, anti-oxidative and anti-cancer activities. However the effect of vitexin on hair regeneration has not been elaborated. Methods The proliferation of human dermal papilla cells (hDPCs) was examined by cell counting and continuous cell culture after vitexin compound 1 (VB-1) was treated. The expression of lef1, wnt5a, bmp2, bmp4, alpl and vcan was examined by RT-PCR. The expression of dkk1, tgf-β1, active-β-Catenin, and AXIN2 was examined by RT-PCR or immunoblotting. Hair shaft growth was measured in the absence or presence of VB-1. Results We demonstrated that VB-1 significantly promotes the proliferation of hDPCs in a concentration-dependent manner within a certain concentration range. Among the hair growth-related genes investigated, dkk1 was clearly down-regulated in hDPCs treated with VB-1. The increased active β-Catenin and decreased AXIN2 protein levels suggest that VB-1 facilitates Wnt/β-catenin signaling in hDPCs in vitro. The expression of DP signature genes was also upregulated after VB-1 treatment. Our study further indicated that VB-1 promotes human hair follicle (HF) growth by HF organ culture assay. Discussion VB-1 may exert hair growth-promoting effects via augmenting Wnt/β-catenin signaling in hDPCs.
Collapse
Affiliation(s)
- Jieshu Luo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yingzi Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yingjun Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jinsong Ding
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| |
Collapse
|
17
|
Foster AR, Nicu C, Schneider MR, Hinde E, Paus R. Dermal white adipose tissue undergoes major morphological changes during the spontaneous and induced murine hair follicle cycling: a reappraisal. Arch Dermatol Res 2018; 310:453-462. [PMID: 29704126 DOI: 10.1007/s00403-018-1831-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 03/23/2018] [Accepted: 04/16/2018] [Indexed: 12/14/2022]
Abstract
In murine skin, dermal white adipose tissue (DWAT) undergoes major changes in thickness in synchrony with the hair cycle (HC); however, the underlying mechanisms remain unclear. We sought to elucidate whether increased DWAT thickness during anagen is mediated by adipocyte hypertrophy or adipogenesis, and whether lipolysis or apoptosis can explain the decreased DWAT thickness during catagen. In addition, we compared HC-associated DWAT changes between spontaneous and depilation-induced hair follicle (HF) cycling to distinguish between spontaneous and HF trauma-induced events. We show that HC-dependent DWAT remodelling is not an artefact caused by fluctuations in HF down-growth, and that dermal adipocyte (DA) proliferation and hypertrophy are HC-dependent, while classical DA apoptosis is absent. However, none of these changes plausibly accounts for HC-dependent oscillations in DWAT thickness. Contrary to previous studies, in vivo BODIPY uptake suggests that increased DWAT thickness during anagen occurs via hypertrophy rather than hyperplasia. From immunohistomorphometry, DWAT thickness likely undergoes thinning during catagen by lipolysis. Hence, we postulate that progressive, lipogenesis-driven DA hypertrophy followed by dynamic switches between lipogenesis and lipolysis underlie DWAT fluctuations in the spontaneous HC, and dismiss apoptosis as a mechanism of DWAT reduction. Moreover, the depilation-induced HC displays increased DWAT thickness, area, and DA number, but decreased DA volume/area compared to the spontaneous HC. Thus, DWAT shows additional, novel HF wounding-related responses during the induced HC. This systematic reappraisal provides important pointers for subsequent functional and mechanistic studies, and introduces the depilation-induced murine HC as a model for dissecting HF-DWAT interactions under conditions of wounding/stress.
Collapse
Affiliation(s)
- April R Foster
- Centre for Dermatology Research, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, and Manchester Academic Health Science Centre, Manchester, UK
| | - Carina Nicu
- Centre for Dermatology Research, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, and Manchester Academic Health Science Centre, Manchester, UK
| | - Marlon R Schneider
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich, Germany
| | - Eleanor Hinde
- Centre for Dermatology Research, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, and Manchester Academic Health Science Centre, Manchester, UK
| | - Ralf Paus
- Centre for Dermatology Research, The University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, and Manchester Academic Health Science Centre, Manchester, UK.
- Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
18
|
Abstract
Although the major white adipose depots evolved primarily to store energy, secrete hormones and thermo-insulate the body, multiple secondary depots developed additional specialized and unconventional functions. Unlike any other fat tissue, dermal white adipose tissue (dWAT) evolved a large repertoire of novel features that are central to skin physiology, which we discuss in this Review. dWAT exists in close proximity to hair follicles, the principal appendages of the skin that periodically grow new hairs. Responding to multiple hair-derived signals, dWAT becomes closely connected to cycling hair follicles and periodically cycles itself. At the onset of new hair growth, hair follicles secrete activators of adipogenesis, while at the end of hair growth, a reduction in the secretion of activators or potentially, an increase in the secretion of inhibitors of adipogenesis, results in fat lipolysis. Hair-driven cycles of dWAT remodelling are uncoupled from size changes in other adipose depots that are controlled instead by systemic metabolic demands. Rich in growth factors, dWAT reciprocally signals to hair follicles, altering the activation state of their stem cells and modulating the pace of hair regrowth. dWAT cells also facilitate skin repair following injury and infection. In response to wounding, adipose progenitors secrete repair-inducing activators, while bacteria-sensing adipocytes produce antimicrobial peptides, thus aiding innate immune responses in the skin.
Collapse
Affiliation(s)
- Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, 2011 Biological Sciences III, University of California, Irvine, Irvine, California 92697, USA
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, California 92697, USA
- Center for Complex Biological Systems, 2620 Biological Sciences III, University of California, Irvine, Irvine, California 92697, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, 2011 Biological Sciences III, University of California, Irvine, Irvine, California 92697, USA
- Sue and Bill Gross Stem Cell Research Center, 845 Health Sciences Road, University of California, Irvine, Irvine, California 92697, USA
- Center for Complex Biological Systems, 2620 Biological Sciences III, University of California, Irvine, Irvine, California 92697, USA
| |
Collapse
|
19
|
Abstract
Adipose tissue depots can exist in close association with other organs, where they assume diverse, often non-traditional functions. In stem cell-rich skin, bone marrow, and mammary glands, adipocytes signal to and modulate organ regeneration and remodeling. Skin adipocytes and their progenitors signal to hair follicles, promoting epithelial stem cell quiescence and activation, respectively. Hair follicles signal back to adipocyte progenitors, inducing their expansion and regeneration, as in skin scars. In mammary glands and heart, adipocytes supply lipids to neighboring cells for nutritional and metabolic functions, respectively. Adipose depots adjacent to skeletal structures function to absorb mechanical shock. Adipose tissue near the surface of skin and intestine senses and responds to bacterial invasion, contributing to the body's innate immune barrier. As the recognition of diverse adipose depot functions increases, novel therapeutic approaches centered on tissue-specific adipocytes are likely to emerge for a range of cancers and regenerative, infectious, and autoimmune disorders.
Collapse
Affiliation(s)
- Rachel K Zwick
- Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA
| | - Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA
| | - Valerie Horsley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, 845 Health Sciences Road, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
20
|
Skin and Its Regenerative Powers: An Alliance between Stem Cells and Their Niche. Dev Cell 2017; 43:387-401. [PMID: 29161590 DOI: 10.1016/j.devcel.2017.10.001] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/03/2017] [Accepted: 10/02/2017] [Indexed: 12/17/2022]
Abstract
Tissues have a natural capacity to replace dying cells and to heal wounds. This ability resides in resident stem cells, which self-renew, preserve, and repair their tissue during homeostasis and following injury. The skin epidermis and its appendages are subjected to daily assaults from the external environment. A high demand is placed on renewal and regeneration of the skin's barrier in order to protect the body from infection and dehydration and to heal wounds. This review focuses on the epithelial stem cells of skin, where they come from, where they reside, and how they function in normal homeostasis and wound repair.
Collapse
|
21
|
HOU CHUN, MIAO YONG, JI HANG, WANG SUSHENG, LIANG GANG, ZHANG ZHIHUA, HONG WEIJIN. 6-Gingerol inhibits hair cycle via induction of MMP2 and MMP9 expression. ACTA ACUST UNITED AC 2017; 89:2707-2717. [DOI: 10.1590/0001-3765201720170354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/02/2017] [Indexed: 02/02/2023]
Affiliation(s)
- CHUN HOU
- Guangzhou Medical University, P.R. China
| | - YONG MIAO
- Southern Medical University, P.R. China
| | - HANG JI
- Guangzhou Medical University, P.R. China
| | | | - GANG LIANG
- Guangzhou Medical University, P.R. China
| | | | | |
Collapse
|
22
|
Sardella C, Winkler C, Quignodon L, Hardman JA, Toffoli B, Giordano Attianese GMP, Hundt JE, Michalik L, Vinson CR, Paus R, Desvergne B, Gilardi F. Delayed Hair Follicle Morphogenesis and Hair Follicle Dystrophy in a Lipoatrophy Mouse Model of Pparg Total Deletion. J Invest Dermatol 2017; 138:500-510. [PMID: 28964716 DOI: 10.1016/j.jid.2017.09.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 09/08/2017] [Accepted: 09/08/2017] [Indexed: 01/06/2023]
Abstract
PPARγ regulates multiple aspects of skin physiology, including sebocyte differentiation, keratinocyte proliferation, epithelial stem cell survival, adipocyte biology, and inflammatory skin responses. However, the effects of its global deletion, namely of nonredundant key functions of PPARγ signaling in mammalian skin, are yet unknown because of embryonic lethality. Here, we describe the skin and hair phenotype of a whole-body PPARγ-null mouse (PpargΔ/Δ), obtained by preserving PPARγ expression in the placenta. PpargΔ/Δ mice exhibited total lipoatrophy and complete absence of sebaceous glands. Right after birth, hair follicle (HF) morphogenesis was transiently delayed, along with reduced expression of HF differentiation markers and of transcriptional regulators necessary for HF development. Later, adult PpargΔ/Δ mice developed scarring alopecia and severe perifollicular inflammation. Skin analyses in other models of lipodystrophy, AZIPtg/+ and Adipoq-Cretg/+Ppargfl/fl mice, coupled with skin graft experiments, showed that the early defects observed in hair morphogenesis were caused by the absence of adipose tissue. In contrast, the late alteration of HF cycle and appearance of inflammation were observed only in PpargΔ/Δ mice and likely were due to the lack sebaceous glands. Our findings underscore the increasing appreciation for the importance of adipose tissue-mediated signals in HF development and function.
Collapse
Affiliation(s)
- Chiara Sardella
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Carine Winkler
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Laure Quignodon
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jonathan A Hardman
- Centre for Dermatology Research, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Barbara Toffoli
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | - Jennifer E Hundt
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Liliane Michalik
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Charles R Vinson
- Center for Cancer Research, National Cancer Institute, Laboratory of Metabolism, Bethesda, Maryland, USA
| | - Ralf Paus
- Centre for Dermatology Research, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Béatrice Desvergne
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Federica Gilardi
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
23
|
Dunnill CJ, Al-Tameemi W, Collett A, Haslam IS, Georgopoulos NT. A Clinical and Biological Guide for Understanding Chemotherapy-Induced Alopecia and Its Prevention. Oncologist 2017; 23:84-96. [PMID: 28951499 DOI: 10.1634/theoncologist.2017-0263] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy-induced alopecia (CIA) is the most visibly distressing side effect of commonly administered chemotherapeutic agents. Because psychological health has huge relevance to lifestyle, diet, and self-esteem, it is important for clinicians to fully appreciate the psychological burden that CIA can place on patients. Here, for the first time to our knowledge, we provide a comprehensive review encompassing the molecular characteristics of the human hair follicle (HF), how different anticancer agents damage the HF to cause CIA, and subsequent HF pathophysiology, and we assess known and emerging prevention modalities that have aimed to reduce or prevent CIA. We argue that, at present, scalp cooling is the only safe and U.S. Food and Drug Administration-cleared modality available, and we highlight the extensive available clinical and experimental (biological) evidence for its efficacy. The likelihood of a patient that uses scalp cooling during chemotherapy maintaining enough hair to not require a wig is approximately 50%. This is despite different types of chemotherapy regimens, patient-specific differences, and possible lack of staff experience in effectively delivering scalp cooling. The increased use of scalp cooling and an understanding of how to deliver it most effectively to patients has enormous potential to ease the psychological burden of CIA, until other, more efficacious, equally safe treatments become available. IMPLICATIONS FOR PRACTICE Chemotherapy-induced alopecia (CIA) represents perhaps the most distressing side effect of chemotherapeutic agents and is of huge concern to the majority of patients. Scalp cooling is currently the only safe option to combat CIA. Clinical and biological evidence suggests improvements can be made, including efficacy in delivering adequately low temperature to the scalp and patient-specific cap design. The increased use of scalp cooling, an understanding of how to deliver it most effectively, and biological evidence-based approaches to improve its efficacy have enormous potential to ease the psychological burden of CIA, as this could lead to improvements in treatment and patient quality-of-life.
Collapse
Affiliation(s)
- Christopher John Dunnill
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, Huddersfield, United Kingdom
| | - Wafaa Al-Tameemi
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Andrew Collett
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, Huddersfield, United Kingdom
| | - Iain Stuart Haslam
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, Huddersfield, United Kingdom
| | - Nikolaos Theodoros Georgopoulos
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, Huddersfield, United Kingdom
| |
Collapse
|
24
|
Krieger K, Millar SE, Mikuda N, Krahn I, Kloepper JE, Bertolini M, Scheidereit C, Paus R, Schmidt-Ullrich R. NF-κB Participates in Mouse Hair Cycle Control and Plays Distinct Roles in the Various Pelage Hair Follicle Types. J Invest Dermatol 2017; 138:256-264. [PMID: 28942365 DOI: 10.1016/j.jid.2017.08.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 07/06/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022]
Abstract
The transcription factor NF-κB controls key features of hair follicle (HF) development, but the role of NF-κB in adult HF cycle regulation remains obscure. Using NF-κB reporter mouse models, strong NF-κB activity was detected in the secondary hair germ of late telogen and early anagen HFs, suggesting a potential role for NF-κB in HF stem/progenitor cell activation during anagen induction. At mid-anagen, NF-κB activity was observed in the inner root sheath and unilaterally clustered in the HF matrix, which indicates that NF-κB activity is also involved in hair fiber morphogenesis during HF cycling. A mouse model with inducible NF-κB suppression in the epithelium revealed pelage hair-type-dependent functions of NF-κB in cycling HFs. NF-κB participates in telogen-anagen transition in awl and zigzag HFs, and is required for zigzag hair bending and guard HF cycling. Interestingly, zigzag hair shaft bending depends on noncanonical NF-κB signaling, which previously has only been associated with lymphoid cell biology. Furthermore, loss of guard HF cycling suggests that in this particular hair type, NF-κB is indispensable for stem cell activation, maintenance, and/or growth.
Collapse
Affiliation(s)
- Karsten Krieger
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sarah E Millar
- Departments of Dermatology and Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nadine Mikuda
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Inge Krahn
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Marta Bertolini
- Department of Dermatology, University of Münster, Münster, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Ralf Paus
- Department of Dermatology, University of Münster, Münster, Germany; Centre for Dermatology Research, University of Manchester, Manchester, UK
| | - Ruth Schmidt-Ullrich
- Signal Transduction in Tumor Cells, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
25
|
Abstract
The skin is a complex landscape containing regions in which hair follicles exhibit
different types of behavior.
Collapse
Affiliation(s)
- Zhou Yu
- Peking University-Tsinghua University-National Institute of Biological Sciences, Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
26
|
Liao CP, Booker RC, Morrison SJ, Le LQ. Identification of hair shaft progenitors that create a niche for hair pigmentation. Genes Dev 2017; 31:744-756. [PMID: 28465357 PMCID: PMC5435888 DOI: 10.1101/gad.298703.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/13/2017] [Indexed: 11/25/2022]
Abstract
Liao et al. report the identification of hair shaft progenitors in the matrix that are differentiated from follicular epithelial cells expressing transcription factor KROX20. Expression of stem cell factor (SCF) by these cells is necessary for the maintenance of differentiated melanocytes and for hair pigmentation. Hair differentiates from follicle stem cells through progenitor cells in the matrix. In contrast to stem cells in the bulge, the identities of the progenitors and the mechanisms by which they regulate hair shaft components are poorly understood. Hair is also pigmented by melanocytes in the follicle. However, the niche that regulates follicular melanocytes is not well characterized. Here, we report the identification of hair shaft progenitors in the matrix that are differentiated from follicular epithelial cells expressing transcription factor KROX20. Depletion of Krox20 lineage cells results in arrest of hair growth, confirming the critical role of KROX20+ cells as antecedents of structural cells found in hair. Expression of stem cell factor (SCF) by these cells is necessary for the maintenance of differentiated melanocytes and for hair pigmentation. Our findings reveal the identities of hair matrix progenitors that regulate hair growth and pigmentation, partly by creating an SCF-dependent niche for follicular melanocytes.
Collapse
Affiliation(s)
| | | | - Sean J Morrison
- Department of Pediatrics.,Children's Research Institute.,Simmons Comprehensive Cancer Center.,Hamon Center for Regenerative Science and Medicine.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Lu Q Le
- Department of Dermatology.,Simmons Comprehensive Cancer Center.,Hamon Center for Regenerative Science and Medicine
| |
Collapse
|
27
|
Wang Q, Oh JW, Lee HL, Dhar A, Peng T, Ramos R, Guerrero-Juarez CF, Wang X, Zhao R, Cao X, Le J, Fuentes MA, Jocoy SC, Rossi AR, Vu B, Pham K, Wang X, Mali NM, Park JM, Choi JH, Lee H, Legrand JMD, Kandyba E, Kim JC, Kim M, Foley J, Yu Z, Kobielak K, Andersen B, Khosrotehrani K, Nie Q, Plikus MV. A multi-scale model for hair follicles reveals heterogeneous domains driving rapid spatiotemporal hair growth patterning. eLife 2017; 6:22772. [PMID: 28695824 PMCID: PMC5610035 DOI: 10.7554/elife.22772] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 06/29/2017] [Indexed: 01/27/2023] Open
Abstract
The control principles behind robust cyclic regeneration of hair follicles (HFs) remain unclear. Using multi-scale modeling, we show that coupling inhibitors and activators with physical growth of HFs is sufficient to drive periodicity and excitability of hair regeneration. Model simulations and experimental data reveal that mouse skin behaves as a heterogeneous regenerative field, composed of anatomical domains where HFs have distinct cycling dynamics. Interactions between fast-cycling chin and ventral HFs and slow-cycling dorsal HFs produce bilaterally symmetric patterns. Ear skin behaves as a hyper-refractory domain with HFs in extended rest phase. Such hyper-refractivity relates to high levels of BMP ligands and WNT antagonists, in part expressed by ear-specific cartilage and muscle. Hair growth stops at the boundaries with hyper-refractory ears and anatomically discontinuous eyelids, generating wave-breaking effects. We posit that similar mechanisms for coupled regeneration with dominant activator, hyper-refractory, and wave-breaker regions can operate in other actively renewing organs.
Collapse
Affiliation(s)
- Qixuan Wang
- Department of Mathematics, University of California, Irvine, United States,Center for Complex Biological Systems, University of California, Irvine, United States
| | - Ji Won Oh
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States,Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea,Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Korea,Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Anukriti Dhar
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Tao Peng
- Department of Mathematics, University of California, Irvine, United States
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Christian Fernando Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Ran Zhao
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States,Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaoling Cao
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States,Department of Burn Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jonathan Le
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Melisa A Fuentes
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Shelby C Jocoy
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Antoni R Rossi
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Brian Vu
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Kim Pham
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Xiaoyang Wang
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States
| | - Nanda Maya Mali
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea,Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Korea
| | - Jung Min Park
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea,Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Korea
| | - June-Hyug Choi
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea,Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Korea
| | - Hyunsu Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, Korea
| | - Julien M D Legrand
- UQ Diamantina Institute, Experimental Dermatology Group, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Eve Kandyba
- Department of Pathology, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, United States
| | - Jung Chul Kim
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea
| | - Moonkyu Kim
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea
| | - John Foley
- Department of Dermatology, Medical Sciences Program, Indiana University School of Medicine, Bloomington, United States
| | - Zhengquan Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health and State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Krzysztof Kobielak
- Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States,Centre of New Technologies, CeNT, University of Warsaw, Warsaw, Poland
| | - Bogi Andersen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States,Departments of Medicine and Biological Chemistry, University of California, Irvine, United States
| | - Kiarash Khosrotehrani
- UQ Diamantina Institute, Experimental Dermatology Group, Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, United States,Center for Complex Biological Systems, University of California, Irvine, United States,Department of Developmental and Cell Biology, University of California, Irvine, United States, (QN)
| | - Maksim V Plikus
- Center for Complex Biological Systems, University of California, Irvine, United States,Department of Developmental and Cell Biology, University of California, Irvine, United States,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, United States, (MVP)
| |
Collapse
|
28
|
Sánchez-Danés A, Blanpain C. Maintaining hair follicle stem cell identity in a dish. EMBO J 2016; 36:132-134. [PMID: 28003314 DOI: 10.15252/embj.201696051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
| | - Cédric Blanpain
- IRIBHM, Université Libre de Bruxelles, Brussels, Belgium.,WELBIO, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
29
|
Chacón-Martínez CA, Klose M, Niemann C, Glauche I, Wickström SA. Hair follicle stem cell cultures reveal self-organizing plasticity of stem cells and their progeny. EMBO J 2016; 36:151-164. [PMID: 27940653 DOI: 10.15252/embj.201694902] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/01/2016] [Accepted: 11/08/2016] [Indexed: 12/17/2022] Open
Abstract
Understanding how complex tissues are formed, maintained, and regenerated through local growth, differentiation, and remodeling requires knowledge on how single-cell behaviors are coordinated on the population level. The self-renewing hair follicle, maintained by a distinct stem cell population, represents an excellent paradigm to address this question. A major obstacle in mechanistic understanding of hair follicle stem cell (HFSC) regulation has been the lack of a culture system that recapitulates HFSC behavior while allowing their precise monitoring and manipulation. Here, we establish an in vitro culture system based on a 3D extracellular matrix environment and defined soluble factors, which for the first time allows expansion and long-term maintenance of murine multipotent HFSCs in the absence of heterologous cell types. Strikingly, this scheme promotes de novo generation of HFSCs from non-HFSCs and vice versa in a dynamic self-organizing process. This bidirectional interconversion of HFSCs and their progeny drives the system into a population equilibrium state. Our study uncovers regulatory dynamics by which phenotypic plasticity of cells drives population-level homeostasis within a niche, and provides a discovery tool for studies on adult stem cell fate.
Collapse
Affiliation(s)
| | - Markus Klose
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Catherin Niemann
- Institute for Biochemistry II, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sara A Wickström
- Paul Gerson Unna Group "Skin Homeostasis and Ageing", Max Planck Institute for Biology of Ageing, Cologne, Germany .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
30
|
Blanco S, Bandiera R, Popis M, Hussain S, Lombard P, Aleksic J, Sajini A, Tanna H, Cortés-Garrido R, Gkatza N, Dietmann S, Frye M. Stem cell function and stress response are controlled by protein synthesis. Nature 2016; 534:335-40. [PMID: 27306184 PMCID: PMC5040503 DOI: 10.1038/nature18282] [Citation(s) in RCA: 327] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/21/2016] [Indexed: 12/18/2022]
Abstract
Whether protein synthesis and cellular stress response pathways interact to control stem cell function is currently unknown. Here we show that mouse skin stem cells synthesize less protein than their immediate progenitors in vivo, even when forced to proliferate. Our analyses reveal that activation of stress response pathways drives both a global reduction of protein synthesis and altered translational programmes that together promote stem cell functions and tumorigenesis. Mechanistically, we show that inhibition of post-transcriptional cytosine-5 methylation locks tumour-initiating cells in this distinct translational inhibition programme. Paradoxically, this inhibition renders stem cells hypersensitive to cytotoxic stress, as tumour regeneration after treatment with 5-fluorouracil is blocked. Thus, stem cells must revoke translation inhibition pathways to regenerate a tissue or tumour.
Collapse
Affiliation(s)
- Sandra Blanco
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Roberto Bandiera
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Martyna Popis
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Shobbir Hussain
- Department of Biology & Biochemistry, University of Bath,
Claverton Down, Bath BA2 7AY, United Kingdom
| | - Patrick Lombard
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Jelena Aleksic
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Abdulrahim Sajini
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Hinal Tanna
- University of Cambridge, CR-UK, Cambridge Institute, Li Ka Shing
Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Rosana Cortés-Garrido
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Nikoletta Gkatza
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Sabine Dietmann
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| | - Michaela Frye
- Wellcome Trust – Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, United
Kingdom
| |
Collapse
|
31
|
FOXC1 maintains the hair follicle stem cell niche and governs stem cell quiescence to preserve long-term tissue-regenerating potential. Proc Natl Acad Sci U S A 2016; 113:E1506-15. [PMID: 26912458 DOI: 10.1073/pnas.1601569113] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Adult tissue stem cells (SCs) reside in niches, which orchestrate SC behavior. SCs are typically used sparingly and exist in quiescence unless activated for tissue growth. Whether parsimonious SC use is essential to conserve long-term tissue-regenerating potential during normal homeostasis remains poorly understood. Here, we examine this issue by conditionally ablating a key transcription factor Forkhead box C1 (FOXC1) expressed in hair follicle SCs (HFSCs). FOXC1-deficient HFSCs spend less time in quiescence, leading to markedly shortened resting periods between hair cycles. The enhanced hair cycling accelerates HFSC expenditure, and impacts hair regeneration in aging mice. Interestingly, although FOXC1-deficient HFs can still form a new bulge that houses HFSCs for the next hair cycle, the older bulge is left unanchored. As the new hair emerges, the entire old bulge, including its reserve HFSCs and SC-inhibitory inner cell layer, is lost. We trace this mechanism first, to a marked increase in cell cycle-associated transcripts upon Foxc1 ablation, and second, to a downstream reduction in E-cadherin-mediated inter-SC adhesion. Finally, we show that when the old bulge is lost with each hair cycle, overall levels of SC-inhibitory factors are reduced, further lowering the threshold for HFSC activity. Taken together, our findings suggest that HFSCs have restricted potential in vivo, which they conserve by coupling quiescence to adhesion-mediated niche maintenance, thereby achieving long-term tissue homeostasis.
Collapse
|
32
|
Axin2 marks quiescent hair follicle bulge stem cells that are maintained by autocrine Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2016; 113:E1498-505. [PMID: 26903625 DOI: 10.1073/pnas.1601599113] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
How stem cells maintain their identity and potency as tissues change during growth is not well understood. In mammalian hair, it is unclear how hair follicle stem cells can enter an extended period of quiescence during the resting phase but retain stem cell potential and be subsequently activated for growth. Here, we use lineage tracing and gene expression mapping to show that the Wnt target gene Axin2 is constantly expressed throughout the hair cycle quiescent phase in outer bulge stem cells that produce their own Wnt signals. Ablating Wnt signaling in the bulge cells causes them to lose their stem cell potency to contribute to hair growth and undergo premature differentiation instead. Bulge cells express secreted Wnt inhibitors, including Dickkopf (Dkk) and secreted frizzled-related protein 1 (Sfrp1). However, the Dickkopf 3 (Dkk3) protein becomes localized to the Wnt-inactive inner bulge that contains differentiated cells. We find that Axin2 expression remains confined to the outer bulge, whereas Dkk3 continues to be localized to the inner bulge during the hair cycle growth phase. Our data suggest that autocrine Wnt signaling in the outer bulge maintains stem cell potency throughout hair cycle quiescence and growth, whereas paracrine Wnt inhibition of inner bulge cells reinforces differentiation.
Collapse
|
33
|
Mastrogiannaki M, Lichtenberger BM, Reimer A, Collins CA, Driskell RR, Watt FM. β-Catenin Stabilization in Skin Fibroblasts Causes Fibrotic Lesions by Preventing Adipocyte Differentiation of the Reticular Dermis. J Invest Dermatol 2016; 136:1130-1142. [PMID: 26902921 PMCID: PMC4874948 DOI: 10.1016/j.jid.2016.01.036] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 12/31/2022]
Abstract
The Wnt/β-catenin pathway plays a central role in epidermal homeostasis and regeneration, but how it affects fibroblast fate decisions is unknown. We investigated the effect of targeted β-catenin stabilization in dermal fibroblasts. Comparative gene expression profiling of stem cell antigen 1- (Sca1-) and Sca1+ neonatal fibroblasts from upper and lower dermis, respectively, confirmed that Sca1+ cells had a preadipocyte signature and showed differential expression of Wnt/β-catenin–associated genes. By targeting all fibroblasts or selectively targeting Dlk1+ lower dermal fibroblasts, we found that β-catenin stabilization between developmental stages E16.5 and P2 resulted in a reduction in the dermal adipocyte layer with a corresponding increase in dermal fibrosis and an altered hair cycle. The fibrotic phenotype correlated with a reduction in the potential of Sca1+ fibroblasts to undergo adipogenic differentiation ex vivo. Our findings indicate that Wnt/β-catenin signaling controls adipogenic cell fate within the lower dermis, which potentially contributes to the pathogenesis of fibrotic skin diseases.
Collapse
Affiliation(s)
- Maria Mastrogiannaki
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Beate M Lichtenberger
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Andreas Reimer
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Charlotte A Collins
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Ryan R Driskell
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
34
|
Oh JW, Kloepper J, Langan EA, Kim Y, Yeo J, Kim MJ, Hsi TC, Rose C, Yoon GS, Lee SJ, Seykora J, Kim JC, Sung YK, Kim M, Paus R, Plikus MV. A Guide to Studying Human Hair Follicle Cycling In Vivo. J Invest Dermatol 2016; 136:34-44. [PMID: 26763421 PMCID: PMC4785090 DOI: 10.1038/jid.2015.354] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 12/17/2022]
Abstract
Hair follicles (HFs) undergo lifelong cyclical transformations, progressing through stages of rapid growth (anagen), regression (catagen), and relative "quiescence" (telogen). Given that HF cycling abnormalities underlie many human hair growth disorders, the accurate classification of individual cycle stages within skin biopsies is clinically important and essential for hair research. For preclinical human hair research purposes, human scalp skin can be xenografted onto immunocompromised mice to study human HF cycling and manipulate long-lasting anagen in vivo. Although available for mice, a comprehensive guide on how to recognize different human hair cycle stages in vivo is lacking. In this article, we present such a guide, which uses objective, well-defined, and reproducible criteria, and integrates simple morphological indicators with advanced, (immuno)-histochemical markers. This guide also characterizes human HF cycling in xenografts and highlights the utility of this model for in vivo hair research. Detailed schematic drawings and representative micrographs provide examples of how best to identify human HF stages, even in suboptimally sectioned tissue, and practical recommendations are given for designing human-on-mouse hair cycle experiments. Thus, this guide seeks to offer a benchmark for human hair cycle stage classification, for both hair research experts and newcomers to the field.
Collapse
Affiliation(s)
- Ji Won Oh
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea; Department of Immunology, Kyungpook National University School of Medicine, Daegu, Korea; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | | | - Ewan A Langan
- Department of Dermatology, University of Lübeck, Lübeck, Germany; Comprehensive Centre for Inflammation Research, University of Lübeck, Germany
| | - Yongsoo Kim
- Division of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joongyeub Yeo
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, California, USA
| | - Min Ji Kim
- Department of Dermatology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Christian Rose
- Dermatohistologisches Labor Rose/Bartsch, Lübeck, Germany
| | - Ghil Suk Yoon
- Department of Pathology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Seok-Jong Lee
- Department of Dermatology, Kyungpook National University School of Medicine, Daegu, Korea
| | - John Seykora
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jung Chul Kim
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea; Department of Immunology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Young Kwan Sung
- Department of Immunology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Moonkyu Kim
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Korea; Department of Immunology, Kyungpook National University School of Medicine, Daegu, Korea.
| | - Ralf Paus
- Dermatology Research Centre, Institute of Inflammation and Repair, University of Manchester, Manchester, UK; Department of Dermatology, University of Münster, Münster, Germany.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
35
|
Zhang H, Zhu NX, Huang K, Cai BZ, Zeng Y, Xu YM, Liu Y, Yuan YP, Lin CM. iTRAQ-Based Quantitative Proteomic Comparison of Early- and Late-Passage Human Dermal Papilla Cell Secretome in Relation to Inducing Hair Follicle Regeneration. PLoS One 2016; 11:e0167474. [PMID: 27907131 PMCID: PMC5132394 DOI: 10.1371/journal.pone.0167474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 11/15/2016] [Indexed: 02/05/2023] Open
Abstract
Alopecia is an exceedingly prevalent problem that lacks effective therapy. Recently, research has focused on early-passage dermal papilla cells (DPCs), which have hair inducing activity both in vivo and in vitro. Our previous study indicated that factors secreted from early-passage DPCs contribute to hair follicle (HF) regeneration. To identify which factors are responsible for HF regeneration and why late-passage DPCs lose this potential, we collected 48-h-culture medium (CM) from both of passage 3 and 9 DPCs, and subcutaneously injected the DPC-CM into NU/NU mice. Passage 3 DPC-CM induced HF regeneration, based on the emergence of a white hair coat, but passage 9 DPC-CM did not. In order to identify the key factors responsible for hair induction, CM from passage 3 and 9 DPCs was analyzed by iTRAQ-based quantitative proteomic technology. We identified 1360 proteins, of which 213 proteins were differentially expressed between CM from early-passage vs. late-passage DPCs, including SDF1, MMP3, biglycan and LTBP1. Further analysis indicated that the differentially-expressed proteins regulated the Wnt, TGF-β and BMP signaling pathways, which directly and indirectly participate in HF morphogenesis and regeneration. Subsequently, we selected 19 proteins for further verification by multiple reaction monitoring (MRM) between the two types of CM. These results indicate DPC-secreted proteins play important roles in HF regeneration, with SDF1, MMP3, biglycan, and LTBP1 being potential key inductive factors secreted by dermal papilla cells in the regeneration of hair follicles.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
| | - Ning-Xia Zhu
- Department of Cardiology, First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Keng Huang
- Emergency Department, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Bo-Zhi Cai
- Tissue Engineering Laboratory, First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Yang Zeng
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Yang Liu
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yan-Ping Yuan
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
| | - Chang-Min Lin
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
- * E-mail:
| |
Collapse
|
36
|
Geyfman M, Plikus MV, Treffeisen E, Andersen B, Paus R. Resting no more: re-defining telogen, the maintenance stage of the hair growth cycle. Biol Rev Camb Philos Soc 2015; 90:1179-96. [PMID: 25410793 PMCID: PMC4437968 DOI: 10.1111/brv.12151] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 09/12/2014] [Accepted: 10/07/2014] [Indexed: 12/17/2022]
Abstract
The hair follicle (HF) represents a prototypic ectodermal-mesodermal interaction system in which central questions of modern biology can be studied. A unique feature of these stem-cell-rich mini-organs is that they undergo life-long, cyclic transformations between stages of active regeneration (anagen), apoptotic involution (catagen), and relative proliferative quiescence (telogen). Due to the low proliferation rate and small size of the HF during telogen, this stage was conventionally thought of as a stage of dormancy. However, multiple lines of newly emerging evidence show that HFs during telogen are anything but dormant. Here, we emphasize that telogen is a highly energy-efficient default state of the mammalian coat, whose function centres around maintenance of the hair fibre and prompt responses to its loss. While actively retaining hair fibres with minimal energy expenditure, telogen HFs can launch a new regeneration cycle in response to a variety of stimuli originating in their autonomous micro-environment (including its stem cell niche) as well as in their external tissue macro-environment. Regenerative responses of telogen HFs change as a function of time and can be divided into two sub-stages: early 'refractory' and late 'competent' telogen. These changing activities are reflected in hundreds of dynamically regulated genes in telogen skin, possibly aimed at establishing a fast response-signalling environment to trauma and other disturbances of skin homeostasis. Furthermore, telogen is an interpreter of circadian output in the timing of anagen initiation and the key stage during which the subsequent organ regeneration (anagen) is actively prepared by suppressing molecular brakes on hair growth while activating pro-regenerative signals. Thus, telogen may serve as an excellent model system for dissecting signalling and cellular interactions that precede the active 'regenerative mode' of tissue remodeling. This revised understanding of telogen biology also points to intriguing new therapeutic avenues in the management of common human hair growth disorders.
Collapse
Affiliation(s)
- Mikhail Geyfman
- Department of Ophthalmology, University of California, Irvine, CA 92697, USA
| | - Maksim V. Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| | - Elsa Treffeisen
- Department of Dermatology, Kligman Labouratories, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Bogi Andersen
- Department of Biological Chemistry, University of California Irvine, CA 92697, USA
- Department of Medicine, University of California Irvine, CA 92697, USA
- Institute for Genomics and Bioinformatics, University of California, Irvine, CA 92697, USA
| | - Ralf Paus
- Department of Dermatology, University of Luebeck, Luebeck, Germany
- Institute of Inflammation and Repair, and Dermatology Centre, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PL, UK
| |
Collapse
|
37
|
Expression profiling reveals genes involved in the regulation of wool follicle bulb regression and regeneration in sheep. Int J Mol Sci 2015; 16:9152-66. [PMID: 25915029 PMCID: PMC4463583 DOI: 10.3390/ijms16059152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/08/2015] [Accepted: 04/10/2015] [Indexed: 01/04/2023] Open
Abstract
Wool is an important material in textile manufacturing. In order to investigate the intrinsic factors that regulate wool follicle cycling and wool fiber properties, Illumina sequencing was performed on wool follicle bulb samples from the middle anagen, catagen and late telogen/early anagen phases. In total, 13,898 genes were identified. KRTs and KRTAPs are the most highly expressed gene families in wool follicle bulb. In addition, 438 and 203 genes were identified to be differentially expressed in wool follicle bulb samples from the middle anagen phase compared to the catagen phase and the samples from the catagen phase compared to the late telogen/early anagen phase, respectively. Finally, our data revealed that two groups of genes presenting distinct expression patterns during the phase transformation may have important roles for wool follicle bulb regression and regeneration. In conclusion, our results demonstrated the gene expression patterns in the wool follicle bulb and add new data towards an understanding of the mechanisms involved in wool fiber growth in sheep.
Collapse
|
38
|
Driskell I, Oeztuerk-Winder F, Humphreys P, Frye M. Genetically induced cell death in bulge stem cells reveals their redundancy for hair and epidermal regeneration. Stem Cells 2015; 33:988-98. [PMID: 25447755 PMCID: PMC4583782 DOI: 10.1002/stem.1910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/22/2014] [Accepted: 11/12/2014] [Indexed: 12/17/2022]
Abstract
Adult mammalian epidermis contains multiple stem cell populations in which quiescent and more proliferative stem and progenitor populations coexist. However, the precise interrelation of these populations in homeostasis remains unclear. Here, we blocked the contribution of quiescent keratin 19 (K19)-expressing bulge stem cells to hair follicle formation through genetic ablation of the essential histone methyltransferase Setd8 that is required for the maintenance of adult skin. Deletion of Setd8 eliminated the contribution of bulge cells to hair follicle regeneration through inhibition of cell division and induction of cell death, but the growth and morphology of hair follicles were unaffected. Furthermore, ablation of Setd8 in the hair follicle bulge blocked the contribution of K19-postive stem cells to wounded epidermis, but the wound healing process was unaltered. Our data indicate that quiescent bulge stem cells are dispensable for hair follicle regeneration and epidermal injury in the short term and support the hypothesis that quiescent and cycling stem cell populations are equipotent.
Collapse
Affiliation(s)
- Iwona Driskell
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
39
|
Hsu YC, Li L, Fuchs E. Transit-amplifying cells orchestrate stem cell activity and tissue regeneration. Cell 2014; 157:935-49. [PMID: 24813615 DOI: 10.1016/j.cell.2014.02.057] [Citation(s) in RCA: 278] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/16/2014] [Accepted: 02/24/2014] [Indexed: 12/17/2022]
Abstract
Transit-amplifying cells (TACs) are an early intermediate in tissue regeneration. Here, using hair follicles (HFs) as a paradigm, we show that emerging TACs constitute a signaling center that orchestrates tissue growth. Whereas primed stem cells (SCs) generate TACs, quiescent SCs only proliferate after TACs form and begin expressing Sonic Hedgehog (SHH). TAC generation is independent of autocrine SHH, but the TAC pool wanes if they can't produce SHH. We trace this paradox to two direct actions of SHH: promoting quiescent-SC proliferation and regulating dermal factors that stoke TAC expansion. Ingrained within quiescent SCs' special sensitivity to SHH signaling is their high expression of GAS1. Without sufficient input from quiescent SCs, replenishment of primed SCs for the next hair cycle is compromised, delaying regeneration and eventually leading to regeneration failure. Our findings unveil TACs as transient but indispensable integrators of SC niche components and reveal an intriguing interdependency of primed and quiescent SC populations on tissue regeneration.
Collapse
Affiliation(s)
- Ya-Chieh Hsu
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Lishi Li
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
40
|
Guerrero-Juarez CF, Ramos R, Oh JW, Hsi TC, Plikus MV. Light-emitting hair follicles: studying skin regeneration with in vivo imaging. J Invest Dermatol 2014; 134:1496-1498. [PMID: 24825056 DOI: 10.1038/jid.2014.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cutting-edge imaging technologies and new luminescent and fluorescent genetic tools now make it possible to study hair regeneration in vivo in real time at the microscopic single-cell level and at the macroscopic level of hair follicle populations. These technologies also allow for noninvasive assessment of the skin's clinically relevant homeostatic parameters, such as oxidative stress levels and pH.
Collapse
Affiliation(s)
- Christian Fernando Guerrero-Juarez
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California, USA.
| |
Collapse
|